Abstract
Developmental pharmacology describes the impact of maturation on drug disposition (pharmacokinetics, PK) and drug effects (pharmacodynamics, PD) throughout the paediatric age range. This paper, written by a multidisciplinary group of experts, summarizes current knowledge, and provides suggestions to pharmaceutical companies, regulatory agencies and academicians on how to incorporate the latest knowledge regarding developmental pharmacology and innovative techniques into neonatal and paediatric drug development.
Biological aspects of drug absorption, distribution, metabolism and excretion throughout development are summarized. Although this area made enormous progress during the last two decades, remaining knowledge gaps were identified. Minimal risk and burden designs allow for optimally informative but minimally invasive PK sampling, while concomitant profiling of drug metabolites may provide additional insight in the unique PK behaviour in children. Furthermore, developmental PD needs to be considered during drug development, which is illustrated by disease‐ and/or target organ‐specific examples. Identifying and testing PD targets and effects in special populations, and application of age‐ and/or population‐specific assessment tools are discussed. Drug development plans also need to incorporate innovative techniques such as preclinical models to study therapeutic strategies, and shift from sequential enrolment of subgroups, to more rational designs.
To stimulate appropriate research plans, illustrations of specific PK/PD‐related as well as drug safety‐related challenges during drug development are provided. The suggestions made in this joint paper of the Innovative Medicines Initiative conect4children Expert group on Developmental Pharmacology and the European Society for Developmental, Perinatal and Paediatric Pharmacology, should facilitate all those involved in drug development.
Keywords: developmental pharmacology, drug development, paediatrics

1. DEVELOPMENTAL PHARMACOLOGY
Pharmacotherapy is a powerful tool in preventive and curative medicine, with shifts in practices based on perceived needs. In children, an increase in outpatient prescription drug use for asthma, attention‐deficit/hyperactivity disorder (ADHD) and contraception is reported, but a decrease in antihistamines, antibiotics, and upper respiratory combinations drugs. 1 , 2 Based on a survey in 2010, amoxicillin was the most frequently prescribed drug in children up to 11 years, and methylphenidate in adolescents. 2 Besides drug utilization, drug disposition and effects may differ between populations. General clinical pharmacology principles apply to all populations and refer to the processes involved in drug disposition (pharmacokinetics, PK) and drug effects (pharmacodynamics, PD). PK is determined by drug absorption and distribution, followed by metabolism of most compounds and excretion of the compound and/or its metabolites. PD in part relates to characteristics of drug receptors or targets as well as postreceptor effects. For the paediatric population (i.e. from early neonatal life through adolescence), developmental changes in biological processes involved in PK and PD need to be considered (developmental pharmacology). 3 , 4 Due to rapid physiological changes (e.g. maturation of liver and kidney function, organ‐specific growth), developmental impact on PK and PD is most remarkable in the neonatal period and first year of life. However, PK and/or PD may also differ from adults during childhood and adolescence. 3 , 4 As a consequence, safe and effective therapy for an individual child cannot be ensured by linear extrapolation of adult dosing recommendations. Knowledge of ontogeny of biological processes involved improves PK and PD predictions, and supports individualized and targeted drug therapy. 5 Multidisciplinary collaboration between basic scientists, paediatric subspecialists, pharmacists, (clinical) pharmacologists, pharmacometricians, biomedical engineers, veterinarians and other experts expands our understanding rapidly. Besides ontogeny, nonmaturational factors such as environment, disease states, or co‐treatment can also influence PK/PD. 6 The goals of this paper are: (i) to summarize current knowledge; (ii) to provide illustrations for pharmaceutical companies, regulatory agencies and academicians about how to incorporate the latest knowledge regarding developmental pharmacology; and (iii) to underline why a comprehensive approach for such incorporation should be considered. In addition, some innovative techniques for paediatric drug development are summarized. The scope of this paper includes pharmacology (PK and PD), while other aspects of drug development are not covered. This paper reflects a collaboration between researchers from the Innovative Medicines Initiative conect4children (IMI c4c) Expert group on Developmental Pharmacology, and the European Society for Developmental, Perinatal and Paediatric Pharmacology (ESDPPP). 7 , 8
2. DEVELOPMENTAL PK
In the outpatient setting, drugs are commonly administered orally. In contrast, in an intensive care setting the parenteral (mainly intravenous) route is used in 71.3, 60.7 and 68.7% of drug administrations in adult, paediatric and neonatal intensive care unit patients, respectively. 9 For most drugs, children are sufficiently different from adults to require dosing adaptations. Understanding the fate of a compound in the body is essential to select the optimal dosing strategy for each individual. Size and age‐related changes in drug absorption, distribution, metabolism and excretion pathways have been extensively studied. 3 , 10 , 11 , 12 Although knowledge of developmental PK has increased, knowledge gaps still remain. Within the developmental PK section, we will provide current evidence on maturational aspects of absorption, distribution, metabolism and excretion (ADME) processes. Neonates with intrauterine growth restriction (IUGR) and adolescents were selected as special paediatric populations to further illustrate these concepts. Furthermore, we will discuss how including metabolite concentrations in bioanalytical approaches can provide useful data for paediatric predictive PK models, and how improvements can be made towards less invasive sampling methods to gather data on drug disposition in children.
2.1. Developmental biological processes involved in drug disposition
2.1.1. Absorption
Absorption determines how concentration changes after extravascular administration, including absorption rate (how rapid?) and bioavailability (how much?), both impacted by growth and maturation. Enteral (buccal, rectal, oral) or nonenteral (nasal, ophthalmic, dermal, inhalational, intramuscular, intrathecal) routes are relevant.
For enteral administration, maturation includes gastric and intestinal pH and motility, pancreatic lipase activity, bile acid secretion, and presystemic (first pass) drug metabolism and transport in the intestinal wall. The gastric fluid composition (bile acids, osmolarity, pH) displays age‐dependent changes, further affected by feeding frequency and type. 13 The capacity of the newborn to produce gastric acid appears similar to older populations, despite the higher pH in their gastric fluid. 13 , 14 This discrepancy is probably explained by the impact of frequency and volume of milk feeds, alkalinizing the gastric content. Consequently, plasma concentrations of the acid‐labile antibiotic penicillin reached 5–6 times higher values in neonates compared to older children. 15 Gastric emptying is another example of the merged effect of maturation and feeding practices. In a meta‐analysis of data from preterm neonates to adults, not age but meal type (aqueous>breast milk>formula milk>semi‐solid, solid) was the main determinant of gastric emptying, with collinearity between age and meal type. 16 Pancreatic lipase activity is typically low at birth and evolves during infancy, somewhat compensated by lipase activity (bile salt‐stimulated lipase) in fresh human milk. 17 The bile acid pool size, bile flow and its ileal reabsorption also display maturation, resulting in lower duodenal bile acid concentrations in neonates. 10 The activity of intestinal drug‐metabolizing enzymes (DMEs) and drug transporters (DTs) co‐determine age‐dependent drug bioavailability. 18 , 19 Intestinal drug metabolism matures slowly, as exemplified by midazolam's decreasing bioavailability over the paediatric age range (reflecting increasing first pass metabolism in the intestine and liver), although nonmaturational factors also determine a significant part of the variability. 20 For example, Blake et al. illustrated that dextromethorphan and caffeine metabolism in infants were affected by the type of nutrition (human milk vs. formula). 21
For nonenteral administration routes that display an absorption phase, absorption kinetics relates to the drug diffusion rate as well as tissue perfusion rate (i.e. regional blood flow), with possible bypass of the first‐pass effect. Absorption through the skin can be more rapid and/or more extensive in children (e.g. topical timolol for infantile haemangioma in preterm neonates, or minoxidil for paediatric hair disorders) 22 , 23 prompting attention to possible unintended (systemic) side‐effects after local application.
2.1.2. Distribution
Following absorption, most drugs will gain access from the systemic circulation to other body compartments. Distribution profiles depend on drug (e.g. size, lipophilicity), patient (e.g. blood flow, biological barrier characteristics including DT expression, membrane composition), or both (e.g. plasma/tissue protein binding). The apparent volume of distribution (Vd, L or L/kg) reflects the degree to which a drug is distributed in body tissue relative to the plasma. Weight is the obvious main driver of the absolute volume of distribution (L), while age‐related changes in drug protein binding and body composition in part explain developmental differences in distribution, when expressed as relative volume (L/kg). The major drug binding protein albumin (primarily binds neutral to acidic drugs) increases in concentration during the first 1–3 years. 24 Data on α‐1‐acid glycoprotein (binds basic or cationic drugs) are sparse, but paediatric reference ranges indicate an increase up to 5 years of life. 25 Besides proteins abundance, binding of competitors at protein binding sites can alter binding affinity and influence unbound, pharmacologically active, drug fractions. The unbound fraction of albumin‐bound drugs, cefazolin, flucloxacillin, vancomycin, cefoperazone and phenobarbital, is higher in neonates compared to older populations. 26 , 27 , 28 , 29 , 30 Concerning body composition, the total body water fraction (>70%) is higher in neonates compared to children and adolescents (around 60%). 31 , 32 With maturation the fraction of total body water declines, while the fraction of fat mass increases. One study in nonobese children (age 7–14 years) demonstrated a mean percentage body fat of 20% in boys and 25% in girls, 33 but the obesity epidemic in children, adolescents and adults has resulted in large variations in fat mass. 34 Variation in body composition may require a different drug dosing. 35
Multiple brain‐specific factors impact drug distribution to the brain, and their influence differs throughout development. Blood–brain (BBB) and choroidal blood–cerebrospinal fluid (BCSFB) barrier function is still immature in children. Efflux transporters acting on amphiphilic drugs are likely to be more efficient around birth at the BCSFB, while at the BBB, some transporter expression is downregulated compared to adult levels. 36 Verscheijden et al. recently described location and transporter‐specific maturation of ATP‐binding cassette transporters in both human BBB and BCSFB. 37 Besides, cerebral blood flow, drug binding to and uptake by neural cells, drug metabolism, volume of cerebral fluid compartments and rate of cerebrospinal fluid secretion contribute to cerebral drug bioavailability. 36
In addition to development, nonmaturational factors (e.g. diseases, co‐treatment) affects drug distribution. It has been documented that sepsis alters drug distribution, resulting in lower amoxicillin concentrations and a longer half‐life in neonates as compared to nonsystemic disease conditions. 38 Also, body composition may be altered in conditions such as severe illness or eating disorders. 32 , 39 Extracorporeal membrane oxygenation (ECMO) in neonates leads to a larger Vd for both lipophilic and hydrophilic drugs. 40 During paediatric ECMO the Vd (L/kg) of e.g. analgo‐sedatives also increases, showing the importance of including infants and children treated with ECMO in PK studies for drugs relevant to this population. 41
2.1.3. Metabolism
Drug metabolism takes place in the liver and a variety of other organs, and is highly variable. The impact of maturation is highest in the first years of life. Drugs can be metabolized by Phase I and II DMEs, which show enzyme‐ and organ‐specific maturational patterns. Recent proteomics analyses have quickly increased understanding of these patterns. 42 , 43 Three patterns can be identified with: (i) most DMEs being low at birth and increasing to adult levels during the first months of life; (ii) high at birth and decreasing thereafter and; iii) stable expression. Most DMEs (e.g. CYP1A2, 2A6, 2D6; 2E1, 3A4) follow the first pattern, with birth and external factors contributing to rapid maturation. Based on in vitro data, adult values of CYP1A2 activity and protein expression are reached at age 5–15 and 1–5 years of age, respectively. For CYP2E1 activity is low in foetuses and reaches 50% of adult values in infants. Its foetal protein expression is variable and adult values are reached at 1–5 years of age. 44 In vivo data display additional variability. 45 For the CYP3A family, protein expression is 65–80% in foetuses but remains relatively constant at later ages. Interestingly, CYP3A4 enzyme activity hereby increases after birth (reaching 30–40% of adult values at 1 month, 50% at 6–12 months and 100% at age 1–5 y), while CYP3A7 is highly active in foetuses and early neonatal life, with a subsequent decrease in activity during the first weeks of life. 44 , 46 , 47 This shift between CYP3A7 and 3A4 occurs immediately after birth. 46 A detailed overview on multilevel ontogeny patterns of hepatic DT and DMEs is recently compiled by van Groen et al. 44 When doses are linearly extrapolated, immature drug metabolism can lead to toxicity. Exceptions may occur such as for paracetamol, where immature uridine 5′‐diphospho‐glucuronosyltransferase (UGT) activity is compensated by high sulfation activity directly after birth. 48 For many metabolized drugs, clearance is increased in toddlers and young infants resulting in subtherapeutic exposure, which is often not due to increased DME activity per se, but overall increased metabolic capacity as a result of a relatively large liver size at this age. 49 The interplay of maturation with other factors further adds variability to paediatric drug metabolism. Inflammation/sepsis leads to decreased CYP3A4 mediated midazolam clearance, 50 while genetic variation further affects omeprazole and tacrolimus disposition. 51 , 52 Moreover, treatment modalities may lead to variable disposition, e.g. during therapeutic hypothermia. 53
2.1.4. Excretion
Excretion occasionally occurs through the hepato‐biliary or pulmonary route, but most commonly by the kidneys (glomerular filtration rate [GFR], tubular secretion and tubular reabsorption). Ontogeny of biliary excretion appears to be fast, attaining adult activity within the first weeks to months. 54 Elimination kinetics in the lung is determined by alveolar ventilation, functional residual capacity and cardiac output. 55
Based on aminoglycoside and vancomycin datasets, GFR maturation has been described, using a bodyweight‐dependent exponent and postnatal age (PNA). 56 By pooling of GFR estimates (polyfructose, chrome‐ethylenediaminetetraacetic acid, mannitol or iohexol) GFR has been shown to reach half the mature value based on allometric body size by 48 weeks postmenstrual age (PMA), with full maturity being attained before 2 years of age. 57 In a recent analysis on differences in GFR between preterm and term neonates, Salem et al. documented that both postnatal and gestational age (GA) are relevant in GFR development until 1.25 years, as PMA ignores birth as a pivotal event. 58 Compared to glomerular filtration, tubular functions appear to mature more slowly. 59 , 60 Our knowledge is limited, but recent kidney drug transporter expression data show different rates and patterns of maturation, which appears aligned with PK data of corresponding substrates. 61 , 62
In addition to maturation, GFR variability is further affected by nonmaturational covariates. To illustrate both ends of the spectrum, critically ill children may display enhanced kidney perfusion and glomerular hyperfiltration, resulting in augmented renal clearance. 63 In contrast, chronic kidney disease (CKD) and acute kidney injury are associated with reduced clearance. The impact of CKD is not limited to decreased excretion, as it may lead to modifications in absorption, distribution, transport, and/or metabolism. 64
2.1.5. Special populations
As mentioned earlier, two specific paediatric populations will be discussed, namely neonates with IUGR and adolescents, to further stress the relevance and diversity in developmental PK. IUGR is the result of foetal growth failure caused by various factors, displaying foetal and postnatal features of malnutrition, while small for GA (SGA) is based on any birth weight <10th percentile for a given GA and may reflect genetic influences on size. 65 , 66 Although definitions differ, both terms are often used interchangeably, as illustrated below. Sparse data are available on the impact of IUGR/SGA on PK. Hepatic DME and DT expression are affected by IUGR. 67 , 68 To illustrate its relevance, S‐ibuprofen clearance was 3.11‐fold higher in SGA compared to appropriate for GA preterms. 69 IUGR also results in decreased renal clearance due to impaired renal developmental programming, decreased nephron number and GFR, leading to CKD. 70 Examples are reduced vancomycin, gentamicin or amikacin clearance in SGA compared to appropriate for GA neonates. 71 , 72 , 73 Finally, the impact of IUGR on PK probably remains relevant throughout paediatric life as e.g. GFR remains lower. 74
At the other end of the paediatric spectrum, adolescents are commonly regarded as similar to adults with respect to PK. However, some organ systems undergo intense development during adolescence (e.g. skeleton, reproductive tissue, central nervous system, etc.), with PK implications. Activity of several DME change throughout physical and sexual maturation, with largest variability in infants and adolescents. As an example, alterations in sex and growth hormones appear to reduce DME (CYP1A2) activity, except progesterone which appears to increase CYP3A activity. 75 , 76 To illustrate altered PK, for the oral combined (nomegestrol acetate + oestradiol) contraceptive pill Zoely (Theramex Ireland, Dublin, Ltd), lower exposure of oestradiol (−36%) was observed in adolescents compared to adults. 77 Another study evaluating the effect of lixisenatide observed shorter time to maximal serum concentration (Cmax), and lower absolute Cmax in adolescents compared to adults. 78 Finally, a recent systematic review showed differences regarding dose‐related concentrations in children and adolescents compared to adults for 14 of 26 neuroactive/psychoactive drugs. 79 These examples of age‐dependent PK differences underscore (i) the need to involve paediatric pharmacologists to evaluate when detailed studies on drug efficacy and safety are needed in paediatric subgroups, and (ii) that developmental age rather than administrative (chronological) age needs to be considered when assessing PK(/PD) in adolescents. 76 Approved dosing for adults and adolescents seems highly equivalent, allowing allometric scaling with subsequent limited PK studies on a case‐by‐case approach. 80 , 81 Nevertheless, the unique developmental setting of adolescence often requests population‐specific PK(/PD) attention. 76
2.2. Utility of measuring drug metabolite levels to support PK studies in children
About a decade ago, Anderson and Holford already pointed towards knowledge and expertise gaps in paediatric pharmacotherapy: the poorly studied impact of metabolites on effect was 1 of these factors. 82 Indeed, while metabolism generally leads to loss of activity, several drugs that are also used in neonates and children are known to generate metabolites that are pharmacologically active. The fact that this level of clinically relevant detail on metabolite disposition remains unknown for many drugs used in paediatric populations underscores the need for increased efforts to measure drug metabolites. The advances in the performance of bioanalytical methods in the past decade can facilitate this ambition. Indeed, recently developed methods have pushed the limits to new heights, not only in terms of analytical sensitivity (lower quantification limits) and sample volume requirements (often below 10 μL or even less), but also in terms of the ability for concomitant measurement of parent and multiple metabolites in a single injection for liquid chromatography with tandem mass spectrometry (LC–MS/MS) analysis. 83 , 84 Also, the use of a [14C]‐microtracer with accelerated mass spectrometry for metabolite profiling has been reported. 85 In the current section, we aim to illustrate the possible utility of measuring drug metabolite concentrations to support PK studies in neonates and children. While metabolite levels will not always provide additional insights regarding the disposition of a given drug, in several cases metabolite profiles will support: (i) enhancing the knowledge of in vivo impact of ontogeny of elimination pathways; and (ii) improved understanding of the PKPD profiles of drugs generating pharmacologically/toxicologically active metabolites. As described in detail below, morphine and paracetamol generate a pharmacologically and a toxicologically relevant metabolite, respectively. In addition, metabolites may contribute to unravel developmental toxicity mechanisms like for ifosfamide 86 or valproic acid. 87
2.2.1. Morphine and its metabolites
Morphine undergoes biotransformation to morphine‐3‐glucuronide and morphine‐6‐glucuronide (M6G). Only M6G has a μ‐opioid agonism. The PK‐related effect of morphine in neonates and children thus depends not only on ontogeny of the morphine elimination process, but also on specific ontogeny profiles of M6G formation and elimination, justifying quantification of M6G next to morphine also in paediatric morphine PK studies. A meta‐analysis reported moderate (2‐fold) to high (10‐fold) interindividual variability in morphine clearance (and in relative M6G levels) depending on the paediatric age group, but especially in critically ill patients. 88 Moreover, the highest interpatient variability in dose–exposure relationship was observed in neonates and infants. Specifically in neonates morphine elimination is driven by hepatic maturation, while renal maturation determines its glucuronides excretion. As illustrated in a meta‐analysis by Knosgaard et al., 84 PMA and PNA provide the best elimination predictions of morphine and its glucuronides, respectively. By additionally taking into account size‐related parameters, the authors constructed a model to describe morphine and metabolites PK in (pre)term neonates.
2.2.2. Paracetamol and its metabolites
Several independent studies have addressed the maturation of elimination of paracetamol and its phase‐2 metabolites (paracetamol glucuronide, paracetamol sulfate). 89 , 90 Whereas glucuronidation matures during the first 2 years of life, sulfation shows a different developmental pattern with higher expression of sulfotransferase (SULT)1A1 and SULT2A1 in infants and young children compared to adults. 42 In addition, a population PK study demonstrated that the oxidative paracetamol biotransformation pathway (eventually yielding a.o. mercapturic acid metabolites) was subject to only small maturational changes. 91 Consistently, in very preterm neonates, paracetamol glucuronidation was very low, whereas other pathways (sulfation, glutathione conjugation) were of relatively higher importance compared to adults. 92 Interestingly, the clinical relevance of UGT1A9 polymorphism, the glucuronosyltransferase isoform involved in paracetamol glucuronidation, resulted in up to 42% decreased glucuronide formation clearance in some neonates. This finding illustrates that the impact of polymorphisms on top of maturational covariates should not be ignored a priori in neonatal pharmacology. 93 In addition, measuring paracetamol glucuronide concentrations thus contributed to a more accurate identification of the factors (polymorphism and CL maturation) determining variability in neonatal paracetamol exposure. A similar concept was recently presented for omeprazole. Based on a population PK model in infants for omeprazole and 2 metabolites, the complex interplay of both ontogeny and polymorphism of CYP2C19 was illustrated. 52 For older paediatric age groups, knowledge gaps exist regarding paracetamol PK, as illustrated in a review on paracetamol use in overweight children and adolescents. 94 It remains uncertain whether the documented increased clearance in obese adults also applies to obese adolescents. This urges to determine PK of paracetamol and its (hepatoxicity‐related) metabolites in this population, which can then support acceptable dosing recommendations in children and adolescents.
2.3. Minimal risk and burden designs for PK in vulnerable populations
The study of PK in infants and children has historically been challenging due to sample size, heterogeneity of study population and patient burden (blood sampling). 95 Optimal PK sampling strategy must consider both timing and number of samples per patient, and also minimizing sample volume using innovative bioanalytical methods. Trial designs need to minimize risk and burden for participants and their families. This covers not only sampling, but broader trial‐related and ethical considerations. 96 The child's interest should always prevail over that of science and society. 96
2.3.1. Timing and number of samples per patient
Each sample is precious and should only be collected if it is predicted to provide optimal and needed information. From a PK modelling perspective, optimal sampling design allows parameter estimation with maximal precision. Modelling and simulation are strongly recommended in paediatric investigation plans 97 and methods for optimally designing sampling schedules and to derive the required number of study participants 98 are now available. From previously developed PK models in adults or older children, it is possible to derive optimal sampling times with the highest chance of estimating PK parameters most precisely in the population of interest. Opportunistic PK sampling design could mitigate the challenges associated with PK studies in children. However, it should consider the density and quality of sampling, as well as the stability of the drug. 99 Inappropriate design will result in biased 100 or imprecise 101 PK model estimates. Judicious use of optimal design 102 or simulation estimation 103 can, however, mean that with a few samples precise estimates can be derived making studies minimally invasive and maximally informative. Mathematically optimal sampling design might not be clinically optimal in terms of number and timing of samples. It is therefore important to take the practicalities of sampling and ethical aspects into consideration when designing PK studies. 96 Furthermore, reliability of extrapolation methods may differ between paediatric subpopulations and should be considered to limit the number of samples as much as possible. 104
2.3.2. Low volume sampling
Microsampling, which is the collection of smaller‐than‐normal plasma samples for bio‐analysis, may provide a solution and includes mainly dried blood spots (DBS), dry plasma spots, volumetric absorptive microsampling or capillary microsampling techniques. In DBS or dry plasma spots, a small volume of blood or plasma is applied on an absorbent paper which is dried after saturation has occurred. In the laboratory, the blood or plasma is eluted out of the paper and analysed. The advantage of DBS is that analytes have higher stability in ambient conditions for several days or months. 105 Application of DBS and rapid LC–MS/MS‐based methods have allowed a deepening knowledge of propranolol PK in (pre)term neonates. 106 Cohen‐Wolkowiez et al. reported on the feasibility of using DBS concentrations in combination with plasma samples for PK model building of piperacillin and tazobactam in infants. 101 A quite similar, optimized technique is volumetric absorptive microsampling, in which a fixed blood volume is absorbed by the porous, hydrophilic tip of the device. After the tip is dried, it is sent to the lab for drug extraction and bio‐analysis. In capillary microsampling, blood is collected in a capillary tube and subsequently centrifuged in this tube before bio‐analysis. Overall, microsampling seems promising as it would allow a significant reduction in the blood volume required.
Besides minimally invasive blood sampling, approaches for sampling in other matrices are developed/under development. Microneedle technology is known as a (transdermal) drug delivery system. 107 Nevertheless, as it creates (a) transient channel(s) across the skin, it can also be used for sampling. Microneedle‐based interstitial fluid (ISF) sampling can complement conventional blood or urine sampling. In adults, microneedle‐based ISF from human skin was collected to quantify selected biomarkers, and was well tolerated. 108 To illustrate its applicability, caffeine PK in adults and glucose response after a meal and insulin administration in diabetic children were reported to be similar in plasma and ISF. 108 Although this technique is mainly applied in research settings, it is promising as microneedle insertion was less painful compared to subcutaneous catheters in diabetic children and adolescents. 109
A noninvasive technique for measuring drug exposure is using saliva as matrix. The advantages of saliva monitoring in paediatric PK trials are acknowledged by the Food and Drug Administration (FDA) as it reduces blood sampling, is easy to collect and causes minimal discomfort. 110 The usefulness of saliva for therapeutic drug monitoring has been studied for e.g. anti‐epileptics, antiretrovirals, antipsychotics, antibiotics and antifungals, but is dependent on the physicochemical properties of the drug. 111 , 112 , 113 , 114
2.3.3. Microdosing
Microdosing is another promising method to minimize patient burden while studying PK. The dose is 1/100th of the no observed adverse effect level. A microdose can be quantified using the most sensitive ultra‐performance LC techniques or when labelled with [14C], as microtracer quantified by accelerator mass spectrometry. A prerequisite is dose linearity from the microdose up to therapeutic ranges. 115 In children, it has shown feasible to perform such studies which can also be used to study age‐related variation in drug and metabolite PK. Studies characterising paracetamol, midazolam and ursochol PK are published. 20 , 116 , 117 , 118 The recent FDA guidance on neonatal drug studies suggests to consider microdosing to explore ontogeny. 119 Moreover, a recent proof‐of‐concept study showed the feasibility to safely use a [14C] microtracer to elucidate paediatric metabolite pathways, which may address the challenges discussed above. 85
3. DEVELOPMENTAL PD
There is limited progress in knowledge on how growth and development impact PD. 4 To determine the effects of growth and development on PD, the greatest challenge is probably the robust and direct quantification of drug effects. Receptors for most drugs are not in the vascular space, but in the tissues. Because of technical and ethical constraints, such tissue compartments cannot be easily accessed repeatedly in children. 120 Therefore, PD evaluation usually depends on indirect measurements of drug action. To mitigate these challenges, researchers started to explore the use of (functional) biomarkers as tools for assessing developmental PD. 121 , 122 Biomarkers of PD as clinical substitutes for defining PD in paediatric patients are currently still limited. 123
Developmental PD investigates the age‐related maturation of biological systems and how this affects drug response (i.e. potency, efficacy or therapeutic range). We aim to illustrate that developmental aspects can be considered in drug targets and in PD endpoints, using 2 groups of conditions as examples (section 3.1 and 3.2), and in disease‐specific PD assessment (section 3.3). As these sections are based on illustrations, no generalizable conclusions can be drawn.
3.1. Identifying and testing treatment targets and effects in rare diseases
Basic science has elucidated the pathogenetic mechanisms of disorders that often present in the paediatric population, such as spinal muscular atrophy (SMA), Pompe's disease and fragile X syndrome. 124 , 125 , 126 This knowledge is relevant to paediatric drug development because it provides the opportunity to identify new drug targets (Table 1). Mechanism‐targeted drug development may rather lead to disease‐modifying therapies than the traditional symptom‐targeted approaches, at least in conditions with relatively simple mechanisms. 126 , 127 Nevertheless, for e.g. fragile X syndrome, specific treatment hypotheses were formulated following understanding of the imbalance between excitatory glutamatergic transmission and inhibitory GABAergic system, but clinical trials have not yet provided evidence of efficacy for the proposed treatments. 125 However, even in this setting, mechanism‐targeted drug development may serve to guide research. A specific challenge for paediatric drug development is identifying if and how a treatment response is dependent on the developmental stage of the child. Possible windows of opportunity for the treatment of disorders that often present in the paediatric population, adds to the complexity of paediatric pharmacology (Table 1). Even more challenging is drug development for conditions such as autism spectrum disorder, whose pathogenesis has not been clarified yet and is probably heterogeneous. In this area, there are ongoing trials based on hypotheses of neuronal transmission abnormalities. 138 , 139
TABLE 1.
Examples that developmental pharmacodynamics (PD) needs to be considered during drug development
| Developmental PD topic | Category | Examples |
|---|---|---|
|
Type of drug target Where the drug acts on? |
Mechanism‐targeted therapy |
‐ neuromuscular diseases: Building on the understanding of SMA genetics, novel compounds that specifically address the basic deficit, as nusinersen, were developed and approved for clinical use. 127 ‐ metabolic diseases: For Pompe's disease, caused by an enzyme deficiency, a targeted therapy is developed (enzyme replacement therapy with recombinant human α‐glucosidase from CHO cells (alglucosidase alfa). 126 |
|
Developmental changes in drug target Why is drug action different? |
Molecular |
‐ receptor structure: The GABA receptor subunit composition changes with age, and this explains paradoxical seizures in preterm infants and neonates when benzodiazepines are administered. 128 , 129 ‐ receptor density: Increased expression of the μ‐opioid receptor postnatally in rats is documented. Human studies are lacking, but an altered expression might explain why neonates are more sensitive to morphine. 128 ‐ receptor affinity: High concentrations of high‐affinity receptors for insulin are present on mononuclear leukocytes in neonatal cord blood (receptor affinity 5.9 × 108 M−1 vs. 2.9 × 108M−1 in neonates vs. adults) 130 |
| Biochemical | ‐ vancomycin AUC/MIC target: As only unbound drug can have pharmacological effect, this target should be based on unbound vancomycin. Maturational changes in vancomycin protein binding (i.e. median fraction unbound of 0.9 vs. 0.6 in neonates vs. adults) suggest altered AUC/MIC target throughout development. 131 | |
| Physiological |
‐ ontogeny of target organ system: Maturation of the adaptive immune system (CD4 T cell count) 132 and the coagulation system (vitamin K‐dependent coagulation factors) 133 explain differences in drug action with age. ‐ sensitivity of target organ system: The potency of the immunosuppressant cyclosporine at inhibiting peripheral blood monocytes proliferation and interleukin‐2 expression was 2‐fold and 7‐fold lower in infants (0–1 years) compared to older children. This might indicate altered target concentrations and dosing. 134 |
|
|
Age‐and disease‐specific PD endpoints and assessment tools Which drug effect to assess? How to assess drug effect? |
Endpoint category | ‐ vital signs: Blood pressure in children, and consequently the diagnostic thresholds for degree of hypertension, vary with age, height and sex. 135 |
| Assessment tools |
‐ for efficacy of psychoactive drugs (e.g. symptom assessment): Hyperactivity is the most apparent symptom of ADHD in young children, but this gradually attenuates with age while inattention, impulsivity, and deficits in organization and planning become evident features in adolescence. 136 ‐ for safety of psychoactive drugs (e.g. ADR assessment): Higher incidence of sedation reported as ADR of antipsychotics and antidepressants in children compared to adults, related to the underlying ADR mechanism (off‐target drug effect and sensitivity of paediatric subgroups to a specific ADR). 137 |
|
|
Therapeutic window of opportunity When to achieve optimal drug effect? |
Neurologic system | ‐ maturation of glutamatergic/GABAergic balance: Given the rapid changes in brain structure and function due to synaptogenesis and pruning during the first years of life, a treatment addressing the glutamatergic/GABAergic imbalance could have an effect at a younger age (e.g. until 36 months) but not if administered later. 128 , 129 |
SMA: spinal muscular atrophy, CHO: Chinese hamster ovary, GABA: γ‐aminobutyric acid, AUC: area under the concentration‐time curve, MIC: minimal inhibitory concentration, ADHD: attention deficit hyperactivity disorder, ADR: adverse drug reaction.
The evaluation of PD effects and the impact of growth and development are challenging to study in rare inherited diseases. The variable clinical phenotypes and time points of diagnosis, and the low incidence make it difficult to evaluate PD and separate the impact of maturation and natural disease course from other factors. Moreover, the drug of interest is usually the only treatment option, making placebo‐controlled designs ethically critical. Innovative methods can overcome these challenges. Clinical trial endpoints can be optimized by modelling natural history data (registries), 140 fallback tests for co‐primary endpoints, 141 and goal attainment scaling. 142 Confirmatory adaptive designs can minimize the number of needed patients and improve the significance of results. 143 The dilemma of placebo control in untreatable diseases can be addressed by delayed start randomization. 144 Furthermore, registry‐based studies can support risk–benefit analysis. 145
3.2. Developmental PD: The adaptive immune system as illustration
In the early days of human immunodeficiency virus (HIV) therapy it was thought that CD4 T cell count was an unreliable biomarker of disease progression in children younger than 5 years. 146 Despite having normal CD4 counts, infants and young children often had uncontrolled viral loads and poor clinical outcomes. This was due to the fact that thymic output displays age‐related activity, resulting in infants and young children having up to 5‐fold higher normal CD4 counts than adolescents. 132 , 147 By leveraging biological prior data on thymic output inferred from T‐cell receptor excision circles and T‐cell turnover inferred from Ki67 expression, 148 one can scale PD models for expected changes in T‐cell production and turnover. PD models using this scaling can be developed and applied in paediatric studies. 149 For the immunosuppressant cyclosporine, in vitro age‐dependent PD has been reported (Table 1). 134
3.3. Developmental PD: Neurological and psychiatric disorders as illustration
The structural and functional changes in the brain throughout infancy, childhood, adolescence and young adulthood are reflected in continuous modifications in the phenotypic manifestations of psychopathology (e.g. ADHD, Table 1). 136 It is important that symptom rating scales take this developmental trajectory into account. Developmentally appropriate assessment tools should be sufficiently sensitive and valid to treatment‐related effects in children with neurological disorders for use in clinical trials. In addition to disease‐specific core symptoms, also comorbidities may require their specific assessment tools. Besides efficacy, safety profiles can differ between children and adults. For antipsychotics and antidepressants, a significantly higher incidence and a different profile of adverse drug reactions (ADRs) was reported in children (including adolescents) compared to adults (Table 1). 137 Central nervous system changes including synaptogenesis, connectivity, and differential maturation of neurotransmitter systems continue throughout adolescence and can even take 3 decades. 150 , 151 This can also contribute to differential response to pharmacotherapy between adolescents and adults. 128
4. INNOVATIVE TOOLS FOR PAEDIATRIC DRUG DEVELOPMENT
Drug development plans not only need to incorporate the latest advances on developmental PK (section 2) and PD (section 3), but also innovative techniques. While some innovative approaches mentioned earlier (e.g. microsampling section 2.3) are already more established, the current section provides illustrations of experimental tools. Multidisciplinary collaboration creates opportunities to model pathophysiological processes, identify molecular targets and test pharmacotherapy. Table 2 gives examples of innovative tools available in the developmental pharmacology field.
TABLE 2.
Examples of innovative tools for paediatric drug development
| Tool | Aim | Example |
|---|---|---|
| Patient derived inducible pluripotent stem cells | To model cellular phenotypes of inherited diseases for drug discovery and characterization of pharmacodynamics |
Duchenne muscular dystrophy 152 Spinal muscular atrophy 153 |
| Organoids | To study molecular pathophysiology mechanisms, drug disposition and drug effects |
Nephrotic syndrome 154 Genetic kidney diseases, including cystinosis, ADPKD 155 Biliary atresia 158 |
| Juvenile animal models | To develop a preclinical model for paediatric drug research and a juvenile PBPK model: Maturational, or disease‐related |
Juvenile Göttingen minipig model ‐ for physiology data (maturational) 159 ‐ for perinatal asphyxia (disease) 160 |
| To study therapeutic strategies for developmental lung diseases | Juvenile rabbit model for BPD 161 | |
| To identify genes important in human traits and disorders | Zebrafish mutant model for ASD 162 | |
| Endogenous biomarkers | To assess endogenous metabolic activity reflecting drug metabolic activity | 4β–OHC and 6β‐OHF as surrogates for in vivo CYP3A4 activity 160 , 163 , 164 , 165 |
ADPKD: autosomal dominant polycystic kidney disease; PBPK: physiology‐based pharmacokinetics, BPD: bronchopulmonary dysplasia, ASD: autism spectrum disorder, 4β–OHC: 4‐β‐hydroxycholesterol, 6β‐OHF: 6‐β‐hydroxycortisol.
For understanding the ontogeny of the drug target organ system, in vitro research, and preclinical (animal) models can be implemented. For the latter, species‐specific differences in organ maturation, logistical disadvantages and imbalance between disease model and drug development are limitations to consider. 166 Finding preclinical (animal) models that display physiology and development comparable to human models is challenging. 167 Differences often relate to ADME characteristics, size or receptors. Although underexplored, juvenile pig models are of interest to investigate disease mechanisms (for neonates several [e.g. asphyxia, 168 necrotizing enterocolitis, 169 IUGR 170 and resuscitation 171 ] are described), and subsequently to assess PK/PD. Disposition of dexmedetomidine during hypothermia, and an approved paediatric investigation plan for 2‐iminobiotin, both in perinatal asphyxia pig models illustrate the potential and limitations of juvenile animal models. 172 Such data can be used to develop a PBPK framework, and allow to distinguish the impact of asphyxia vs. hypothermia on PK, which is not feasible in a clinical setting. 160
In section 4.1, we discuss opportunities and challenges of examples of preclinical cellular systems and additional animal disease models. Innovative methodology is also being developed to increase understanding of the ADME ontogeny. This will be illustrated in section 4.2 by selected endogenous biomarkers reflecting ADME relevant protein activity: the applicability of this work in paediatrics is limited, and this is considered as a research tool.
4.1. Examples of experimental and preclinical models for paediatric drug development
4.1.1. Ex vivo 3‐dimensional organotypic bone culture model
The growth plate (physis) is a hierarchic organized complex system and can be described as an organ. It is the site where elongation of the long bones occurs in longitudinal growth during development. Particularly in paediatrics and the growing bone, it is paramount to investigate (adverse) and/or osteogenic effects of drugs and biomaterial‐based devices on the physis and the growing skeleton in the biological growth mimicking environment. 173 , 174 Unfortunately, investigating growth at the physis is challenging since an optimal model is currently lacking and inconsistencies between in vitro and in vivo studies exist. The current in vitro models focus on the behaviour of developing osteoblast progenitors, osteoclasts or endothelial cells following drug administration. However, the events occurring in the growth plate during development are not mirrored in these in vitro models. 174 , 175 New approaches can be found in use of ex vivo 3‐dimensional organotypic cultures. 176 An ex vivo bone growth model is based on bone slices from femurs of early postnatal rats, cultured for several weeks, and can further be implemented in preclinical, toxicological and therapeutic investigations. 176
4.1.2. Mouse models for preclinical efficacy assessment
Mouse models can be used to study disease mechanisms in association with development and growth. While requirements are defined for toxicity assessment of new drugs, no regulation is available for preclinical pharmacology data used to support human trials. 177 Irrespective, an animal model can be useful for validation of target engagement (proof‐of‐concept studies). Clinically oriented preclinical studies can support assessment of efficacy also at young ages, support toxicology and dose‐finding studies. However, translation of the efficacy profile from mice to humans remains challenging, due to differences in (patho)physiology, the issue of age‐matching, presence of different genetic modifiers in mice and humans, and finally by the quality of preclinical data. 178 The absence of requirements for reporting standards in scientific publications has led to efforts in compiling facultative ARRIVE guidelines, 179 but the implementation of these guidelines is still very limited. 180 In the neuromuscular community, a (facultative) advisory board for clinical trials uncovered the challenges of applying results from poorly designed preclinical studies to human trials to improve translatability. 181 Scientific and clinical communities, and industry need to be aware of the risk of using inadequately validated mice model (or other animal model) data for premature translation to a human trial.
4.2. Endogenous biomarkers reflecting ADME relevant protein activity
Although their relevance is still controversial, endogenous biomarkers have been suggested as possible surrogates for enzymatic activity in adults. 182 Examples are plasma 4β‐hydroxycholesterol (4β–OHC, with 4β‐OHC/cholesterol ratio) and urinary 6β‐hydroxycortisol (6β‐OHF, with 6β‐OHF/cortisol ratio) for CYP3A4 activity. It is important to know that paediatric data on these markers are only explorative.
Developmental patterns are described: at birth, neonates have lower 4β‐OHC, but the level increased significantly between birth and age 4 months to reach adult values. However, a median 4β‐OHC/cholesterol ratio at birth (0.19) was already comparable to adults, and this ratio did not change until 4 months. 163 In children (age 1–17 y) with epilepsy, carbamazepine induces CYP3A4/5, with subsequent 4β‐OHC increase. 183 Takaki et al. demonstrated that several oxysterols in urine and plasma undergo developmental changes and may be promising candidates for becoming biomarkers for paediatric liver disease. 184 Also 6β‐OHF levels display age‐dependency, with maximum values at 14–20 years for both sexes. 164 Concerning 6β‐OHF/cortisol ratios, higher values are reported in neonates (PNA 1–15 days) compared to infants (30–359 days), 185 while others found different results. 186 A correlation between this ratio and GA and birth weight is described. 186 The ratio at birth in preterm infants was lower than term cases and remained stable the first 14 days, while in term neonates the ratio decreased after birth to preterm levels at PNA 5 days. This ratio further declines with age and remains stable from 21–25 years onwards. 164
Following (i) confirmation of the patterns observed and (ii) demonstration of the correlation with DME activity, these data might inform future PBPK models for drugs undergoing metabolic elimination. They may contribute to define changes in DME activity due to strong inducers/inhibitors.
Based on a recent systems biology analysis, 187 several endogenous compounds were listed as substrates for human drug transporters of the SLC (solute carrier) family. These compounds might be candidates for future biomarkers of in vivo activities of various SLC isoforms in children. Finally, testotsterone glucuronide normalized by androsterone glucuronide seems a promising urinary UGT2B17 biomarker in children. In line with the developmental pattern of UGT2B17 expression, this marker was significantly associated with sex, age and copy number variation. 188
5. REFLECTIONS ON HOW KNOWLEDGE REGARDING DEVELOPMENTAL PHARMACOLOGY SHOULD BE IMPLEMENTED IN THE PAEDIATRIC DRUG DEVELOPMENT PATHWAY
Due to the anticipated relevance of developmental PK and PD on the design of paediatric clinical trials, there is regulatory guidance and a framework to support paediatric drug development. 189 This has been converted in a paediatric decision tree (Figure 1). 190
FIGURE 1.

Paediatric study decision tree with identified scenarios (from Manolis and Pons, Br J Clin Pharmacol 2009, 190 with permission from Rightslink, John Wiley and Sons and Copyright Clearance Center). PK, pharmacokinetics; PD pharmacodynamics
The use of this pathway depends on the availability of an underlying rationale as to what can or cannot be extrapolated from adult to paediatric data and between different paediatric age groups. Bridging can be done by extrapolation, modelling and simulation and has been used for regulatory evaluation. 190 , 191 Based on assumptions on (dis)similarities in disease and concentration–response profiles, PK, PD and safety data are collected to design the paediatric drug development programme. Data sources may include adult studies, paediatric studies, preclinical (animal) models, in vitro and in silico models. 192 Ollivier et al. described the shift that occurred within paediatric pharmacotherapy from implicit (subjective, driven by practical experience, resulting in eminence‐based off‐label use), to explicit (based on and driven by scientific rationale) extrapolation. 104 This shift on how to approach off‐label drug use is also reflected in the joint policy statement of the European Academy of Paediatrics and the ESDPPP. 193
The studies used to construct the paediatric programme need to be planned early in the adult programme and may be included in the adult programme (e.g. identifying dose‐exposure‐response relationships). Understanding developmental pharmacology is needed to plan and interpret these studies to deploy appropriate methods and existent data.
The goal of developmental pharmacology is to have sufficient evidence about the disposition and effects of drugs to inform and support authorization and accurate pharmacotherapy in children. Large, traditional clinical trials are tools to gather data and knowledge, but are not always the best approach to fill these knowledge gaps. First, in the presence of a well‐characterized condition and strong evidence for similar drug effects across age groups, well‐justified extrapolation supported by modern tools can generate a similar strength of dosing and efficacy evidence as large clinical trials. Modern tools include pharmacometrics, PBPK, and clinical biomarkers. Extrapolation requires fewer patients than required for large trials, as illustrated for HIV or partial‐onset seizures, 194 and can also reduce the number of samples per patient. Second, when efficacy trials are done, integrating trial‐derived and pharmacometrics‐derived results can reduce the uncertainties that the trials address, e.g. refinement towards more targeted and efficient designs. The international paediatric multiple sclerosis study group recommends reconsidering paediatric study designs (including studying safety instead of efficacy and the ethical considerations of using placebo for a highly active disease), as disease mechanisms are shared across the age span. 195 This study group illustrates that knowledge regarding developmental pharmacology can be combined with drug information to justify recruitment of adolescents (or children) to adult studies as long as sufficient information on drug and target population (including ontogeny) is present. This approach is endorsed in FDA recommendations about study eligibility criteria that suggest increasing diversity in patient enrolment, including children, pregnant or lactating women in e.g. confirmatory clinical trials. 196 However, crucial to such an inclusive approach is that developmental PK and PD characterization are considered from the earliest drug development stages. Similarly, sequential enrolment of paediatric age groups, moving to younger age groups only when studies in an older age group have demonstrated safety and efficacy should no longer be taken for granted, unless justified and based on clear scientific rationale such as potential developmental safety concern. Arbitrary sequential enrolment of paediatric subgroups (from older, going to younger ages) in drug development has been questioned in both EMA and FDA, as this may lead to delays in data availability, especially in the younger ages. 96 , 196 This may result in prolonged off‐label practices in these subgroups, with subsequent difficulties to conduct relevant trials in these groups, once the drug is marketed.
This is because sufficient information to justify starting studies in multiple age‐groups can emerge from modelling and simulation. 196 Because of the more limited information on ontogeny and pathophysiology, Ollivier et al. stated that extrapolation remains most difficult in (pre)term neonates. 104 , 194 Depending on the neonatal drug development programme, each of the earlier mentioned data sources (adult studies, paediatric studies, preclinical (animal) models, in vitro and in silico models) can be considered to varying degrees and levels of confidence to guide dosing. Using an illustrative approach, neonatal programmes on meropenem (disease similar to children or adults), clopidogrel or thyroid hormone (diseases related but not similar) or caffeine and surfactants (diseases unique to neonates) have been described. 192
5.1. Paediatric drug development pathway, challenges related to PK and PD
PK data are crucial to paediatric drug development as exposure will determine (side‐) effects (Figure 1). However, this is not unique to children, so that sufficient adult—or rarely—preclinical (animal) data (section 4.1, Table 2) should be gathered to understand the concentration–time profile (PK) and optimal concentration–response (PD) profile. 192 Repurposing is an important setting, as another indication may require new dose‐findings studies, like ibuprofen for closure of a patent ductus arteriosus, or sildenafil for treatment of pulmonary hypertension. 197 , 198 The earlier mentioned modelling and simulation or PBPK modelling should provide supportive guidance on conducting and designing PK studies, to reduce individual burden (e.g. number and volume of samples, sample technique, opportunistic or scavenged, time window, maximize flexible sampling approach). This should be based on a priori simulations that justify the approach taken and maximize the information used as the source for extrapolation.
Characterizing the impact of development on subsequent PD is still challenging. Paediatric biomarkers should be sufficiently sensitive to discriminate time‐dependent changes from medicine‐ or intervention‐related effects to enable modelling of drug–response relationships across the paediatric spectrum. 4 Again, modelling and simulation can support trial design and conduct, while strategies related to enrichment (e.g. treatments for SMA targeting type I vs. II vs. III, or cystic fibrosis drug modulator therapies, targeted to a specific mutation) can also be useful. 196 Robust modelling efforts necessitate rich data on e.g. the natural disease course and functional outcome variables while considering impact of maturation/development (e.g. sharing of clinical research data on SMA, or natural trend data of lung function tests in cystic fibrosis), most relevant in the field of rare diseases. 199 , 200 We should focus on patient (and when relevant parent/family)‐centred outcome variables, using approaches to include children and young people's opinions. 201 , 202
5.2. Paediatric drug development pathway, challenges related to safety
Drug safety remains 1 of the key elements of the benefit/risk balance assessment by regulatory authorities, health care providers and the public. Standardization of pharmacovigilance and safety is key to facilitate comparison and is embedded in guidance documents. This resulted in regulatory requirements to assess seriousness, causality and severity, irrespective of the population considered, but all have their issues when applied in paediatrics.
Serious adverse event outcomes are clearly defined as including: death, life‐threatening events, inpatient hospitalization or prolongation of hospitalization, persistent or significant incapacity or disruption of daily life functions, or a congenital anomaly. However, it may be difficult to disentangle prolonged hospitalization in specific subpopulations such as preterm neonates. 203 Causality assessment is another issue, as e.g. the Naranjo algorithm or Uppsala causality tool are not tailored to all paediatric subpopulations. 204 To stress the relevance of availability of these tools, the inter‐rater reliability for different tools when applied to suspected ADRs in neonates remained fair (κ ~0.3). 205 Similar issues exist for severity assessment, subdivided into mild, moderate, severe, life threatening or death (grade 1–5). To limit inter‐rater variability, consensus documents related to e.g. vaccine trials (Division of Acquired Immune Deficiency Syndrome—table, adults, paediatrics), oncology (Common Terminology Criteria for Adverse Events, adults and paediatrics) or, recently, neonatology have been reported. 206 , 207 , 208 , 209 To illustrate their relevance, grading in part depends on the impact on age‐appropriate instrumental activities of daily living. To limit variability and standardize event assessment, the impact on daily living should be translated to a population‐specific setting, such as reported by the generic severity criteria for the neonatal ADR scale. 206
It is important that safety biomarkers used in drug development programmes, also display age‐ and non–age‐related differences in patterns or quantification. QT(c) times are commonly used biomarkers to prevent cardiac arrhythmias, but these markers display maturational and nonmaturational changes. 210 Recognition and quantification of adverse events of the kidney necessitate age‐appropriate reference intervals of kidney function and injury markers. 211 , 212 Besides these ‘general’ safety biomarkers, growth, pubertal development (and reproduction) or neurocognitive and behavioural development are of specific relevance to this population.
6. GENERAL SUGGESTIONS AND CONCLUSION
Knowledge on developmental pharmacology is rapidly expanding, and should be captured in paediatric drug development to achieve safe, effective and individualized pharmacotherapy. A few general suggestions can be made:
To reduce knowledge gaps in developmental PK, profiling drug metabolites can support mechanistic PK studies and provide insight into in vivo impact of ontogeny of elimination pathways, while minimal risk and burden designs allow for optimally informative but minimally invasive PK sampling. In addition, integration of innovative methodology to better understand ADME ontogeny, can provide paediatric drug development initiatives with more profound data on drug disposition in paediatric subgroups. However, limitations of currently underexplored preclinical models and still explorative biomarkers need to be considered. Furthermore, linking exposure (PK) to effect (PD) remains difficult. Clearly establishing robust dose–response relationships is important before exposure‐based individualized dosing can be pursued. We are aware that this paper focusses on maturational factors impacting PK/PD. Nonmaturational factors further contribute to inter‐ and intrapatient PK/PD variability. As impact of both types of factors cannot always be distinguished from each other in clinical trials, preclinical models might offer add‐on benefit (e.g. to separate the impact of asphyxia vs. hypothermia on PK). 160
In addition to developmental PK, PD data are even more relevant, but difficult to collect, as drug efficacy and safety are often considered as final endpoints of pharmacotherapy. Therefore, real world paediatric patient data (e.g. laboratory results from routine clinical care, registries, electronic health files) are useful to support paediatric PD endpoints. 213 Based on disease‐ and target organ‐specific examples, we illustrated that developmental PD should be considered during drug development. However, many age‐ and disease‐specific, validated markers and references values covering the paediatric range are currently lacking for a reliable PD assessment. Finally, when short‐term efficacy and safety data become more evident, focus on long‐term clinical data on paediatric pharmacotherapy is appropriate.
The Göttingen minipig is the reference pig strain for PK studies in drug development, but its use remains limited. 166 Regulatory guidelines recommend to use the same species in juvenile and adult toxicity studies. However, species selection for adult studies is not clearly defined and lacks consistency. Optimal selection criteria would probably increase Göttingen minipig use for adult toxicity studies. Also for drug indications limited to children, this strain is the preferred model for preclinical PK and toxicity studies. 166 Further ADME research and PBPK models for this species are needed. 214
PK and PD characterization in paediatric subgroups should be considered from the earliest drug development stages. To uniform trial‐related approaches, the use of recent instruments e.g. the paediatric decision tree for extrapolation (Figure 1), or assessments of seriousness, causality and severity of adverse effects in paediatric drug development is suggested.
Continued improvements on targeted and efficient clinical trial designs should become standard in paediatric drug development. 189 , 206 As highlighted in section 5, some of these reflections are supported by scientific societies on developmental pharmacology (e.g. European Academy of Paediatrics, ESDPPP) and/or regulatory agencies (FDA, European Medicines Agency). In addition, developmental pharmacology expertise, as part of strategic feasibility—such as the advice offered by the European public‐private conect4children consortium—should be integrated in paediatric drug development. We hereby also want to stress the importance of shifting from an arbitrary sequential enrolment of paediatric subgroups to more rational study designs.
In conclusion, this paper summarizes current evidence of developmental PK and PD, and provides scientific insights and suggestions to incorporate the latest knowledge on developmental pharmacology and innovative techniques into paediatric drug development. The developmental pharmacology research field should be driven by developmental (patho)physiology, and multidisciplinary collaboration between academia, industry, regulatory agencies, health‐care workers, patients and parents. As an illustration on how to put this call to action, conect4children project (www.conect4children.org) initiated a strategic feasibility advice service to build the bridge between all these partners involved. The aim is to provide innovative methodology advice, including on developmental pharmacology and sharing the latest knowledge, e.g. through this White Paper.
DISCLAIMER
The publication reflects the author's view and neither IMI nor the European Union, EFPIA, or any Associated Partners are responsible for any use that may be made of the information contained therein.
[Correction added on 8 August 2021, after first online publication: Disclaimer has been added in this current version.]
COMPETING INTERESTS
This paper reflects a collaboration between researchers from the IMI c4c Expert group on Developmental Pharmacology, and the ESDPPP. All co‐authors are member of IMI c4c and/or ESDPPP. S.N.W. is co‐chair of the IMI2 C4C strategic feasibility expert work package and director of the secretariat. The authors have no other competing interests to declare for this paper.
CONTRIBUTORS
A.S. conceptualized and drafted the paper. All co‐authors (A.S., P.A., G.C., P.D.C., S.d.W., J.M.K., F.B.L., C.M., P.P., M.F.S., J.S., M.A.T., B.V., W.Z., A.M.W., R.W., J.v.d.A. and K.A.) contributed to writing and revision of the paper. K.A. provided intermittent review. A.S., R.W., S.d.W., J.v.d.A., M.A.T. and K.A. critically revised the prefinal version. All co‐authors (A.S., P.A., G.C., P.D.C., S.d.W., J.M.K., F.B.L., C.M., P.P., M.F.S., J.S., M.A.T., B.V., W.Z., A.M.W., R.W., J.v.d.A. and K.A.) revised the paper draft and approved the final text version.
ACKNOWLEDGEMENTS
The research activities of A.S. are supported by the Clinical Research and Education Council of the University Hospitals Leuven. W.Z. received funding from Young Taishan Scholars Program of Shandong Province and National Science and Technology Major Projects for “Major New Drugs Innovation and Development” (2017ZX09304029‐002). For P.P., the project “International Mobility of Researchers at Charles University” (reg. n. CZ.02.2.69/0.0/0.0/16_027/0008495) is supported by the Operational Program Research, Development and Education.
The c4c project has received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement No 777 389. The Joint Undertaking receives support from the European Union's Horizon 2020 research and innovation programme and EFPIA.
A.M.W. is consultant to the company Bioretec Oy (Finland). In the last 2 years, B.V. has received consultant fees or honoraria from Medice, Lundbeck, and Angelini Pharmaceuticals, and from lawfirms Goodwin & Procter, Haynes & Boone. S.N.W. is consultant for Khondrion and receives clinical trial funding from GSK and Pfizer. C.M. has served as consultant, advisor or CME speaker unrelated to the present work for Janssen, Angelini, Servier, Nuvelution, Otsuka, Lundbeck and Esteve.
Smits A, Annaert P, Cavallaro G, et al. Current knowledge, challenges and innovations in developmental pharmacology: A combined conect4children Expert Group and European Society for Developmental, Perinatal and Paediatric Pharmacology White Paper. Br J Clin Pharmacol. 2022;88(12):4965-4984. 10.1111/bcp.14958
On behalf of the Innovative Medicines Initiative conect4children (IMI c4c) Expert group on Developmental Pharmacology and the European Society for Developmental, Perinatal and Paediatric Pharmacology (ESDPPP)
Funding information AMW is consultant to the company Bioretec Oy (Finland); CM has served as consultant, advisor or CME speaker unrelated to the present work for Janssen, Angelini, Servier, Nuvelution, Otsuka, Lundbeck and Esteve; In the last 2 years, BV has received consultant fees or honoraria from Medice, Lundbeck, and Angelini Pharmaceuticals, and from lawfirms Goodwin & Procter, Haynes & Boone.; SNW is consultant for Khondrion and receives clinical trial funding from GSK and Pfizer.; The conect4children (c4c) project has received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement No 777389. The Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation program and EFPIA., Grant/Award Number: 777389
REFERENCES
- 1. Hales CM, Kit BK, Gu Q, Ogden CL. Trends in Prescription Medication Use Among Children and Adolescents‐United States, 1999‐2014. JAMA. 2018;319(19):2009‐2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Chai G, Governale L, McMahon AW, Trinidad JP, Staffa J, Murphy D. Trends of outpatient prescription drug utilization in US children, 2002‐2010. Pediatrics. 2012;130(1):23‐31. [DOI] [PubMed] [Google Scholar]
- 3. Kearns GL, Abdel‐Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology‐‐drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157‐1167. [DOI] [PubMed] [Google Scholar]
- 4. van den Anker J, Reed MD, Allegaert K, Kearns GL. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J Clin Pharmacol. 2018;58(Suppl 10):S10‐S25. [DOI] [PubMed] [Google Scholar]
- 5. Krekels EHJ, van Hasselt JGC, van den Anker JN, Allegaert K, Tibboel D, Knibbe CAJ. Evidence‐based drug treatment for special patient populations through model‐based approaches. Eur J Pharm Sci. 2017;109S:S22‐S26. [DOI] [PubMed] [Google Scholar]
- 6. Allegaert K, Simons SHP, Tibboel D, Krekels EH, Knibbe CA, van den Anker JN. Non‐maturational covariates for dynamic systems pharmacology models in neonates, infants, and children: Filling the gaps beyond developmental pharmacology. Eur J Pharm Sci. 2017;109S:S27‐S31. [DOI] [PubMed] [Google Scholar]
- 7. conect4children IMI. https://www.imi.europa.eu/projects-results/project-factsheets/c4c. Accessed December 5, 2020.
- 8. European Society for Developmental Perinatal and Paediatric Pharmacology https://www.esdppp.org/. Accessed December 5, 2020.
- 9. Sridharan K, Hasan H, Al Jufairi M, Al Daylami A. Abdul Azeez Pasha S, Al Ansari E. Drug utilisation in adult, paediatric and neonatal intensive care units, with an emphasis on systemic antimicrobials. Anaesthesiol Intensive Ther. 2021;53(1):18‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Nicolas JM, Bouzom F, Hugues C, Ungell AL. Oral drug absorption in pediatrics: the intestinal wall, its developmental changes and current tools for predictions. Biopharm Drug Dispos. 2017;38(3):209‐230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Mooij MG, Nies AT, Knibbe CA, et al. Development of Human Membrane Transporters: Drug Disposition and Pharmacogenetics. Clin Pharmacokinet. 2016;55(5):507‐524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Allegaert K, van den Anker J. Ontogeny of Phase I Metabolism of Drugs. J Clin Pharmacol. 2019;59(Suppl 1):S33‐S41. [DOI] [PubMed] [Google Scholar]
- 13. Van Den Abeele J, Rayyan M, Hoffman I, Van de Vijver E, Zhu W, Augustijns P. Gastric fluid composition in a paediatric population: Age‐dependent changes relevant for gastrointestinal drug disposition. Eur J Pharm Sci. 2018;123:301‐311. [DOI] [PubMed] [Google Scholar]
- 14. Mooij MG, de Koning BA, Huijsman ML, de Wildt SN. Ontogeny of oral drug absorption processes in children. Expert Opin Drug Metab Toxicol. 2012;8(10):1293‐1303. [DOI] [PubMed] [Google Scholar]
- 15. Huang NN, High RH. Comparison of serum levels following the administration of oral and parenteral preparations of penicillin to infants and children of various age groups. J Pediatr. 1953;42(6):657‐658. [DOI] [PubMed] [Google Scholar]
- 16. Kresser M. On a simple and reliable method for the determination of free hemoglobin in the plasma. Dtsch Gesundheitsw. 1967;22(48):2270‐2271. [PubMed] [Google Scholar]
- 17. McClean P, Weaver LT. Ontogeny of human pancreatic exocrine function. Arch Dis Child. 1993;68(1SpecNo):62‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Fakhoury M, Litalien C, Medard Y, et al. Localization and mRNA expression of CYP3A and P‐glycoprotein in human duodenum as a function of age. Drug Metab Dispos. 2005;33(11):1603‐1607. [DOI] [PubMed] [Google Scholar]
- 19. Mooij MG, Schwarz UI, de Koning BA, et al. Ontogeny of human hepatic and intestinal transporter gene expression during childhood: age matters. Drug Metab Dispos. 2014;42(8):1268‐1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. van Groen BD, Krekels EHJ, Mooij MG, et al. The Oral Bioavailability and Metabolism of Midazolam in Stable Critically Ill Children: A Pharmacokinetic Microtracing Study. Clin Pharmacol Ther. 2020;109(1):140‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Blake MJ, Abdel‐Rahman SM, Pearce RE, Leeder JS, Kearns GL. Effect of diet on the development of drug metabolism by cytochrome P‐450 enzymes in healthy infants. Pediatr Res. 2006;60(6):717‐723. [DOI] [PubMed] [Google Scholar]
- 22. Frommelt P, Juern A, Siegel D, et al. Adverse Events in Young and Preterm Infants Receiving Topical Timolol for Infantile Hemangioma. Pediatr Dermatol. 2016;33(4):405‐414. [DOI] [PubMed] [Google Scholar]
- 23. Lemes LR, Melo DF, de Oliveira DS, de La‐Rocque M, Zompero C, Ramos PM. Topical and oral minoxidil for hair disorders in pediatric patients: What do we know so far? Dermatol Ther. 2020;e13950. [DOI] [PubMed] [Google Scholar]
- 24. Sethi PK, White CA, Cummings BS, Hines RN, Muralidhara S, Bruckner JV. Ontogeny of plasma proteins, albumin and binding of diazepam, cyclosporine, and deltamethrin. Pediatr Res. 2016;79(3):409‐415. [DOI] [PubMed] [Google Scholar]
- 25. Adeli K, Higgins V, Trajcevski K, White‐Al Habeeb N. The Canadian laboratory initiative on pediatric reference intervals: A CALIPER white paper. Crit Rev Clin Lab Sci. 2017;54(6):358‐413. [DOI] [PubMed] [Google Scholar]
- 26. Smits A, Roberts JA, Vella‐Brincat JW, Allegaert K. Cefazolin plasma protein binding in different human populations: more than cefazolin‐albumin interaction. Int J Antimicrob Agents. 2014;43(2):199‐200. [DOI] [PubMed] [Google Scholar]
- 27. Oyaert M, Spriet I, Allegaert K, et al. Factors impacting unbound vancomycin concentrations in different patient populations. Antimicrob Agents Chemother. 2015;59(11):7073‐7079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Pullen J, Stolk LM, Degraeuwe PL, van Tiel FH, Neef C, Zimmermann LJ. Protein binding of flucloxacillin in neonates. Ther Drug Monit. 2007;29(3):279‐283. [DOI] [PubMed] [Google Scholar]
- 29. Painter MJ, Minnigh MB, Gaus L, Scher M, Brozanski B, Alvin J. Neonatal phenobarbital and phenytoin binding profiles. J Clin Pharmacol. 1994;34(4):312‐317. [DOI] [PubMed] [Google Scholar]
- 30. Kan M, Wu YE, Li X, et al. An adapted LC‐MS/MS method for the determination of free plasma concentration of cefoperazone in children: Age‐dependent protein binding. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1144:122081. [DOI] [PubMed] [Google Scholar]
- 31. Friis‐Hansen B. Water distribution in the foetus and newborn infant. Acta Paediatr Scand Suppl. 1983;305:7‐11. [DOI] [PubMed] [Google Scholar]
- 32. Langer T, Limuti R, Tommasino C, et al. Intravenous fluid therapy for hospitalized and critically ill children: rationale, available drugs and possible side effects. Anaesthesiol Intensive Ther. 2018;50(1):49‐58. [DOI] [PubMed] [Google Scholar]
- 33. Haroun D, Wells JC, Williams JE, Fuller NJ, Fewtrell MS, Lawson MS. Composition of the fat‐free mass in obese and nonobese children: matched case‐control analyses. Int J Obes (Lond). 2005;29(1):29‐36. [DOI] [PubMed] [Google Scholar]
- 34. Collaboration NCDRF . Worldwide trends in body‐mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population‐based measurement studies in 128.9 million children, adolescents, and adults. Lancet. 2017;390(10113):2627‐2642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Mulla H, Johnson TN. Dosing dilemmas in obese children. Arch Dis Child Educ Pract Ed. 2010;95(4):112‐117. [DOI] [PubMed] [Google Scholar]
- 36. Ghersi‐Egea JF, Saudrais E, Strazielle N. Barriers to Drug Distribution into the Perinatal and Postnatal Brain. Pharm Res. 2018;35(4):84. [DOI] [PubMed] [Google Scholar]
- 37. Verscheijden LFM, van Hattem AC, Pertijs J, et al. Developmental patterns in human blood‐brain barrier and blood‐cerebrospinal fluid barrier ABC drug transporter expression. Histochem Cell Biol. 2020;154(3):265‐273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. D'Agate S, Musuamba FT, Della PO. Dose Rationale for Amoxicillin in Neonatal Sepsis When Referral Is Not Possible. Front Pharmacol. 2020;11:521933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Vaisman N, Corey M, Rossi MF, Goldberg E, Pencharz P. Changes in body composition during refeeding of patients with anorexia nervosa. J Pediatr. 1988;113(5):925‐929. [DOI] [PubMed] [Google Scholar]
- 40. Raffaeli G, Pokorna P, Allegaert K, et al. Drug Disposition and Pharmacotherapy in Neonatal ECMO: From Fragmented Data to Integrated Knowledge. Front Pediatr. 2019;7:360. 10.3389/fped.2019.00360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Zimmerman KO, Dallefeld SH, Hornik CP, Watt KM. Sedative and Analgesic Pharmacokinetics During Pediatric ECMO. J Pediatr Pharmacol Ther. 2020;25(8):675‐688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Ladumor MK, Bhatt DK, Gaedigk A, et al. Ontogeny of Hepatic Sulfotransferases and Prediction of Age‐Dependent Fractional Contribution of Sulfation in Acetaminophen Metabolism. Drug Metab Dispos. 2019;47(8):818‐831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Bhatt DK, Mehrotra A, Gaedigk A, et al. Age‐ and Genotype‐Dependent Variability in the Protein Abundance and Activity of Six Major Uridine Diphosphate‐Glucuronosyltransferases in Human Liver. Clin Pharmacol Ther. 2019;105(1):131‐141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. van Groen BD, Nicolai J, Kuik AC, et al. Ontogeny of Hepatic Transporters and Drug‐Metabolizing Enzymes in Humans and in Nonclinical Species. Pharmacol Rev. 2021;73(2):597‐678. [DOI] [PubMed] [Google Scholar]
- 45. Salem F, Johnson TN, Abduljalil K, Tucker GT, Rostami‐Hodjegan A. A re‐evaluation and validation of ontogeny functions for cytochrome P450 1A2 and 3A4 based on in vivo data. Clin Pharmacokinet. 2014;53(7):625‐636. [DOI] [PubMed] [Google Scholar]
- 46. 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):625‐634. [DOI] [PubMed] [Google Scholar]
- 47. de Wildt SN, Kearns GL, Leeder JS, van den Anker JN. Cytochrome P450 3A: ontogeny and drug disposition. Clin Pharmacokinet. 1999;37(6):485‐505. [DOI] [PubMed] [Google Scholar]
- 48. Mooij MG, van Duijn E, Knibbe CAJ, et al. Successful Use of [(14)C]Paracetamol Microdosing to Elucidate Developmental Changes in Drug Metabolism. Clin Pharmacokinet. 2017;56(10):1185‐1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hines RN. Ontogeny of human hepatic cytochromes P450. J Biochem Mol Toxicol. 2007;21(4):169‐175. [DOI] [PubMed] [Google Scholar]
- 50. Vet NJ, Brussee JM, de Hoog M, et al. Inflammation and Organ Failure Severely Affect Midazolam Clearance in Critically Ill Children. Am J Respir Crit Care Med. 2016;194(1):58‐66. [DOI] [PubMed] [Google Scholar]
- 51. Gijsen V, Mital S, van Schaik RH, et al. Age and CYP3A5 genotype affect tacrolimus dosing requirements after transplant in pediatric heart recipients. J Heart Lung Transplant. 2011;30(12):1352‐1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zhao W, Leroux S, Biran V, Jacqz‐Aigrain E. Developmental pharmacogenetics of CYP2C19 in neonates and young infants: omeprazole as a probe drug. Br J Clin Pharmacol. 2018;84(5):997‐1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Lutz IC, Allegaert K, de Hoon JN, Marynissen H. Pharmacokinetics during therapeutic hypothermia for neonatal hypoxic ischaemic encephalopathy: a literature review. BMJ Paediatr Open. 2020;4(1):e000685. 10.1136/bmjpo-2020-000685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Johnson TN, Jamei M, Rowland‐Yeo K. How Does In Vivo Biliary Elimination of Drugs Change with Age? Evidence from In Vitro and Clinical Data Using a Systems Pharmacology Approach. Drug Metab Dispos. 2016;44(7):1090‐1098. [DOI] [PubMed] [Google Scholar]
- 55. Anderson BJ, Allegaert K. The pharmacology of anaesthetics in the neonate. Best Pract Res Clin Anaesthesiol. 2010;24(3):419‐431. [DOI] [PubMed] [Google Scholar]
- 56. De Cock RF, Allegaert K, Brussee JM, 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):2643‐2654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Rhodin MM, Anderson BJ, Peters AM, et al. Human renal function maturation: a quantitative description using weight and postmenstrual age. Pediatr Nephrol. 2009;24(1):67‐76. [DOI] [PubMed] [Google Scholar]
- 58. 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. 2020;61(2):159‐171. 10.1002/jcph.1725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Cristea S, Krekels EHJ, Rostami‐Hodjegan A, Allegaert K, Knibbe CAJ. The Influence of Drug Properties and Ontogeny of Transporters on Pediatric Renal Clearance through Glomerular Filtration and Active Secretion: a Simulation‐Based Study. AAPS J. 2020;22(4):87. 10.1208/s12248-020-00468-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Schreuder MF, Bueters RR, Allegaert K. The interplay between drugs and the kidney in premature neonates. Pediatr Nephrol. 2014;29:2083‐2091. [DOI] [PubMed] [Google Scholar]
- 61. Cheung KWK, van Groen BD, Spaans E, et al. A Comprehensive Analysis of Ontogeny of Renal Drug Transporters: mRNA Analyses, Quantitative Proteomics, and Localization. Clin Pharmacol Ther. 2019;106(5):1083‐1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Cheung KWK, van Groen BD, Burckart GJ, Zhang L, de Wildt SN, Huang SM. Incorporating Ontogeny in Physiologically Based Pharmacokinetic Modeling to Improve Pediatric Drug Development: What We Know About Developmental Changes in Membrane Transporters. J Clin Pharmacol. 2019;59(Suppl 1):S56‐S69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Dhont E, Van Der Heggen T, De Jaeger A, Vande Walle J, De Paepe P, De Cock PA. Augmented renal clearance in pediatric intensive care: are we undertreating our sickest patients? Pediatr Nephrol. 2020;35(1):25‐39. [DOI] [PubMed] [Google Scholar]
- 64. Schijvens AM, de Wildt SN, Schreuder MF. Pharmacokinetics in children with chronic kidney disease. Pediatr Nephrol. 2020;35(7):1153‐1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Sharma D, Shastri S, Farahbakhsh N, Sharma P. Intrauterine growth restriction ‐ part 1. J Matern Fetal Neonatal Med. 2016;29(24):3977‐3987. [DOI] [PubMed] [Google Scholar]
- 66. Sharma D, Farahbakhsh N, Shastri S, Sharma P. Intrauterine growth restriction ‐ part 2. J Matern Fetal Neonatal Med. 2016;29(24):4037‐4048. [DOI] [PubMed] [Google Scholar]
- 67. Boehm G, Teichmann B, Krumbiegel P. Hepatic biotransformation capacity in low‐birth‐weight infants as measured with the [15N]methacetin urine test: influences of gestational age, postnatal age, and intrauterine growth retardation. Biol Neonate. 1995;68(1):19‐25. [DOI] [PubMed] [Google Scholar]
- 68. Soo JY, Wiese MD, Berry MJ, McMillen IC, Morrison JL. Intrauterine growth restriction may reduce hepatic drug metabolism in the early neonatal period. Pharmacol Res. 2018;134:68‐78. [DOI] [PubMed] [Google Scholar]
- 69. Engbers AGJ, Flint RB, Voller S, et al. Enantiomer specific pharmacokinetics of ibuprofen in preterm neonates with patent ductus arteriosus. Br J Clin Pharmacol. 2020;86(10):2028‐2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Luyckx VA, Perico N, Somaschini M, et al. A developmental approach to the prevention of hypertension and kidney disease: a report from the Low Birth Weight and Nephron Number Working Group. Lancet. 2017;390(10092):424‐428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Frattarelli DA, Ergun H, Lulic‐Botica M, Lehr VT, Aranda JV. Vancomycin elimination in human infants with intrauterine growth retardation. Pediatr Infect Dis J. 2005;24(11):979‐983. [DOI] [PubMed] [Google Scholar]
- 72. Lulic‐Botica M, Sheer T, Edwards D, Thomas RL, Natarajan G. Impact of small‐for‐gestational age (SGA) status on gentamicin pharmacokinetics in neonates. J Clin Pharmacol. 2014;54(1):39‐45. [DOI] [PubMed] [Google Scholar]
- 73. Allegaert K, Anderson BJ, van den Anker JN, Vanhaesebrouck S, de Zegher F. Renal drug clearance in preterm neonates: relation to prenatal growth. Ther Drug Monit. 2007;29(3):284‐291. [DOI] [PubMed] [Google Scholar]
- 74. Smits A, De Cock P, Vermeulen A, Allegaert K. Physiologically based pharmacokinetic (PBPK) modeling and simulation in neonatal drug development: how clinicians can contribute. Expert Opin Drug Metab Toxicol. 2019;15(1):25‐34. [DOI] [PubMed] [Google Scholar]
- 75. Kennedy M. Hormonal regulation of hepatic drug‐metabolizing enzyme activity during adolescence. Clin Pharmacol Ther. 2008;84(6):662‐673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Kearns GL, van den Anker JN. Adolescents and Drug Development: Commentary on a Dawning Paradigm Shift. J Clin Pharmacol. 2021;61(6):740‐743. 10.1002/jcph.1862 [DOI] [PubMed] [Google Scholar]
- 77. EMA . Zoely. https://www.ema.europa.eu/en/medicines/human/EPAR/zoely. Accessed December 5, 2020.
- 78. EU clinical trials register. https://www.clinicaltrialsregister.eu/ctr-search/trial/2011-004584-67/results. Accessed December 6, 2020.
- 79. Fekete S, Hiemke C, Gerlach M. Dose‐Related Concentrations of Neuroactive/Psychoactive Drugs Expected in Blood of Children and Adolescents. Ther Drug Monit. 2020;42(2):315‐324. [DOI] [PubMed] [Google Scholar]
- 80. Momper JD, Mulugeta Y, Green DJ, et al. Adolescent dosing and labeling since the Food and Drug Administration Amendments Act of 2007. JAMA Pediatr. 2013;167(10):926‐932. [DOI] [PubMed] [Google Scholar]
- 81. Noel GJ, Nelson RM, Bucci‐Rechtweg C, et al. Inclusion of Adolescents in Adult Clinical Trials: Report of the Institute for Advanced Clinical Trials for Children's Pediatric Innovation Research Forum. Ther Innov Regul Sci. 2021;55(4):773‐778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Anderson BJ, Holford NH. Tips and traps analyzing pediatric PK data. Paediatr Anaesth. 2011;21(3):222‐237. [DOI] [PubMed] [Google Scholar]
- 83. Cook SF, King AD, van den Anker JN, Wilkins DG. Simultaneous quantification of acetaminophen and five acetaminophen metabolites in human plasma and urine by high‐performance liquid chromatography‐electrospray ionization‐tandem mass spectrometry: Method validation and application to a neonatal pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci. 2015;1007:30‐42. [DOI] [PubMed] [Google Scholar]
- 84. Knosgaard KR, Foster DJ, Kreilgaard M, Sverrisdottir E, Upton RN, van den Anker JN. Pharmacokinetic models of morphine and its metabolites in neonates: Systematic comparisons of models from the literature, and development of a new meta‐model. Eur J Pharm Sci. 2016;92:117‐130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. van Groen BD, van Duijn E, de Vries A, et al. Proof of Concept: First Pediatric [(14) C]microtracer Study to Create Metabolite Profiles of Midazolam. Clin Pharmacol Ther. 2020;108(5):1003‐1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Aleksa K, Matsell D, Krausz K, Gelboin H, Ito S, Koren G. Cytochrome P450 3A and 2B6 in the developing kidney: implications for ifosfamide nephrotoxicity. Pediatr Nephrol. 2005;20(7):872‐885. [DOI] [PubMed] [Google Scholar]
- 87. Ghodke‐Puranik Y, Thorn CF, Lamba JK, et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics. 2013;23(4):236‐241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Altamimi MI, Choonara I, Sammons H. Inter‐individual variation in morphine clearance in children. Eur J Clin Pharmacol. 2015;71(6):649‐655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Krekels EH, van Ham S, Allegaert K, et al. Developmental changes rather than repeated administration drive paracetamol glucuronidation in neonates and infants. Eur J Clin Pharmacol. 2015;71(9):1075‐1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. van der Marel CD, Anderson BJ, van Lingen RA, et al. Paracetamol and metabolite pharmacokinetics in infants. Eur J Clin Pharmacol. 2003;59(3):243‐251. [DOI] [PubMed] [Google Scholar]
- 91. Cook SF, Stockmann C, Samiee‐Zafarghandy S, et al. Neonatal Maturation of Paracetamol (Acetaminophen) Glucuronidation, Sulfation, and Oxidation Based on a Parent‐Metabolite Population Pharmacokinetic Model. Clin Pharmacokinet. 2016;55(11):1395‐1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Flint RB, Roofthooft DW, van Rongen A, et al. Exposure to acetaminophen and all its metabolites upon 10, 15, and 20 mg/kg intravenous acetaminophen in very‐preterm infants. Pediatr Res. 2017;82(4):678‐684. [DOI] [PubMed] [Google Scholar]
- 93. Linakis MW, Cook SF, Kumar SS, et al. Polymorphic Expression of UGT1A9 is Associated with Variable Acetaminophen Glucuronidation in Neonates: A Population Pharmacokinetic and Pharmacogenetic Study. Clin Pharmacokinet. 2018;57(10):1325‐1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Zempsky WT, Bhagat PK, Siddiqui K. Practical Challenges‐Use of Paracetamol in Children and Youth Who are Overweight or Obese: A Narrative Review. Paediatr Drugs. 2020;22(5):525‐534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Giacoia GP, Taylor‐Zapata P, Zajicek A. Drug studies in newborns: a therapeutic imperative. Clin Perinatol. 2012;39(1):11‐23. [DOI] [PubMed] [Google Scholar]
- 96. Ethical considerations for clinical trials on medicinal products conducted with minors. Recommendations of the expert group on clinical tirals for the implementation of Regulation (EU) No 536/2014 on clinical trials on medicinal products for human use. https://ec.europa.eu/health/sites/health/files/files/eudralex/vol-10/2017_09_18_ethical_consid_ct_with_minors.pdf. Published 2017. Accessed April 5, 2021.
- 97. Manolis E, Osman TE, Herold R, et al. Role of modeling and simulation in pediatric investigation plans. Paediatr Anaesth. 2011;21(3):214‐221. [DOI] [PubMed] [Google Scholar]
- 98. Stockmann C, Barrett JS, Roberts JK, Sherwin C. 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(11):630‐640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Leroux S, Turner MA, Guellec CB, et al. Pharmacokinetic Studies in Neonates: The Utility of an Opportunistic Sampling Design. Clin Pharmacokinet. 2015;54(12):1273‐1285. [DOI] [PubMed] [Google Scholar]
- 100. Standing JF, Anderson BJ, Holford NH, Lutsar I, Metsvaht T. Comment on Pharmacokinetic Studies in Neonates: The Utility of an Opportunistic Sampling Design. Clin Pharmacokinet. 2015;54(12):1287‐1288. [DOI] [PubMed] [Google Scholar]
- 101. Cohen‐Wolkowiez M, Watt KM, Zhou C, et al. Developmental pharmacokinetics of piperacillin and tazobactam using plasma and dried blood spots from infants. Antimicrob Agents Chemother. 2014;58(5):2856‐2865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. 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(7):1908‐1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Standing JF, Ongas MO, Ogwang C, et al. Dosing of Ceftriaxone and Metronidazole for Children With Severe Acute Malnutrition. Clin Pharmacol Ther. 2018;104(6):1165‐1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Ollivier C, Thomson A, Manolis E, et al. Commentary on the EMA Reflection Paper on the use of extrapolation in the development of medicines for paediatrics. Br J Clin Pharmacol. 2019;85(4):659‐668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Chen J, Hsieh Y. Stabilizing drug molecules in biological samples. Ther Drug Monit. 2005;27(5):617‐624. [DOI] [PubMed] [Google Scholar]
- 106. 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;78‐79:34‐38. [DOI] [PubMed] [Google Scholar]
- 107. Duarah S, Sharma M, Wen J. Recent advances in microneedle‐based drug delivery: Special emphasis on its use in paediatric population. Eur J Pharm Biopharm. 2019;136:48‐69. [DOI] [PubMed] [Google Scholar]
- 108. Samant PP, Niedzwiecki MM, Raviele N, et al. Sampling interstitial fluid from human skin using a microneedle patch. Sci Transl Med. 2020;12(571):eaaw0285. 10.1126/scitranslmed.aaw0285 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Norman JJ, Brown MR, Raviele NA, Prausnitz MR, Felner EI. Faster pharmacokinetics and increased patient acceptance of intradermal insulin delivery using a single hollow microneedle in children and adolescents with type 1 diabetes. Pediatr Diabetes. 2013;14(6):459‐465. [DOI] [PubMed] [Google Scholar]
- 110. Food and Drug Administration. General clinical pharmacology considerations for pediatric studies for drugs and biological products. Guidance for industry. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/general-clinical-pharmacology-considerations-pediatric-studies-drugs-and-biological-products. Published 2014. Accessed December 6th 2020.
- 111. Patsalos PN, Berry DJ. Therapeutic drug monitoring of antiepileptic drugs by use of saliva. Ther Drug Monit. 2013;35(1):4‐29. [DOI] [PubMed] [Google Scholar]
- 112. Soldin SJ, Rakhmanina NY, Spiegel HM, Sever JL. Therapeutic drug monitoring for patients with HIV infection: Children's National Medical Center, Washington DC experience. Ther Drug Monit. 2004;26(2):107‐109. [DOI] [PubMed] [Google Scholar]
- 113. Patteet L, Cappelle D, Maudens KE, Crunelle CL, Sabbe B, Neels H. Advances in detection of antipsychotics in biological matrices. Clin Chim Acta. 2015;441:11‐22. [DOI] [PubMed] [Google Scholar]
- 114. Kiang TK, Ensom MH. A Qualitative Review on the Pharmacokinetics of Antibiotics in Saliva: Implications on Clinical Pharmacokinetic Monitoring in Humans. Clin Pharmacokinet. 2016;55(3):313‐358. [DOI] [PubMed] [Google Scholar]
- 115. Bosgra S, Vlaming ML, Vaes WH. To Apply Microdosing or Not? Recommendations to Single Out Compounds with Non‐Linear Pharmacokinetics. Clin Pharmacokinet. 2016;55(1):1‐15. [DOI] [PubMed] [Google Scholar]
- 116. Mooij MG, van Duijn E, Knibbe CA, et al. Pediatric microdose study of [(14)C]paracetamol to study drug metabolism using accelerated mass spectrometry: proof of concept. Clin Pharmacokinet. 2014;53(11):1045‐1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Garner CR, Park KB, French NS, et al. Observational infant exploratory [(14)C]‐paracetamol pharmacokinetic microdose/therapeutic dose study with accelerator mass spectrometry bioanalysis. Br J Clin Pharmacol. 2015;80(1):157‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Gordi T, Baillie R, le Vuong T, et al. Pharmacokinetic analysis of 14C‐ursodiol in newborn infants using accelerator mass spectrometry. J Clin Pharmacol. 2014;54(9):1031‐1037. [DOI] [PubMed] [Google Scholar]
- 119. Food and Drug Administration. General clinical pharmacology considerations for neonatal studies for drugs and biological products guidance for industry. Published 2019. Accessed January 15, 2020.
- 120. Kearns GL, Artman M. Functional Biomarkers: an Approach to Bridge Pharmacokinetics and Pharmacodynamics in Pediatric Clinical Trials. Curr Pharm Des. 2015;21(39):5636‐5642. [DOI] [PubMed] [Google Scholar]
- 121. Hoffman EP, Schwartz BD, Mengle‐Gaw LJ, et al. Vamorolone trial in Duchenne muscular dystrophy shows dose‐related improvement of muscle function. Neurology. 2019;93(13):e1312‐e1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Conklin LS, Damsker JM, Hoffman EP, et al. Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first‐in‐class dissociative steroidal anti‐inflammatory drug. Pharmacol Res. 2018;136:140‐150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Conklin LS, Hoffman EP, van den Anker J. Developmental Pharmacodynamics and Modeling in Pediatric Drug Development. J Clin Pharmacol. 2019;59(Suppl 1):S87‐S94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Wirth B, Brichta L, Schrank B, et al. Mildly affected patients with spinal muscular atrophy are partially protected by an increased SMN2 copy number. Hum Genet. 2006;119(4):422‐428. [DOI] [PubMed] [Google Scholar]
- 125. Berry‐Kravis EM, Lindemann L, Jonch AE, et al. Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov. 2018;17(4):280‐299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. van der Ploeg AT, Reuser AJ. Pompe's disease. Lancet. 2008;372(9646):1342‐1353. [DOI] [PubMed] [Google Scholar]
- 127. Ricci F, Vacchetti M, Brusa C, et al. New pharmacotherapies for genetic neuromuscular disorders: opportunities and challenges. Expert Rev Clin Pharmacol. 2019;12(8):757‐770. [DOI] [PubMed] [Google Scholar]
- 128. Mulla H. Understanding developmental pharmacodynamics: importance for drug development and clinical practice. Paediatr Drugs. 2010;12(4):223‐233. [DOI] [PubMed] [Google Scholar]
- 129. Rakhade SN, Jensen FE. Epileptogenesis in the immature brain: emerging mechanisms. Nat Rev Neurol. 2009;5(7):380‐391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Thorsson AV, Hintz RL. Insulin receptors in the newborn. Increase in receptor affinity and number. N Engl J Med. 1977;297(17):908‐912. [DOI] [PubMed] [Google Scholar]
- 131. Leroux S, van den Anker JN, Smits A, Pfister M, Allegaert K. Maturational changes in vancomycin protein binding affect vancomycin dosing in neonates. Br J Clin Pharmacol. 2019;85(5):865‐867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Bains I, Antia R, Callard R, Yates AJ. Quantifying the development of the peripheral naive CD4+ T‐cell pool in humans. Blood. 2009;113(22):5480‐5487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Andrew M, Paes B, Johnston M. Development of the hemostatic system in the neonate and young infant. Am J Pediatr Hematol Oncol. 1990;12(1):95‐104. [DOI] [PubMed] [Google Scholar]
- 134. Marshall JD, Kearns GL. Developmental pharmacodynamics of cyclosporine. Clin Pharmacol Ther. 1999;66(1):66‐75. [DOI] [PubMed] [Google Scholar]
- 135. Coulthard MG. Single blood pressure chart for children up to 13 years to improve the recognition of hypertension based on existing normative data. Arch Dis Child. 2020;105(8):778‐783. [DOI] [PubMed] [Google Scholar]
- 136. Faraone SV, Asherson P, Banaschewski T, et al. Attention‐deficit/hyperactivity disorder. Nat Rev Dis Primers. 2015;1(1):15020. 10.1038/nrdp.2015.20 [DOI] [PubMed] [Google Scholar]
- 137. Liu XI, Schuette P, Burckart GJ, et al. A Comparison of Pediatric and Adult Safety Studies for Antipsychotic and Antidepressant Drugs Submitted to the United States Food and Drug Administration. J Pediatr. 2019;208:236‐242. e233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Nelson SB, Valakh V. Excitatory/Inhibitory Balance and Circuit Homeostasis in Autism Spectrum Disorders. Neuron. 2015;87(4):684‐698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Lemonnier E, Villeneuve N, Sonie S, et al. Effects of bumetanide on neurobehavioral function in children and adolescents with autism spectrum disorders. Transl Psychiatry. 2017;7(5):e1124. 10.1038/tp.2017.101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Garbade SF, Zielonka M, Komatsuzaki S, et al. Quantitative retrospective natural history modeling for orphan drug development. J Inherit Metab Dis. 2021;44:99‐109. 10.1002/jimd.12304 [DOI] [PubMed] [Google Scholar]
- 141. Ristl R, Frommlet F, Koch A, Posch M. Fallback tests for co‐primary endpoints. Stat Med. 2016;35(16):2669‐2686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Gaasterland CM, Jansen‐van der Weide MC, Weinreich SS, van der Lee JH. A systematic review to investigate the measurement properties of goal attainment scaling, towards use in drug trials. BMC Med Res Methodol. 2016;16:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Lagler FB, Hirschfeld S, Kindblom JM. Challenges in clinical trials for children and young people. Arch Dis Child. 2020;106(4):321‐325. 10.1136/archdischild-2019-318676 [DOI] [PubMed] [Google Scholar]
- 144. Spineli LM, Jenz E, Grosshennig A, Koch A. Critical appraisal of arguments for the delayed‐start design proposed as alternative to the parallel‐group randomized clinical trial design in the field of rare disease. Orphanet J Rare Dis. 2017;12(1):140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. European Medicines Agency. Guideline on registry‐based studies (Draft). 24 September 2020. (EMA/502388/2020). https://www.ema.europa.eu/en/guideline-registry-based-studies. Accessed December 13, 2020.
- 146. Study HIVPPMC . Predictive value of absolute CD4 cell count for disease progression in untreated HIV‐1‐infected children. AIDS. 2006;20(9):1289‐1294. [DOI] [PubMed] [Google Scholar]
- 147. Wade AM, Ades AE. Age‐related reference ranges: significance tests for models and confidence intervals for centiles. Stat Med. 1994;13(22):2359‐2367. [DOI] [PubMed] [Google Scholar]
- 148. Bains I, Thiebaut R, Yates AJ, Callard R. Quantifying thymic export: combining models of naive T cell proliferation and TCR excision circle dynamics gives an explicit measure of thymic output. J Immunol. 2009;183(7):4329‐4336. [DOI] [PubMed] [Google Scholar]
- 149. Hoare RL, Veys P, Klein N, Callard R, Standing JF. Predicting CD4 T‐Cell Reconstitution Following Pediatric Hematopoietic Stem Cell Transplantation. Clin Pharmacol Ther. 2017;102(2):349‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Smith RF, McDonald CG, Bergstrom HC, Ehlinger DG, Brielmaier JM. Adolescent nicotine induces persisting changes in development of neural connectivity. Neurosci Biobehav Rev. 2015;55:432‐443. [DOI] [PubMed] [Google Scholar]
- 151. D. OR . Brain maturation ‐ it covers three decades: considerations of the development of ADHD. In Convergence. Interdisciplinary Communcations 2004/2005 ( Ostreng, W. ed.) ‐‐. 14–151 (Centre for Advanced Study, Norwegian Academy of Science and Letters, Oslo, Norway. https://duepublico2.uni-due.de/servlets/MCRFileNodeServlet/duepublico_derivate_00027779/CAS_Oslo_2005_Brain_Maturation_ADHD.pdf. Published 2005. Accessed April 12, 2021. [Google Scholar]
- 152. Shoji E, Sakurai H, Nishino T, et al. Early pathogenesis of Duchenne muscular dystrophy modelled in patient‐derived human induced pluripotent stem cells. Sci Rep. 2015;5(1):12831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Yoshida M, Kitaoka S, Egawa N, et al. Modeling the early phenotype at the neuromuscular junction of spinal muscular atrophy using patient‐derived iPSCs. Stem Cell Reports. 2015;4(4):561‐568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Veissi S, Smeets B, van den Heuvel LP, Schreuder MF, Jansen J. Nephrotic syndrome in a dish: recent developments in modeling in vitro. Pediatr Nephrol. 2020;35(8):1363‐1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Rooney KM, Woolf AS, Kimber SJ. Towards Modelling Genetic Kidney Diseases with Human Pluripotent Stem Cells. Nephron. 2021;1‐12. [DOI] [PubMed] [Google Scholar]
- 156. Vonk AM, van Mourik P, Ramalho AS, et al. Protocol for Application, Standardization and Validation of the Forskolin‐Induced Swelling Assay in Cystic Fibrosis Human Colon Organoids. STAR Protoc. 2020;1(1):100019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Prior N, Inacio P, Huch M. Liver organoids: from basic research to therapeutic applications. Gut. 2019;68(12):2228‐2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Chen S, Li P, Wang Y, et al. Rotavirus Infection and Cytopathogenesis in Human Biliary Organoids Potentially Recapitulate Biliary Atresia Development. MBio. 2020;11(4):e01968‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Van Peer E, Downes N, Casteleyn C, Van Ginneken C, Weeren A, Van Cruchten S. Organ data from the developing Gottingen minipig: first steps towards a juvenile PBPK model. J Pharmacokinet Pharmacodyn. 2016;43(2):179‐190. [DOI] [PubMed] [Google Scholar]
- 160. Smits A, Annaert P, Van Cruchten S, Allegaert K. A Physiology‐Based Pharmacokinetic Framework to Support Drug Development and Dose Precision During Therapeutic Hypothermia in Neonates. Front Pharmacol. 2020;11:587. 10.3389/fphar.2020.00587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Salaets T, Tack B, Jimenez J, et al. Simvastatin attenuates lung functional and vascular effects of hyperoxia in preterm rabbits. Pediatr Res. 2020;87(7):1193‐1200. [DOI] [PubMed] [Google Scholar]
- 162. Colon‐Rodriguez A, Uribe‐Salazar JM, Weyenberg KB, et al. Assessment of Autism Zebrafish Mutant Models Using a High‐Throughput Larval Phenotyping Platform. Front Cell Dev Biol. 2020;8:586296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Nylen H, Sergel S, Forsberg L, et al. Cytochrome P450 3A activity in mothers and their neonates as determined by plasma 4beta‐hydroxycholesterol. Eur J Clin Pharmacol. 2011;67(7):715‐722. [DOI] [PubMed] [Google Scholar]
- 164. Nakamura J, Yakata M. Age‐ and sex‐related differences in urinary excretion of 6 beta‐hydroxycortisol in humans. Clin Chim Acta. 1985;152(1–2):193‐197. [DOI] [PubMed] [Google Scholar]
- 165. Mao J, Martin I, McLeod J, et al. Perspective: 4beta‐hydroxycholesterol as an emerging endogenous biomarker of hepatic CYP3A. Drug Metab Rev. 2017;49(1):18‐34. [DOI] [PubMed] [Google Scholar]
- 166. Ayuso M, Buyssens L, Stroe M, et al. The Neonatal and Juvenile Pig in Pediatric Drug Discovery and Development. Pharmaceutics. 2020;13(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Bueters R, Bael A, Gasthuys E, Chen C, Schreuder MF, Frazier KS. Ontogeny and Cross‐species Comparison of Pathways Involved in Drug Absorption, Distribution, Metabolism, and Excretion in Neonates (Review): Kidney. Drug Metab Dispos. 2020;48(5):353‐367. [DOI] [PubMed] [Google Scholar]
- 168. Cheung PY, Gill RS, Bigam DL. A swine model of neonatal asphyxia. J Vis Exp. 2011;56. 10.3791/3166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Burrin D, Sangild PT, Stoll B, et al. Translational Advances in Pediatric Nutrition and Gastroenterology: New Insights from Pig Models. Annu Rev Anim Biosci. 2020;8:321‐354. [DOI] [PubMed] [Google Scholar]
- 170. Bauer R, Walter B, Brust P, Fuchtner F, Zwiener U. Impact of asymmetric intrauterine growth restriction on organ function in newborn piglets. Eur J Obstet Gynecol Reprod Biol. 2003;110(Suppl 1):S40‐S49. [DOI] [PubMed] [Google Scholar]
- 171. Barouxis D, Chalkias A, Syggelou A, Iacovidou N, Xanthos T. Research in human resuscitation: what we learn from animals. J Matern Fetal Neonatal Med. 2012;25(Suppl 5):44‐46. [DOI] [PubMed] [Google Scholar]
- 172. Agency EM . EMA Decision of 15 May 2020 on the acceptance of a modificaion of an agreed paediatric investigation plan for 2‐iminobiotin (EMEA‐001070‐PIP01–10‐M02). Accessed.
- 173. Wik EH, Martinez‐Silvan D, Farooq A, Cardinale M, Johnson A, Bahr R. Skeletal maturation and growth rates are related to bone and growth plate injuries in adolescent athletics. Scand J Med Sci Sports. 2020;30(5):894‐903. [DOI] [PubMed] [Google Scholar]
- 174. Wang W, Yeung KWK. Bone grafts and biomaterials substitutes for bone defect repair: A review. Bioact Mater. 2017;2(4):224‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Kohli N, Ho S, Brown SJ, et al. Bone remodelling in vitro: Where are we headed?: ‐A review on the current understanding of physiological bone remodelling and inflammation and the strategies for testing biomaterials in vitro. Bone. 2018;110:38‐46. [DOI] [PubMed] [Google Scholar]
- 176. Srinivasaiah S, Musumeci G, Mohan T, et al. A 300 mum Organotypic Bone Slice Culture Model for Temporal Investigation of Endochondral Osteogenesis. Tissue Eng Part C Methods. 2019;25(4):197‐212. [DOI] [PubMed] [Google Scholar]
- 177. Langhof H, Chin WWL, Wieschowski S, Federico C, Kimmelman J, Strech D. Preclinical efficacy in therapeutic area guidelines from the U.S. Food and Drug Administration and the European Medicines Agency: a cross‐sectional study. Br J Pharmacol. 2018;175(22):4229‐4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Willmann R, Luca A, Nagaraju K, Ruegg MA. Best Practices and Standard Protocols as a Tool to Enhance Translation for Neuromuscular Disorders. J Neuromuscul Dis. 2015;2(2):113‐117. [DOI] [PubMed] [Google Scholar]
- 179. Kilkenny C, Browne WJ, Cuthi I, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. Vet Clin Pathol. 2012;41(1):27‐31. [DOI] [PubMed] [Google Scholar]
- 180. Hair K, Macleod MR, Sena ES, Collaboration II . A randomised controlled trial of an Intervention to Improve Compliance with the ARRIVE guidelines (IICARus). Res Integr Peer Rev. 2019;4:12. 10.1186/s41073-019-0069-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Willmann R, Lee J, Turner C, et al. Improving translatability of preclinical studies for neuromuscular disorders: lessons from the TREAT‐NMD Advisory Committee for Therapeutics (TACT). Dis Model Mech. 2020;13(2):dmm042903. 10.1242/dmm.042903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Tomalik‐Scharte D, Lutjohann D, Doroshyenko O, Frank D, Jetter A, Fuhr U. Plasma 4beta‐hydroxycholesterol: an endogenous CYP3A metric? Clin Pharmacol Ther. 2009;86(2):147‐153. [DOI] [PubMed] [Google Scholar]
- 183. Wide K, Larsson H, Bertilsson L, Diczfalusy U. Time course of the increase in 4beta‐hydroxycholesterol concentration during carbamazepine treatment of paediatric patients with epilepsy. Br J Clin Pharmacol. 2008;65(5):708‐715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Takaki Y, Mizuochi T, Takei H, et al. Urinary and serum oxysterols in children: developmental pattern and potential biomarker for pediatric liver disease. Sci Rep. 2020;10(1):6752. 10.1038/s41598-020-63758-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Vauzelle‐Kervroedan F, Rey E, Pariente‐Khayat A, et al. Non invasive in vivo study of the maturation of CYP IIIA in neonates and infants. Eur J Clin Pharmacol. 1996;51(1):69‐72. [DOI] [PubMed] [Google Scholar]
- 186. Nakamura H, Hirai M, Ohmori S, et al. Changes in urinary 6beta‐hydroxycortisol/cortisol ratio after birth in human neonates. Eur J Clin Pharmacol. 1998;53(5):343‐346. [DOI] [PubMed] [Google Scholar]
- 187. Engelhart DC, Granados JC, Shi D, et al. Systems Biology Analysis Reveals Eight SLC22 Transporter Subgroups, Including OATs, OCTs, and OCTNs. Int J Mol Sci. 2020;21(5):1791. 10.3390/ijms21051791 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Zhang H, Basit A, Wolford C, et al. Normalized Testosterone Glucuronide as a Potential Urinary Biomarker for Highly Variable UGT2B17 in Children 7‐18 Years. Clin Pharmacol Ther. 2020;107(5):1149‐1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. European Medicines Agency . Reflection paper on the use of extrapolation in the development of medicines for paediatrics. https://www.ema.europa.eu/en/documents/scientific-guideline/adopted-reflection-paper-use-extrapolation-development-medicines-paediatrics-revision-1_en.pdf. Published 2018. Accessed December 8, 2020.
- 190. Manolis E, Pons G. Proposals for model‐based paediatric medicinal development within the current European Union regulatory framework. Br J Clin Pharmacol. 2009;68(4):493‐501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Bi Y, Liu J, Li L, et al. Role of Model‐Informed Drug Development in Pediatric Drug Development, Regulatory Evaluation, and Labeling. J Clin Pharmacol. 2019;59(Suppl 1):S104‐S111. [DOI] [PubMed] [Google Scholar]
- 192. Van den Anker JN, McCune S, Annaert P, et al. Approaches to Dose Finding in Neonates, Illustrating the Variability between Neonatal Drug Development Programs. Pharmaceutics. 2020;12(7):685. 10.3390/pharmaceutics12070685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Schrier L, Hadjipanayis A, Stiris T, et al. Off‐label use of medicines in neonates, infants, children, and adolescents: a joint policy statement by the European Academy of Paediatrics and the European society for Developmental Perinatal and Pediatric Pharmacology. Eur J Pediatr. 2020;179(5):839‐847. [DOI] [PubMed] [Google Scholar]
- 194. Ollivier C, Mulugeta YL, Ruggieri L, Saint‐Raymond A, Yao L. Paediatric extrapolation: A necessary paradigm shift. Br J Clin Pharmacol. 2019;85(4):675‐679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Waubant E, Banwell B, Wassmer E, et al. Clinical trials of disease‐modifying agents in pediatric MS: Opportunities, challenges, and recommendations from the IPMSSG. Neurology. 2019;92(22):e2538‐e2549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. US Food and Drug administration . Enhancing the Diversity of clinical trial populations ‐ Eligibility criteria, enrollment practices, and trial designs guidance for industry. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enhancing-diversity-clinical-trial-populations-eligibility-criteria-enrollment-practices-and-trial. Published 2020. Accessed December 9, 2020.
- 197. Flint RB, Ter Heine R, Spaans E, et al. Simulation‐based suggestions to improve ibuprofen dosing for patent ductus arteriosus in preterm newborns. Eur J Clin Pharmacol. 2018;74(12):1585‐1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Zhang Q, Xu B, Lv J, Wang Z, Du J. Safety and effect of sildenafil on treating paediatric pulmonary arterial hypertension: a meta‐analysis on the randomised controlled trials. Cardiol Young. 2020;30(12):1882‐1889. [DOI] [PubMed] [Google Scholar]
- 199. Lochmuller H, Evans D, Farwell W, et al. Position Statement: Sharing of Clinical Research Data in Spinal Muscular Atrophy to Accelerate Research and Improve Outcomes for Patients. J Neuromuscul Dis. 2018;5(2):131‐133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. De Boeck K, Lee T, Amaral M, et al. Cystic fibrosis drug trial design in the era of CFTR modulators associated with substantial clinical benefit: stakeholders' consensus view. J Cyst Fibros. 2020;19(5):688‐695. [DOI] [PubMed] [Google Scholar]
- 201. Matza LS, Patrick DL, Riley AW, et al. Pediatric patient‐reported outcome instruments for research to support medical product labeling: report of the ISPOR PRO good research practices for the assessment of children and adolescents task force. Value Health. 2013;16(4):461‐479. [DOI] [PubMed] [Google Scholar]
- 202. Gaillard S, Malik S, Preston J, et al. Involving children and young people in clinical research through the forum of a European Young Persons' Advisory Group: needs and challenges. Fundam Clin Pharmacol. 2018;32(4):357‐362. [DOI] [PubMed] [Google Scholar]
- 203. Davis JM, Baer GR, McCune S, et al. Standardizing Safety Assessment and Reporting for Neonatal Clinical Trials. J Pediatr. 2020;219:243‐249. e241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. World Health Organization . The use of the WHO‐UMC system for standardised case causality assessment. https://www.who.int/medicines/areas/quality_safety/safety_efficacy/WHOcausality_assessment.pdf. Accessed December 9, 2020.
- 205. Roberts EK, Hawcutt DB, Turner MA. Prospective identification and causality evaluation of suspected adverse drug reactions in neonates. Br J Clin Pharmacol. 2020;87(3):1541‐1546. 10.1111/bcp.14485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Salaets T, Turner MA, Short M, et al. Development of a neonatal adverse event severity scale through a Delphi consensus approach. Arch Dis Child. 2019;104(12):1167‐1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events. http://ucdmc.ucdavis.edu/clinicaltrials/StudyTools/Documents/DAIDS_AE_GradingTable_FinalDec2004.pdf. Accessed December 15, 2020.
- 208. Administration UFaD . Division of Microbiology and Infectious Diseases (DMID) Pediatric Toxicity Tables. http://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/Vaccines/ucm091977.pdf. Accessed December 15, 2020.
- 209. Services UDoHaH . CTCAE. Common Terminology Criteria for Adverse Events v5. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf. Published 2017. Accessed December 15, 2020.
- 210. Macfarlane PW. The Influence of Age and Sex on the Electrocardiogram. Adv Exp Med Biol. 2018;1065:93‐106. [DOI] [PubMed] [Google Scholar]
- 211. van Donge T, Staub E, Atkinson A, et al. Age appropriate reference intervals for eight kidney function and injury markers in infants, children and adolescents. Clin Chem Lab Med. 2020:373‐382. 10.1515/cclm-2020-0781 [DOI] [PubMed] [Google Scholar]
- 212. Allegaert K, Smits A, van Donge T, et al. Renal Precision Medicine in Neonates and Acute Kidney Injury: How to Convert a Cloud of Creatinine Observations to Support Clinical Decisions. Front Pediatr. 2020;8:366. 10.3389/fped.2020.00366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Kelly LE, Sinha Y, Barker CIS, Standing JF, Offringa M. Useful pharmacodynamic endpoints in children: selection, measurement, and next steps. Pediatr Res. 2018;83(6):1095‐1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Buyssens L, De Clerck L, Schelstraete W, et al. Hepatic Cytochrome P450 Abundance and Activity in the Developing and Adult Göttingen Minipig: Pivotal Data for PBPK Modeling. Front Pharmacol. 2021;12(535):665644. 10.3389/fphar.2021.665644 [DOI] [PMC free article] [PubMed] [Google Scholar]
