ArticlePDF AvailableLiterature Review

Physiologically Based Pharmacokinetic (PBPK) Modeling in Children

Authors:
  • Aridhia Bioinformatics

Abstract

This review summarizes the present status of physiologically based pharmacokinetic (PBPK) modeling and simulation (M&S) and its application in support of pediatric drug research. We address the reasons that PBPK is suited to the current needs of pediatric drug development and pharmacotherapy in light of the evolution in pediatric PBPK methodologies and approaches, which were originally developed for the purpose of toxicologic evaluation. Also discussed is the current degree of confidence in using PBPK to support pediatric drug development and registration and the key factors essential for robust results and broader adoption of pediatric PBPK M&S.
state art
nature publishing group
40 VOLUME 92 NUMBER 1 | JULY 2012 | www.nature.com/cpt
Pediatric drug development and pharmacotherapy represent new
frontiers for clinical pharmacology. In the past decade, pediatric
drug development, which occurs in the scenarios summarized in
Tabl e 1, has received much attention and support through regu-
latory incentives and requirements in the United States and the
European Union, as well as from an increasing societal aware-
ness of the necessity of performing clinical research in pediatric
patients. Nevertheless, drug research involving pediatric indica-
tions and drug dosing optimization in pediatric clinical practice
remains challenging because of the very nature of conducting
research in pediatric patients, especially in infants and children,
who are considered a vulnerable population.
Although there are many opportunities for applying physiologi-
cally based pharmacokinetic (PBPK) modeling in pediatric clini-
cal pharmacology, there is a lingering concern and skepticism in
the pediatric community about the robustness and reliability of
PBPK-based prediction of drug exposure and ultimately about
ecacy and toxicity, in pediatric patients. is article not only
reviews some of the potential benets and utility of PBPK mod-
eling in pediatrics but also critically discusses current limitations
and deciencies that have so far dampened the condence in this
approach and impeded its more widespread application.
WHY IS PBPK MODELING SUITED TO THE CURRENT
NEEDS IN PEDIATRIC DRUG DEVELOPMENT AND
PHARMACOTHERAPY?
Clinical research in pediatric patients is hampered by inter-
related logistical as well as ethical constraints, for example,
the limited number, extent, and invasiveness of study-related
interventions that can be performed if they are not a part of a
routine therapeutic plan. us, clinical studies in pediatrics are
oen limited with respect to the number of enrolled patients and
treatment arms and in the availability of appropriate primary
and secondary outcome measures.
1
Given that it is inherently more dicult to conduct clinical
studies in infants and children than in adults, pediatric doses of
medicinal products have traditionally been scaled from adult
doses, using functions related to body weight, height, or age.
Such simple allometric approaches may be questionable when
complex absorption and disposition processes are encountered
and can fail to predict exposure accurately, particularly in the
very young.
2
When the adult dose is normalized by body weight
(i.e., mg/kg), under the assumption of a linear relationship
between weight and dose, it indicates that the dose required
will double with a twofold increase in the weight of a child.
When dose adjustment is based on age (i.e., preterm newborns,
term newborns, infants, toddlers, children, and adolescents),
the rapid changes in rates of organ maturation, blood ow, body
composition, and ontogeny of drug elimination and transport
mechanisms occurring in developing children within each age
group are not taken into account.
3
Scaling by body surface
area has been reported to lead to overdosing in neonates and
infants, because of either the inaccuracy of the formula or the
unpredictability of low drug metabolizing enzyme activity at
birth.
2
Moreover, the change in PK parameters across pediat-
ric age strata does not change proportionally with body surface
The last three authors contributed equally to this work.
1
Department of Pediatrics, Division of Clinical Pharmacology & Therapeutics, The Childrens Hospital of Philadelphia, Philadelphia, Pennsylvania, USA;
2
Scientific
Direction, Clinical Pharmacology and Clinical Trials Unit, IRCCS Giannina Gaslini Institute, The Children’s Hospital of Genoa, Genoa, Italy;
3
Department of Clinical
Pharmacy and Pharmacotherapy, Heinrich-Heine University, Düsseldorf, Germany;
4
Department of Pharmaceutical Sciences, College of Pharmacy, University of
Tennessee Health Science Center, Memphis, Tennessee, USA. Correspondence: O Della Casa Alberighi (ornelladellacasa@ospedale-gaslini.ge.it)
Received 18 January 2012; accepted 9 April 2012; advance online publication 6 June 2012. doi:10.1038/clpt.2012.64
Physiologically Based Pharmacokinetic (PBPK)
Modeling in Children
JS Barrett
1
, O Della Casa Alberighi
2
, S Läer
3
and B Meibohm
4
This review summarizes the present status of physiologically based pharmacokinetic (PBPK) modeling and simulation
(M&S) and its application in support of pediatric drug research. We address the reasons that PBPK is suited to the current
needs of pediatric drug development and pharmacotherapy in light of the evolution in pediatric PBPK methodologies
and approaches, which were originally developed for the purpose of toxicologic evaluation. Also discussed is the current
degree of confidence in using PBPK to support pediatric drug development and registration and the key factors essential
for robust results and broader adoption of pediatric PBPK M&S.
state artstate art
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 92 NUMBER 1 | JULY 2012 41
area because it is not a descriptor of metabolic function.
4
On
the other hand, no dierences in drug metabolism may exist
between adolescents and adults.
5
To overcome some of these limitations, the increased use of
pharmacokinetic (PK) and pharmacodynamic (PD) modeling
and simulation (M&S) techniques in pediatric drug develop-
ment and applied pharmacotherapy has been promoted by vari-
ous groups in academia,
6
industr y,
7
and regulatory agencies.
8–10
Two basic areas of M&S application have emerged in recent years
in pediatric clinical pharmacology research. One is an a priori
application of M&S techniques to optimize trial designs, select
dose level and dosing regimens to be studied, develop sampling
schemes, and select outcome measures. is application of M&S
is frequently based on adult data and uses various extrapola-
tion approaches that incorporate PK and PD data from preclini-
cal species, in vitro experiments, and pediatric data from pilot
studies or studies in older pediatric groups.
6,11,12
e second
emerging areas is an a posteriori application of M&S techniques
that has been applied even more frequently to analyze PK and
PK–PD data from pediatric studies, especially using population-
based approaches to derive maximum information content from
the limited data collected in pediatric studies and to establish
a consistent framework that allows the exploration of model-
based dosing recommendations.
13,14
PBPK and PBPK–PD M&S have recently gained increasing
popularity in drug development.
15,16
is has been driven by
major advances in the tools available for performing PBPK
analyses and by a substantial increase in the level of sophis-
tication and accuracy in the underlying physiologic data that
are now available.
16
PBPK and PBPK–PD modeling exemplify
“bottom-upmodeling in systems biology in that specic avail-
able observations, elements, and patterns are combined to form
larger subsystems, which then are linked at many levels to form
a complete multiscale model (see schema in Figure 1).
16
ese
subsystems integrate patient-specic parameters related to
anatomy, physiology, and pathophysiology with drug-specic
properties, such as physicochemical characteristics, metabolic
proles, and pharmacogenomic data. For pediatrics, PBPK mod-
eling also oers a unique opportunity to integrate known matu-
ration trajectories (functions that describe the time course of
organ development until adult physiological status is achieved)
for processes relevant to drug disposition in a consistent frame-
work to better predict drug behavior in various age groups.
17
Such an approach is direly needed—especially in neonates, who
are being delivered at an ever-increasing stage of prematurity
and for whom there is a particular scarcity of drug disposition
data that has been forcing pediatric practitioners to base phar-
macotherapy on empiricism rather than on a scientic ration-
ale.
6,18
Although these PBPK models integrate a wide variety of
biological factors to predict drug PK and thus provide a rational
support for pediatric dose nding, the challenge is to identify
and characterize the ontogeny of the relevant PK and PD proc-
esses that will enable the behavior of a drug to be predicted
across the wide age and physiological development spectrum
of neonates, infants, and children.
19
CONSTRUCTION OF PEDIATRIC PBPK MODELS
As with the traditional scaling approach based on adult data,
the PBPK model provides a computational engine for predict-
ing dose–exposure relationships in children but with a more
physiologically based model.
20
Table 2 provides a template
for the inputs and outputs needed for a typical PBPK model.
Schematically, a PBPK model is a multicompartment model
in which the compartments represent actual organs and other
physiological spaces (Figure 1). Mass balance equations for
each organ describe drug appearance in the organ from arte-
rial blood and its exit into venous blood. e PBPK model is
also constructed to incorporate relevant physiological, phar-
macogenetic, biochemical, and thermodynamic parameters in
a way that organizes much of the knowledge of the drug–body
system.
16
us, PBPK models are more comprehensive than
the empirical models used to analyze routine PK data because
they not only incorporate drug properties but also are built on
a system-specic structure that is independent of the drug.
16,21
e model parameters need to include physiological and drug-
specic parameters, in vitro predictions for distribution and
elimination, and, perhaps, results from in vivo studies in adult
animals.
22,23
e common approach in developing a pediatric
PBPK model is to modify a PBPK model that has been validated
with adult PK data and then to incorporate the dierences in
growth and maturation that can aect all relevant aspects of
drug disposition and PD.
Unique aspects of pediatric PBPK modeling
Human development is characterized by maturation and growth
that aect a child’s morphology, physiology, pathology, and psy-
chology. Maturation and growth oen seem to be a continuum,
but they consist of highly complex sequences of a variety of
concurrent maturation processes from all body compartments,
organs, and interconnected molecular networks that all follow
dierent temporal developmental trajectories. To build a physi-
ologically based model describing PK processes, all developmen-
tal changes aecting drug absorption, distribution, metabolism,
and elimination (ADME) need to be addressed (Tabl e 3). All
of these factors need to be integrated into and qualitatively and
quantitatively assessed in pediatric PBPK models.
Table 1 Scenarios for pediatric drug development
1. Drug has established efficacy and safety in adults, and indication/
disease is similar in adults and children
2. Drug has established efficacy and safety in adults, but indication/
disease has different features or mainly affects children
3. Drug is newly developed for indication/disease that only occurs in
children
4. Drug is newly developed for similar indication/disease in both children
and adults
Scenario 1 could be coupled with scenario 2. Taking the example of epilepsy as a
disease, it includes the following indications: partial-onset seizures and Lennox-
Gastaut syndrome, which are similar in adults and children (scenario 1), allowing
treatment effects to be extrapolated from adults to children, and the infantile
epilepsies, which are specific to children, in which it is not possible to extrapolate
treatment effects including adverse events and PK/PD from adults to children, but
there is a possible model-based extrapolation for PK (scenario 2).
PD, pharmacodynamic; PK, pharmacokinetic.
state artstate art
42 VOLUME 92 NUMBER 1 | JULY 2012 | www.nature.com/cpt
Several groups have developed PBPK models that integrate
maturation and growth processes.
11,12,20,25–27
ese research-
ers then used data sets from the literature or from their own
investigations to validate the performance of their models across
dierent age groups. Model evaluation was based on a compari-
son between predicted and observed drug clearances, volumes of
distribution, and half-lives. Elimination processes investigated
include cytochrome P450s, glucuronidation, sulfonation, and
biliary clearance. e spectrum of drugs investigated include
alfentanil, theophylline, morphine, levetiratectam, levooxacin,
gentamicin, midazolam, carbamazepine, paracetamol, levo-
oxacin, ropivacaine, lorazepam, and fentanyl.
e majority of these studies were based on data sets obtained
aer intravenous (i.v.) drug administration. In each of the PBPK
models, the predictions of drug clearance, distribution volume,
and elimination half-life were within the expected range and
adequately predicted the parameters for the dierent age groups.
Edginton and Willmann
28
also extended their physiologically
based model by including the different pathophysiological
changes that accompany liver cirrhosis. ey adjusted organ
blood ow, cardiac index, plasma binding, hematocrit, func-
tional liver volume, hepatic enzymatic activity, and the glomeru-
lar ltration rate according to the disease state and the age of
pediatric patients. e comparison of predicted and observed
plasma concentrations, as well as clearance values, aer i.v. alfen-
tanil, lidocaine, theophylline, and levetiracetam administration
revealed adequate predictions for the four drugs, thus demon-
strating that PBPK models might be useful in predicting PK of
i.v.
p.o.
Dose
Stomach
Small intestine
Large intestine
Pancreas
Spleen
Liver
Kidney
Testes
Heart
Brain
Fat
Bone
Skin
Muscle
Lung
HO
H
N
O
Portal vein
Gall bladder
V
e
n
o
u
s
b
l
o
o
d
A
r
t
e
r
i
a
l
b
l
o
o
d
Figure 1 Physiologically based pharmacokinetic (PBPK) modeling in a bridging schema using a typical whole-blood PBPK model, in which the tissues and
organs of the body are arranged anatomically and connected via the vascular system. The following extrapolations are considered in pediatrics: (i) The
physiochemical bridge should be based on in vitro experiments and data that need to be correlated with understanding of in vivo drug disposition and transport
across physiologic barriers. (ii) For the animal–human bridge, the need for and timing of juvenile animal studies should be justified, and the target pediatric
age and primary pharmacodynamics in organs/tissues with significant postnatal development (nervous, reproductive, pulmonary, immune, renal, and skeletal
systems; biotransformation enzymes) need to be considered. (iii) For the adult–pediatric bridge, size, ontogeny, and maturation should be accommodated;
some physiologic spaces are still difficult to extrapolate across all age groups (e.g., bone marrow, brain).
state artstate art
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 92 NUMBER 1 | JULY 2012 43
some drugs aer their i.v. administration to pediatric patients
with a variety of diseases. However, the ability of PBPK models
to reliably predict plasma concentrations and PK parameters
aer oral drug administration to patients in various pediatric
age groups is still in question. is is expected because Poulin
et al.
15
also showed in adults that currently available PBPK
models have a substantially lower eectiveness in simulating
adult plasma concentration–time proles aer oral than aer i.v.
administration. erefore, further research is necessary before
oral administration models can be relied on to predict oral PK
for children.
Role of animal studies in pediatric PBPK development
Current guidelines applied to pharmaceutical drug develop-
ment in pediatrics indicate a need for juvenile animal studies
only when previous animal and human safety data are judged
insucient.
29–31
Situations that would justify toxicity studies
in juvenile animals include, but are not limited to, ndings in
nonclinical studies that indicate target organ or systemic toxicity
relevant for developing systems, possible eects on growth and/
or development in the intended age group, or a pharmacological
eect of the test compound that would aect one or more devel-
oping organs. e need for proper selection of species and
appropriate times of testing is exemplied by a PBPK model
developed recently for the neuroaminidase inhibitor oseltamivir
and its active metabolite in infants and neonates with inuenza
to predict exposures aer i.v. dosing.
32
is model addressed
the suitability of using young and adult marmoset monkeys to
represent age-dependent changes in human PK, the sensitiv-
ity of the tested drug and metabolite concentrations in vivo to
age-dependent changes in metabolism and renal function, and
the suitability of data aer oral administration to represent i.v.
disposition. In a stepwise modeling strategy, simulated data were
veried by comparison with data obtained in a juvenile animal
study; prediction in neonates and infants was then made using
a PBPK model accounting for the age dependencies in humans,
which was informed by renements developed for adult humans
and juvenile animals. However, there are still only a few pub-
lished studies about juvenile animals.
33
Of concern is that, despite the growing knowledge of func-
tional dierences between juvenile and adult animals used in
toxicology studies, there are still areas in which there is a lack of
Table 2 Hierarchy of PBPK model inputs and outputs based on intended use in supporting pediatric research and development
Intended use Model inputs
a
Outputs
Candidate screening (CS) •  Drug properties: MW, lipophilicity, solubility, protein 
binding, pKa
•  In vitro metabolism data (V
max
, K
M
, etc.) and experimental
details
•  Pediatric physiology: organ weight, blood flow, CL 
ontogeny, fu ontogeny (usually from software tool
database)
•  Study population (healthy volunteers); clearance pathways 
(CL
R
and CL
H
)
•  Dosing (usually single dose but can vary to incorporate 
simple phase I designs)
•  DDI potential (magnitude and shift of toxicity profiles 
relative to a standard (e.g., single agent relative to
combination)
•  Dose–exposure relation; evaluation of profiles against 
target product profile expectations
FTIP dose finding •  CS inputs
•  Pediatric-specific demographics (age, body weight, height, 
etc.) of study population; clearance pathways
•  Dosing (various algorithms and rules”can be evaluated)
•  Dose–exposure relation; evaluation of profiles and PK 
metrics across age/developmental strata and relative to
adult exposures
•  Comparison of exposure from fixed vs. weight-adjusted 
dosing
Target organ exposures (TOEs) •  FTIP inputs 
•  Species (if comparing animal biodistribution to pediatric 
predictions)
•  Target organs identified (with data/measured 
concentrations), if available
•  Overlays of observed vs. predicted exposures for animal 
studies
•  In pediatrics, predicted exposures in target organs; 
correlation with toxicity or PD measures
Trial design evaluation •  FTIP inputs
•  Design features (e.g., parallel vs. crossover), sampling 
scheme, sample size/strata, population, etc.
•  Replication details
•  Probability of success for scenarios or trial designs to 
achieve clinical milestones or study objectives
Real-time PK safety •  FTIP inputs
•  Measured concentrations (sampling/observations) 
•  Response measures (SAEs, ADRs, PD, etc.)
•  Overlays of observed vs. predicted exposures with 
comparisons across age strata and relative to adult data
Targeted drug delivery •  TOE inputs
•  Delivery inputs (e.g., extravascular route, input rate, 
duration)
•  Cellular constituent targets
•  Imaging data (if relevant)
•  Overlays of observed vs. predicted exposures (or 
equivalent metrics) with comparisons across age strata
and relative to adult data
ADRs, adverse drug reactions; CL, clearance; CL
H
, hepatic clearance; CL
R
, renal clearance; DDI, drug–drug interaction; FTIP, first-time-in-pediatrics; fu, unbound fraction; K
M
,
Michaelis–Menten constant-CYP enzyme affinity; MW, molecular weight; PD, pharmacodynamic; PK, pharmacokinetic; pKa, ionization constant; SAEs, serious adverse events; 
V
max
, maximum enzyme activity.
a
Derived parameters include partition coefficients and permeabilities.
state artstate art
44 VOLUME 92 NUMBER 1 | JULY 2012 | www.nature.com/cpt
robust knowledge of cross-species functional and PK dierences
among juveniles. is means that extrapolation of any toxico-
logical study nding to an immature human may not be easy or
even relevant (e.g., in central nervous system development for
which no species provides a direct correlation to human neu-
rological development). So, at a fundamental level, the devel-
opment of PBPK models in relevant animal species or even in
xenogra models, provides an important means of assessing
the portability of organ-based exposures between these animal
models and patients.
34,35
is is needed to permit a more critical
assessment of why animal models do or do not predict what is
observed clinically.
36,37
An important consideration is the avail-
ability of actual biodistribution data to ne-tune PBPK models,
as the absence of such data forces reliance on predicted tissue
partition coecients, which may be less accurate. In addition,
PBPK assessment of nonsystemic biodistribution targets can
support targeted delivery strategies and oers a tool for assess-
ing druggability during preclinical drug development.
Integration with pediatric pharmacodynamics
An extension of the PBPK concept includes the additional pre-
diction of drug exposure–response relationships in PBPK–PD
models. PBPK–PD modeling, which is aimed at the prediction
of exposure–response relationships in humans, including their
inherent intra- and interindividual variability, oers a unique
opportunity to integrate age- and size-dependent changes in
prediction of drug eects. Exposure is expressed as the time
course of the drug concentration in plasma, whereas response
is expressed as the time course of the intensity of the drug
eect. PBPK–PD models contain specic expressions to quan-
titatively characterize processes on the causal path between
plasma concentration and eect, such as drug target–site dis-
tribution, drug target binding and activation, and transduction
mechanisms. Ultimately, PBPK–PD models can also character-
ize the interaction of drug eect with disease processes.
38
e
distinction between drug-specic and biological system-specic
parameters is crucial for accurately predicting drug eects in
children. Such a distinction needs to consider growth and matu-
ration, as well as the availability and use of child-specic disease
outcomes.
PBPK–PD models have been extended to model risk from
chemical exposure in juvenile animals and in children.
39
However, only a few full PBPK–PD models have been devel-
oped so far for therapeutic drugs,
40
although several groups
have coupled PBPK models with classical compartmental PD
models.
41,42
Modeling the relationship between drug biophase
concentration and response can be tenuous when an a priori
approach is used. In an oncologic example, it was assumed suc-
cessfully that the response in children is superimposable on
that of adults.
43
However, this may not always be the case, as
illustrated by a PBPK–PD study of sotalol in pediatric patients
with supraventricular tachycardia, in which it was found that
exposure–response relationships may dier from adults and
may be age-variant,
14
and by a study of etanercept in pediatric
patients with juvenile idiopathic arthritis.
44
Extension of pediat-
ric PBPK modeling to include a PD component (e.g., covariates
associated with drug–receptor interaction) remains an area of
much-needed future research.
Application of PBPK modeling in pediatric drug development
e initial foray into pediatric PBPK modeling began in the
mid-1980s with studies in which PBPK models were used to
assess the risk of exposure to toxic chemicals, with the ultimate
goal of predicting organ-specic toxicity. Environmental toxi-
cology models have been applied to assess the risks of inadvert-
ent exposure to chemical compounds in unborn and breastfed
infants
45
and in children
46,47
, as well as to environmental agents
such as caeine and theophylline in neonates
27
; such models
have incorporated an early-life-stage renal clearance model for
the risk assessment of chemicals cleared primarily by the kid-
neys.
48
Currently, chemical-specic PBPK models are increas-
ingly being used in risk assessment to account for inter- and
intraspecies dierences in PK and to replace default uncertainty
factors in the derivation of reference values.
48
Workow practices for common pediatric PBPK applications
are shown in Figure 2. e most common implementations
of PBPK modeling in the setting of pediatric drug develop-
ment include rst-time-in-pediatrics (FTIP) dose selection,
49
simulation-based trial design,
12
correlation with target organ
toxicities, real-time assessment of PK–safety relationships, and
assessment of nonsystemic biodistribution targets. PBPK models
can support risk assessment by investigating possible drug–drug
interactions in pediatric populations and the eect of impaired
organ function.
50
FTIP dose selection is a critical milestone and decision point
in pediatric drug development.
48,51
Historically, these estimates
have come from an assessment of the adult therapeutic window
Table 3 Developmental changes affecting drug
pharmacokinetics
Changes in drug absorption
5
due to
Changes in gastric emptying and intestinal transit time
pH changes in different intestinal segments
Changes in intestinal transporters and in enzymes causing first-pass
metabolism
Changes in drug distribution
5
due to
Changes in body fluid compartments (e.g., total body water is 78% in
neonates vs. 55% in adults, extracellular fluid space also is relatively
greater, intracellular fluid space is relatively smaller)
Relative percentage of body fat is lower in children than in adults
Protein binding of drugs is less in infants and children than in adults
Blood–brain barrier is more permeable in infants and children than in
adults
Changes in hepatic metabolism
24,25
Liver size is relatively greater in infants and children than in adults
26
Drug metabolizing enzymes undergo age-specific changes (e.g.,
glucuronidation and sulfation are immature in neonates)
17
Changes in renal excretion of drugs and drug metabolites
24,25
Glomerular filtration rate is less in infants and children than in adults
Renal tubular absorption and secretion are less in infants and children
than in adults
state artstate art
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 92 NUMBER 1 | JULY 2012 45
(if known) and the collective safety/toxicology experience with
a drug molecule and class. Given that timelines are oen com-
pressed and the current regulatory requirement of providing
a Pediatric Investigation Plan, comprehensive adult PK and/
or clinical data are not necessarily available when this decision
point is reached. is necessitates greater reliance on in silico
techniques and preclinical experience to guide FTIP dose selec-
tion. e historical approach in adults was to choose starting
doses as fractions of toxicity indexes
52
(e.g., no adverse eect
level, NOAEL), but more recent criteria are based on drug action
(e.g., minimal anticipated biologic eect level, MABEL).
53
In
any case, these approaches still implicitly rely on extrapolation
of PD relationships observed in animal models
54
or on clinical
experience in adult populations.
55
Currently, PBPK method-
ology and workow for pediatric FTIP prediction
49
is reliant
on drug properties (e.g., physiochemical data, protein binding,
clearance) and is scaled to pediatric organ sizes and ow rates,
even accommodating maturational and ontogeny eects as a
function of the changes in physiologic parameters (e.g., func-
tional enzyme expression). Still, a key element of appropriately
tuning” these models is the need to adjust experimental or pre-
dicted organ uptake on the basis of available actual organ/tissue
biodistribution data in adults and experimental animals,
34,56
preferably aer i.v. administration to avoid any confounding
with absorption-related processes. Likewise, other drug physi-
ochemical and physiologic and drug-specic PK/ADME proper-
ties that serve as model inputs (e.g., ionization constant, protein
binding, cardiac output) may represent additional factors that
greatly inuence projected exposures. As a result, sensitivity
analyses are essential to identify key dependencies and to appro-
priately assess PBPK simulation results.
Clinical trial design can be aided by PBPK-based simulations
that provide the ability to evaluate dose selection, sample size,
and sampling scheme and aid in selecting design constructs
with the greatest potential to yield meaningful results.
13,57,58
By
shiing the scaling from size and age to function (e.g., a con-
cept that relies on inferences from PK parameter distributions
in children) to predict drug exposure in dierent pediatric age
groups, pediatric PBPK models have been suggested as a method
of choice in neonates and infants to guide optimization of the
dosing schedule and sampling times.
19
When incorporated in a
trial design model with appropriate replication, the PBPK model
also provides a means to generate virtual patients that mimic the
intended target population, and it provides a probability-based
Candidate screening
Define physiochemical
inputs from QSPK
relationshipscalculated
based on availability of
preclinical experiments
and data
based on availability of
adult PK, toxicology and
margin of safety
based on availability of
adult PK, and preclinical
distribution data
based on availability of
adult PK, PD and
therapeutic window
Model refinement: Model refinement:
Model refinement: developmental and ontogeny factors reflecting target pediatric
population characteristics
“Optimizeparameters
(e.g., permeability/
partitioning) via animal
distribution: consider
regional effects of
transporters (e.g., brain)
Incorporate proposed
disease progression,
design constructs and
sampling schemes
Screen/rank compound
candidates based on
ability to achieve clinical
exposure targets
Compare exposure
metrics across age strata
for various dose
multiples relative to
adult exposures
(available or targeted)
Simulate exposure
metrics in target tissues
for intended pediatric
population: evaluate
against activity/tox goals
Simulate trial
outcomes for each
design schema and/
or scenario
Model refinement:
Model refinement: Model refinement:
Model refinement:
Define physiochemical inputs from in vitro experimentsmeasured/estimated
parameters or calculated parameters
First-time-in-pediatrics
dose finding
Target organ
exposures
Tr ial design
evaluation
Figure 2 Simplified workflows for common pediatric PBPK applications. PBPK, physiologically based pharmacokinetic; QSPK, quantum sufficit (adjusted
accordingly) PK.
state artstate art
46 VOLUME 92 NUMBER 1 | JULY 2012 | www.nature.com/cpt
assessment of study outcomes.
12
Recent advances in soware
development also permit easier and more convenient identi-
cation of “virtual twins”: simulated patients with demographic
characteristics similar to those of the target population. As the
underlying data for these tools replaces algorithm-based simula-
tions with actual pediatric patient populations for resampling,
this approach should become even more relevant.
Once PK in children can be described and predicted, assump-
tions about the exposure–response relationships, including the
anticipated therapeutic and potentially toxic eects, can further
rene relationships between expected clinical response(s) and
the administered dose. Depending on the clinical needs and
the nature of the eect, either a threshold model (to simulate
PK proles and to design a dosing regimen to achieve target
drug concentrations above the expected threshold of ecacy but
below an anticipated threshold for an adverse event)
59
or PK–PD
models (to provide guidance on the timing of the response, by
integrating PK and PD parameters to simulate response-time
proles for given dosing regimens) can be considered. PBPK–PD
models also can be used to explore the sensitivity of these target
levels to changes in the dierent model system parameters. For
example, a frequent question concerning the inuence of pedi-
atric formulation variables (such as dierent dissolution-rate salt
forms) on clinical biomarkers can be explored in silico through
appropriately parameterized PBPK–PD models.
60
As more studies have been completed in children since the
Best Pharmaceuticals in Children Act and Product Research and
Equity Act have become law, it is increasingly evident that adult
safety proles cannot be transferred directly to children even
when the disease processes are the same.
61
As drug developers
and the pediatric prescribing community seek reasons for these
dierences, exposure-based explanations for target organ toxici-
ties will need to be considered; PBPK metrics oer an approach
to do this, even if the PBPK framework is ultimately used only to
derive cumulative organ exposure or intensity-based metrics.
Finally, one of the main concerns with pediatric drug devel-
opment is that long-term adverse events may not be identied
for many years.
1
is reality can be mitigated by using real-time
evaluation of PBPK results during ongoing pediatric trials to
guide the review of potential PK-mediated safety relationships
and to examine drug safety in the context of the achieved tar-
get exposures (systemic or nonsystemic). In this setting, PBPK
modeling and simulation output can provide inputs to the Data
Safety Monitoring Board to accompany any observed toxicity
or other adverse events.
Examples of successful application of PBPK modeling in
pediatrics
Pediatric PBPK models have been used successfully to predict
dierences in PK between adults and children for several drugs,
such as the disposition of theophylline and midazolam in infants
and children,
11
the maturation of midazolam clearance,
62
and
the prediction of clearance and its variability for 11 drugs
(midazolam—oral and i.v., caeine, carbamazepine, cisapride,
theophylline, diclofenac, omeprazole, S-warfarin, phenytoin,
gentamicin, and vancomycin) in children and infants.
25
ese
models predict the large dierences between half-life and clear-
ance in neonates, infants, and adults.
Edginton et al.
20
extended existing PBPK models for
adults,
63,64
in conjunction with a previously developed age-
specic clearance model,
17
to develop a generic PBPK model
for children that reected age-related physiological changes in
children from birth to 18 years of age. ese authors showed
that this model predicted pediatric plasma PK proles of aceta-
minophen, alfentanil, morphine, theophylline, and levooxacin
reasonably well aer i.v. administration, with appropriate age-as-
sociated trends. Physiological scaling in this model was based on
extensive literature data and took into consideration the fact that
cardiac output, portal vein ow, extracellular water, total body
water, and lipid and protein values are all highly age-dependent.
Although preterm neonates had plasma concentrations greater
than those in adults and older children had concentrations lower
than those in adults (83%, 97%, and 87%, respectively, of the
predicted plasma concentrations), volumes of distribution and
elimination half-lives were within 50% of the study-reported
values. In addition, there was no age-dependent bias in the pre-
diction of distribution volumes or elimination half-lives across
ages ranging from term neonates to 18 years.
20
More recently, a mechanistic pediatric PBPK model that
incorporated interindividual parameter variability was used to
replicate pediatric clinical studies by performing them in silico.
50
Emphasis was placed on drugs used in pediatric anesthesia, and
the PBPK model parameters, which changed in a number of
“what if ” simulations, were used to explore the likely underly-
ing reasons for observed PK proles of drugs such as mida-
zolam, ibuprofen, zolpidem, itraconazole, theophylline, and
caeine. is exemplies the potential application of PBPK
models in pediatric drug clinical investigation (e.g., in assessing
plasma:brain drug concentrations of diclofenac, theophylline,
sildenal, and dextromethorphan) and practice (e.g., in critically
ill children on multiple therapies that present a high potential
for drug–drug interactions).
Extensions of the PBPK model also make it possible to cor-
relate drug disposition with target organ toxicity, an appli-
cation that obviously has great potential for oncology drug
development.
8,65,66
Whereas the time course and severity of
toxicity are oen poorly characterized in a conventional drug
development paradigm, PBPK models have the potential to pro-
vide a more mechanistic understanding of the temporal eects
and the cascade of events that elicit adverse drug reactions and
ultimately toxicity. Accordingly, toxicity-based PD models
driven by target-organ drug exposures would be an excellent
and much-needed extension of current PBPK applications.
67,68
Medical surveillance has also increased the epidemiologic con-
sideration of the benet-to-risk ratio of chronic drug exposures
overlaid on patient disease trajectories.
69,70
Current limitations of pediatric PBPK modeling
Although the list of successful applications of PBPK in pediat-
rics is broad and probably expanding, there are varying degrees
of condence in this approach because its reliability has been
evaluated in only a limited number of published examples or
state artstate art
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 92 NUMBER 1 | JULY 2012 47
unpublished experience within individual companies. Hence,
there remains a clear need to generate reliable experimental data
that can reduce uncertainty in predictions and provide a more
complete understanding of the mechanistic basis for all ADME
processes.
21
Particularly problematic has been the ability of PBPK models
to reliably predict plasma concentrations and PK parameters
aer oral drug administration to patients in various pediatric
age groups. Poulin et al.
15
has shown in adults that currently
available PBPK models have a substantially lower eectiveness
in simulating adult plasma concentration–time proles aer oral
than aer i.v. administration when the models were developed
in the absence of human PK data. ese authors found that up to
69% of the simulations in adults demonstrated a medium to high
degree of accuracy for predicting plasma concentrations aer
i.v. drug administration, whereas this number decreased to 23%
aer oral administration. PBPK prediction of oral absorption
in pediatric patients is further complicated by the facts that the
gastrointestinal tract undergoes distinct developmental changes
and that data about the ontogeny of drug transporters in the gas-
trointestinal tract are sparse.
71
us, PBPK model-predicted bio-
availability of drugs in pediatric patients may be highly biased,
with the consequence that the model-predicted dosage might
result in an inappropriate drug exposure in certain age groups.
erefore, thorough validation of PBPK simulated pediatric
oral doses is necessary before oral administration models can
be relied on to predict dosing regimens for children.
Finally, although PBPK models can be useful in optimizing
the design of pediatric drug studies and in minimizing the par-
ticipation of actual pediatric patients in experimental protocols,
these models are not intended to replace the key clinical studies
of new drugs when it is feasible to carry them out.
8,72
The impact of regulatory authorities on pediatric PBPK
modeling
e increasing use of PBPK modeling in pediatric research and
development is probably a result, in part, of the requirement
to develop a Pediatric Investigation Plan early in the course of
drug development before there has been extensive adult clini-
cal experience.
55
Consequently, it has become more common-
place to use PBPK simulations for predicting proposed pediatric
dose regimens, and sponsors and investigators familiar with the
approach are more comfortable with the results and their inter-
pretation than in the past.
50,73
More generally, the US Food and
Drug Administration has used PBPK M&S to assist in decisions
relating to the need to consider specic clinical pharmacol-
ogy studies, to evaluate proposed study design, and to include
appropriate language in drug labels.
74
It appears that the Food
and Drug Administrations revised protocol for question-based
review will also include questions reecting best practices in the
use of PBPK modeling applications.
74,75
FUTURE DIRECTIONS
As more studies have been completed in children since passage
of the Best Pharmaceuticals in Children Act and the Product
Research and Equity Act, it is increasingly evident that adult
safety proles cannot be transferred directly to children even
when the disease processes are the same.
63
In addition, as drug
developers and the pediatric prescribing community seek rea-
sons for these dierences, exposure-based explanations for
target-organ toxicities will need to be considered, and PBPK
metrics oer an approach to do this even if the PBPK framework
is ultimately used only to derive cumulative organ exposure or
intensity-based metrics. With time it is also possible that these
models will provide insights into (i) pediatric patient popula-
tions dened by altered physiologic states, (ii) nonsystemic
administration in which peripheral exposures are correlated
with outcomes, and (iii) drug dosing requirements appropriate
for chronic administration and relative to dynamic changes in
disease status and progression.
ere is a particular need for reliable experimental data to be
generated in the pediatric population that can reduce uncertainty
in predictions and provide a more complete understanding of
the mechanistic basis for all ADME processes.
21
As microdosing
becomes a viable option for initial clinical testing in children,
this approach may help to ll gaps in the ontogenic information
that is needed and also provide an important quality control
check on the results of PBPK M&S eorts. Extension of these
models to include a PD component that includes covariates
associated with drug–receptor interactions and their maturation
remains an area needing much future research. However, for
PBPK modeling to represent any kind of meaningful alternative
to conventional and better-understood (at least with respect to
the operational characteristics) methods, it must be rigorously
evaluated and reported in a transparent and reproducible man-
ner with agreed-on performance criteria. It will be essential that
PBPK models transparently specify their underlying assump-
tions so that identication of the relevant physiological processes
is clearly dened and guide the development of increasingly reli-
able pediatric physiologically based ADME and PD models to
achieve their potential.
It is apparent that the use of PBPK modeling to support vari-
ous aspects of pediatric research and development is highly
attractive and will probably increase. But for PBPK modeling
to move beyond its application for early-development screen-
ing, users must assess the condence with which the approach
can be used for these applications and determine what is needed
in the future to make these techniques even more robust and
generalizable. It is pertinent that the criteria used in such evalu-
ations be proposed by scientists familiar with pediatric clinical
pharmacology, methodological approaches and alternatives,
computational algorithms, and the assumptions underlying
PBPK models. ese criteria should not be decided by soware
vendors, although their knowledge and development expertise
are important considerations in the evaluation and further evo-
lution of current tools. A further consideration is the necessity
for open disclosure of the details of model construction and
renement when PBPK results are reported and published. Not
unlike an analytical method, the application of PBPK modeling
in all settings must be transparent and reproducible from the
standpoint of the ability of an independent assessor to re-create
modeling outcomes. is is essential for both the integrity of
state artstate art
48 VOLUME 92 NUMBER 1 | JULY 2012 | www.nature.com/cpt
scientic reporting and the ecient regulatory review of PBPK
analyses and for their potential future application in clinical
practice.
A nal but major impediment to the successful application of
PBPK M&S, in both pediatric and other settings, is that there are
far fewer scientists who are appropriately knowledgeable to per-
form these analyses than there are scientists qualied to conduct
traditional PK and PK–PD analyses. For example, the knowledge
of acid/base chemistry, membrane transport, drug metabolism,
and human physiology is perhaps a new requirement for many
members of the M&S and the wider pharmacometric commu-
nity. Unfortunately, the ease with which the newer generations of
PBPK soware can be used may even promote a certain amount
of misuse.” Hence, training and some level of accreditation for
the appropriate knowledge base and skills required to gener-
ate reliable PBPK results will be necessary. e potential of this
technology is great, but so are the risks, particularly where chil-
dren are concerned.
CONFLICT OF INTEREST
The authors declared no conflict of interest.
© 2012 American Society for Clinical Pharmacology and Therapeutics
1. Klassen, T.P., Hartling, L., Hamm, M., van der Lee, J.H., Ursum, J. & Oringa, M.
StaR Child Health: an initiative for RCTs in children. Lancet 374, 1310–1312
(2009).
2. Johnson, T.N. The problems in scaling adult drug doses to children. Arch. Dis.
Child. 93, 207–211 (2008).
3. European Medicines Agency. ICH Topic E11: Note for Guidance on Clinical
Investigation of Medicinal Products in the Paediatric Population (CPMP/
ICH/2711/99) <http://www.ema.europa.eu/docs/en_GB/document_library/
Scientic_guideline/2009/09/WC500002926.pdf>. (Accessed 12 December
2011).
4. Lack, J.A. & Stuart-Taylor, M.E. Calculation of drug dosage and body surface
area of children. Br. J. Anaesth. 78, 601–605 (1997).
5.  Kearns, G.L., Abdel-Rahman, S.M., Alander, S.W., Blowey, D.L., Leeder, J.S. & 
Kauman, R.E. Developmental pharmacology–drug disposition, action, and
therapy in infants and children. N. Engl. J. Med. 349, 1157–1167 (2003).
6. Läer, S., Barrett, J.S. & Meibohm, B. The in silico child: using simulation to guide
pediatric drug development and manage pediatric pharmacotherapy. J. Clin.
Pharmacol. 49, 889–904 (2009).
7. Bellanti, F. & Della Pasqua, O. Modelling and simulation as research tools in
paediatric drug development. Eur. J. Clin. Pharmacol. 67 (suppl. 1), 75–86
(2011).
8. Manolis, E. & Pons, G. Proposals for model-based paediatric medicinal
development within the current European Union regulatory framework.
Br. J. Clin. Pharmacol. 68, 493–501 (2009).
9. Jadhav, P.R. & Kern, S.E. The need for modeling and simulation to design
clinical investigations in children. J. Clin. Pharmacol. 50, 121S–129S (2010).
10. Manolis, E. et al. Role of modeling and simulation in pediatric investigation
plans. Paediatr. Anaesth. 21, 214–221 (2011).
11. Björkman, S. Prediction of drug disposition in infants and children by means
of physiologically based pharmacokinetic (PBPK) modelling: theophylline
and midazolam as model drugs. Br. J. Clin. Pharmacol. 59, 691–704 (2005).
12. Mouksassi, M.S., Marier, J.F., Cyran, J. & Vinks, A.A. Clinical trial simulations
in pediatric patients using realistic covariates: application to teduglutide, a
glucagon-like peptide-2 analog in neonates and infants with short-bowel
syndrome. Clin. Pharmacol. Ther. 86, 667–671 (2009).
13. Meibohm, B., Läer, S., Panetta, J.C. & Barrett, J.S. Population pharmacokinetic
studies in pediatrics: issues in design and analysis. AAPS J. 7, E475–E487
(2005).
14. Läer, S. et al. Development of a safe and eective pediatric dosing regimen for
sotalol based on population pharmacokinetics and pharmacodynamics in
children with supraventricular tachycardia. J. Am. Coll. Cardiol. 46, 1322–1330
(2005).
15. Poulin, P. et al. PHRMA CPCDC initiative on predictive models of human
pharmacokinetics, part 5: prediction of plasma concentration-time proles
in human by using the physiologically-based pharmacokinetic modeling
approach. J. Pharm. Sci. 100, 4127–4157 (2011).
16. Rowland, M., Peck, C. & Tucker, G. Physiologically-based pharmacokinetics
in drug development and regulatory science. Annu. Rev. Pharmacol. Toxicol.
51, 45–73 (2011).
17.  Edginton, A.N., Schmitt, W., Voith, B. & Willmann, S. A mechanistic approach for 
the scaling of clearance in children. Clin. Pharmacokinet. 45, 683–704 (2006).
18. Bartelink, I.H., Rademaker, C.M., Schobben, A.F. & van den Anker,
J.N. Guidelines on paediatric dosing on the basis of developmental
physiology and pharmacokinetic considerations. Clin. Pharmacokinet.
45, 1077–1097 (2006).
19. Cella, M., Gorter de Vries, F., Burger, D., Danhof, M. & Della Pasqua, O. A
model-based approach to dose selection in early pediatric development.
Clin. Pharmacol. Ther. 87, 294–302 (2010).
20.  Edginton, A.N., Schmitt, W. & Willmann, S. Development and evaluation 
of a generic physiologically based pharmacokinetic model for children.
Clin. Pharmacokinet. 45, 1013–1034 (2006).
21. Aarons, L. Physiologically based pharmacokinetic modelling: a sound
mechanistic basis is needed. Br. J. Clin. Pharmacol. 60, 581–583 (2005).
22. Lowe, P.J., Hijazi, Y., Luttringer, O., Yin, H., Sarangapani, R. & Howard, D. On
the anticipation of the human dose in rst-in-man trials from preclinical
and prior clinical information in early drug development. Xenobiotica.
37, 1331–1354 (2007).
23. Jamei, M., Dickinson, G.L. & Rostami-Hodjegan, A. A framework for assessing
inter-individual variability in pharmacokinetics using virtual human
populations and integrating general knowledge of physical chemistry,
biology, anatomy, physiology and genetics: a tale of ‘bottom-up vs ‘top-down’
recognition of covariates. Drug Metab. Pharmacokinet. 24, 53–75 (2009).
24. Alcorn, J. & McNamara, P.J. Ontogeny of hepatic and renal systemic clearance
pathways in infants: part I. Clin. Pharmacokinet. 41, 959–998 (2002).
25. Johnson, T.N., Rostami-Hodjegan, A. & Tucker, G.T. Prediction of the clearance
of eleven drugs and associated variability in neonates, infants and children.
Clin. Pharmacokinet. 45, 931–956 (2006).
26. Johnson, T.N., Tucker, G.T., Tanner, M.S. & Rostami-Hodjegan, A. Changes
in liver volume from birth to adulthood: a meta-analysis. Liver Transpl.
11, 1481–1493 (2005).
27. Ginsberg, G., Hattis, D., Russ, A. & Sonawane, B. Physiologically based
pharmacokinetic (PBPK) modeling of caeine and theophylline in neonates
and adults: implications for assessing childrens risks from environmental
agents. J. Toxicol. Environ. Health Part A 67, 297–329 (2004).
28.  Edginton, A.N. & Willmann, S. Physiology-based simulations of a pathological 
condition: prediction of pharmacokinetics in patients with liver cirrhosis.
Clin. Pharmacokinet. 47, 743–752 (2008).
29. Hurtt, M.E. et al. Juvenile animal studies: testing strategies and design.
Birth Defects Res. B Dev. Reprod. Toxicol. 71, 281–288 (2004).
30. US Food and Drug Administration. Center for Drug Evaluation and
Research. Guidance for Industry: Nonclinical Safety Evaluation of Pediatric
Drug Products, February 2006 <http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm079247.pdf>.
Accessed 16 December 2011.
31. European Medicines Agency. Committee for Medicinal Products for Human
Use. Guideline on the need for non-clinical testing in juvenile animals of
pharmaceuticals for paediatric indications (CHMP/SWP/169215/05), August 
2008 <http://www.ema.europa.eu/docs/en_GB/document_library/Scientic_
guideline/2009/09/WC500003305.pdf>. Accessed 12 December 2011.
32. Parrott, N. et al. Development of a physiologically based model for
oseltamivir and simulation of pharmacokinetics in neonates and infants.
Clin. Pharmacokinet. 50, 613–623 (2011).
33. Baldrick, P. Juvenile animal testing in drug development–is it useful? Regul.
Toxicol. Pharmacol. 57, 291–299 (2010).
34. Zhang, F. et al. Whole-body physiologically based pharmacokinetic model 
for nutlin-3a in mice after intravenous and oral administration. Drug Metab.
Dispos. 39, 15–21 (2011).
35. Bradshaw-Pierce, E.L., Eckhardt, S.G. & Gustafson, D.L. A physiologically
based pharmacokinetic model of docetaxel disposition: from mouse to man.
Clin. Cancer Res. 13, 2768–2776 (2007).
36. Horrobin, D.F. Modern biomedical research: an internally self-consistent
universe with little contact with medical reality? Nat. Rev. Drug Discov.
2, 151–154 (2003).
37.  Kamb, A. Whats wrong with our cancer models? Nat. Rev. Drug Discov.
4, 161–165 (2005).
38. Danhof, M., de Lange, E.C., Della Pasqua, O.E., Ploeger, B.A. & Voskuyl,
R.A. Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD)
modeling in translational drug research. Trends Pharmacol. Sci. 29, 186–191
(2008).
state artstate art
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 92 NUMBER 1 | JULY 2012 49
39. Timchalk, C., Kousba, A.A. & Poet, T.S. An age-dependent physiologically
based pharmacokinetic/pharmacodynamic model for the organophosphorus
insecticide chlorpyrifos in the preweanling rat. Toxicol. Sci. 98, 348–365 (2007).
40. Burghaus, R. et al. Evaluation of the ecacy and safety of rivaroxaban using a
computer model for blood coagulation. PLoS ONE 6, e17626 (2011).
41.  Edginton, A.N., Schmitt, W. & Willmann, S. Application of physiology-based 
pharmacokinetic and pharmacodynamic modeling to individualized target-
controlled propofol infusions. Adv. Ther. 23, 143–158 (2006).
42. Johnson, M. et al. Mechanism-based pharmacokinetic-pharmacodynamic
modeling of the dopamine D2 receptor occupancy of olanzapine in rats.
Pharm. Res. 28, 2490–2504 (2011).
43. Blesch, K.S., Gieschke, R., Tsukamoto, Y., Reigner, B.G., Burger, H.U. & Steimer,
J.L. Clinical pharmacokinetic/pharmacodynamic and physiologically based
pharmacokinetic modeling in new drug development: the capecitabine
experience. Invest. New Drugs 21, 195–223 (2003).
44. Yim, D.S., Zhou, H., Buckwalter, M., Nestorov, I., Peck, C.C. & Lee, H. Population
pharmacokinetic analysis and simulation of the time-concentration prole
of etanercept in pediatric patients with juvenile rheumatoid arthritis. J. Clin.
Pharmacol. 45, 246–256 (2005).
45. Gentry, P.R., Covington, T.R. & Clewell, H.J. 3
rd
. Evaluation of the potential
impact of pharmacokinetic dierences on tissue dosimetry in ospring
during pregnancy and lactation. Regul. Toxicol. Pharmacol. 38, 1–16 (2003).
46. Pelekis, M., Gephart, L.A. & Lerman, S.E. Physiological-model-based derivation
of the adult and child pharmacokinetic intraspecies uncertainty factors for
volatile organic compounds. Regul. Toxicol. Pharmacol. 33, 12–20 (2001).
47. Price, K., Haddad, S. & Krishnan, K. Physiological modeling of age-specic
changes in the pharmacokinetics of organic chemicals in children. J. Toxicol.
Environ. Health Part A 66, 417–433 (2003).
48.  DeWoskin, R.S. & Thompson, C.M. Renal clearance parameters for PBPK model 
analysis of early lifestage dierences in the disposition of environmental
toxicants. Regul. Toxicol. Pharmacol. 51, 66–86 (2008).
49. Edginton, A.N. Knowledge-driven approaches for the guidance of rst-in-
children dosing. Paediatr. Anaesth. 21, 206–213 (2011).
50. Johnson, T.N. & Rostami-Hodjegan, A. Resurgence in the use of physiologically
based pharmacokinetic models in pediatric clinical pharmacology: parallel
shift in incorporating the knowledge of biological elements and increased
applicability to drug development and clinical practice. Paediatr. Anaesth.
21, 291–301 (2011).
51. Baber, N.S. Tripartite meeting. Paediatric regulatory guidelines: do they help
in optimizing dose selection for children? Br. J. Clin. Pharmacol. 59, 660–662
(2005).
52. Reigner, B.G. & Blesch, K.S. Estimating the starting dose for entry into humans:
principles and practice. Eur. J. Clin. Pharmacol. 57, 835–845 (2002).
53. Muller, P.Y., Milton, M., Lloyd, P., Sims, J. & Brennan, F.R. The minimum
anticipated biological eect level (MABEL) for selection of rst human dose in
clinical trials with monoclonal antibodies. Curr. Opin. Biotechnol. 20, 722–729
(2009).
54. Rocchetti, M., Simeoni, M., Pesenti, E., De Nicolao, G. & Poggesi, I. Predicting
the active doses in humans from animal studies: a novel approach in
oncology. Eur. J. Cancer 43, 1862–1868 (2007).
55. Tod, M., Jullien, V. & Pons, G. Facilitation of drug evaluation in children by
population methods and modelling. Clin. Pharmacokinet. 47, 231–243 (2008).
56. Berry, L.M., Roberts, J., Be, X., Zhao, Z. & Lin, M.H. Prediction of V(ss) from in
vitro tissue-binding studies. Drug Metab. Dispos. 38, 115–121 (2010).
57. Holford, N.H., Kimko, H.C., Monteleone, J.P. & Peck, C.C. Simulation of clinical
trials. Annu. Rev. Pharmacol. Toxicol. 40, 209–234 (2000).
58. Barrett, JS. Modeling and simulation in pediatric research and development.
In Clinical Trial Simulations: Applications & Trends (eds. Kimko, H. & Peck, C.),
401–433 (Springer Science + Business Media, New York, 2011).
59. Derendorf, H. & Meibohm, B. Modeling of pharmacokinetic/
pharmacodynamic (PK/PD) relationships: concepts and perspectives.
Pharm. Res. 16, 176–185 (1999).
60.  Lowe, P.J., Tannenbaum, S., Wu, K., Lloyd, P. & Sims, J. On setting the rst 
dose in man: quantitating biotherapeutic drug-target binding through
pharmacokinetic and pharmacodynamic models. Basic Clin. Pharmacol.
Toxicol. 106, 195–209 (2010).
61. Stephenson, T. How children’s responses to drugs dier from adults.
Br. J. Clin. Pharmacol. 59, 670–673 (2005).
62. Anderson, B.J. & Larsson, P. A maturation model for midazolam clearance.
Paediatr. Anaesth. 21, 302–308 (2011).
63. Price, P.S. et al. Modeling interindividual variation in physiological factors used
in PBPK models of humans. Crit. Rev. Toxicol. 33, 469–503 (2003).
64.  Willmann, S. et al. Development of a physiology-based whole-body
population model for assessing the inuence of individual variability on the
pharmacokinetics of drugs. J. Pharmacokinet. Pharmacodyn. 34, 401–431
(2007).
65. Barrett, J. et al. Model-based Drug Development for Oncology Agents. Exp.
Opin. Drug Discov. 2, 185–209 (2007).
66. Steimer, J.L. et al. Modelling the genesis and treatment of cancer: the potential
role of physiologically based pharmacodynamics. Eur. J. Cancer 46, 21–32
(2010).
67. Bruckner, J.V. Dierences in sensitivity of children and adults to
chemical toxicity: the NAS panel report. Regul. Toxicol. Pharmacol. 31,
280–285 (2000).
68.  de Zwart, L.L., Haenen, H.E., Versantvoort, C.H., Wolterink, G., van Engelen, J.G. 
& Sips, A.J. Role of biokinetics in risk assessment of drugs and chemicals in
children. Regul. Toxicol. Pharmacol. 39, 282–309 (2004).
69. Stone, K.D. Atopic diseases of childhood. Curr. Opin. Pediatr. 15, 495–511
(2003).
70. Breitner, J.C. et al. Risk of dementia and AD with prior exposure to NSAIDs in an
elderly community-based cohort. Neurology 72, 1899–1905 (2009).
71. Johnson, T.N. & Thomson, M. Intestinal metabolism and transport of drugs in
children: the eects of age and disease. J. Pediatr. Gastroenterol. Nutr. 47, 3–10
(2008).
72. Hoppu, K. Can we get the necessary clinical trials in children and avoid the
unnecessary ones? Eur. J. Clin. Pharmacol. 65, 747–748 (2009).
73. Khalil, F. & Läer, S. Physiologically based pharmacokinetic modeling:
methodology, applications, and limitations with a focus on its role in pediatric
drug development. J. Biomed. Biotechnol. 90, 74–61 (2011).
74. Zhao, P. et al. Applications of physiologically based pharmacokinetic (PBPK)
modeling and simulation during regulatory review. Clin. Pharmacol. Ther. 89,
259–267 (2011).
75. Huang, S.M. & Rowland, M. The role of physiologically based pharmacokinetic
modeling in regulatory review. Clin. Pharmacol. Ther. 91, 542–549 (2012).
... Physiologically-based-pharmacokinetic (PBPK) modeling has been widely used to investigate the influence of physiological changes in different subjects, or in specific populations, on drug disposition [6][7][8]. The application of PBPK models to predict drug exposure in pregnant women is increasing due to their ability to integrate knowledge from different sources. ...
Article
Full-text available
Physiological changes during pregnancy can alter maternal and fetal drug exposure. The objective of this work was to predict maternal and umbilical ceftazidime pharmacokinetics during pregnancy. Ceftazidime transplacental permeability was predicted from its physicochemical properties and incorporated into the model. Predicted concentrations and parameters from the PBPK model were compared to the observed data. PBPK predicted ceftazidime concentrations in non-pregnant and pregnant subjects of different gestational weeks were within 2-fold of the observations, and the observed concentrations fell within the 5th–95th prediction interval from the PBPK simulations. The calculated transplacental clearance (0.00137 L/h/mL of placenta volume) predicted an average umbilical cord-to-maternal plasma ratio of 0.7 after the first dose, increasing to about 1.0 at a steady state, which also agrees well with clinical observations. The developed maternal PBPK model adequately predicted the observed exposure and kinetics of ceftazidime in the pregnant population. Using a verified population-based PBPK model provides valuable insights into the disposition of drug concentrations in special individuals that are otherwise difficult to study and, in addition, offers the possibility of supplementing sparse samples obtained in vulnerable populations with additional knowledge, informing the dosing adjustment and study design, and improving the efficacy and safety of drugs in target populations.
... The bottlenecks in developing a satisfactory PBPK model and the causes of uncertainty are highlighted in (Table 4) PBPK models can also be used in other scenarios like drug-drug interaction studies. They can be combined with PD models to predict age-appropriate disease outcomes according to the developmental changes among the paediatric population [64]. PK-PD relationships are believed to be comparable between adults and the paediatric population, with some rare exceptions. ...
Article
Full-text available
Age-appropriateness of a formulation is the ability to deliver variable but accurate doses to the paediatric population in a safe and acceptable manner to improve medical adherence and reduce medication errors. Paediatric drug delivery is a challenging area of formulation research due to the existing gap in knowledge. This includes the unknown safety of excipients in the paediatric population, the need for an age-appropriate formulation, the lack of an effective taste-masking method and the lack of paediatric pharmacokinetic data and patient acceptability. It is equally important to establish methods for predicting the biopharmaceutical performance of a paediatric formulation as a function of age. Overcoming the challenges of existing technologies and providing custom-made solutions for the development of age-appropriate formulation is, therefore, a daunting task. Orodispersible films (ODF) are promising as age-appropriate formulations, an unmet need in paediatric drug delivery. New technological improvements in taste masking, improving solubility and rate of dissolution of insoluble drugs, the flexibility of dosing and extemporaneous preparation of these films in a hospital good manufacturing practises (GMP) setup using 3D printing can increase its acceptance among clinicians, patients and caregivers. The current review discusses the problems and possibilities in ODF technology to address the outstanding issues of age-appropriateness, which is the hallmark of patient acceptance and medical adherence in paediatrics.
... The bottlenecks in developing a satisfactory PBPK model and the causes of uncertainty are highlighted in (Table 4) PBPK models can also be used in other scenarios like drug-drug interaction studies. They can be combined with PD models to predict age-appropriate disease outcomes according to the developmental changes among the paediatric population [64]. PK-PD relationships are believed to be comparable between adults and the paediatric population, with some rare exceptions. ...
Article
Opioid use disorders (OUD) during pregnancy are related to neonatal opioid withdrawal syndrome (NOWS). R,S-methadone used to treat OUD and NOWS can penetrate the placenta. High neonatal brain extracellular fluid (bECF) levels of R,S-methadone can induce respiratory depression in newborns. The purpose of this work was to estimate neonatal bECF and saliva levels to establish the neonatal R,S-methadone salivary thresholds for respiratory depression after maternal oral dosing despite the sparse data in pregnancy and newborn populations. An adult physiologically-based pharmacokinetic (PBPK) model for R,S-methadone after intravenous and oral administration was constructed, vetted, and scaled to newborn and pregnancy populations. The pregnancy model predicted the R-methadone and S-methadone doses transplacentally transferred to newborns. Then, the newborn PBPK model was used to estimate newborn exposure after such doses. After maternal oral dosing of R,S-methadone (43.8 mg/day), the neonatal plasma levels were below the respiratory depression threshold. Further, the bECF levels were above the analgesia threshold for more than 96 h. The salivary thresholds for the analgesic effects of R-methadone, S-methadone, and R,S-methadone were estimated herein at 1.7, 43, and 16 ng/mL, respectively. Moreover, the salivary thresholds for the respiratory depression of R-methadone and R,S-methadone were estimated at 58 and 173 ng/mL, respectively. Using neonatal salivary monitoring of methadone can be useful in ensuring newborns' safety during maternal OUD treatment.
Article
Pantoprazole is used to treat gastroesophageal reflux disease (GERD), maintain healing of erosive esophagitis (EE), and control symptoms related to Zollinger–Ellison syndrome (ZES). Pantoprazole is mainly metabolized by cytochrome P450 (CYP) 2C19, converting to 4′-demethyl pantoprazole. CYP2C19 is a genetically polymorphic enzyme, and the genetic polymorphism affects the pharmacokinetics and/or pharmacodynamics of pantoprazole. In this study, we aimed to establish the physiologically based pharmacokinetic (PBPK) model to predict the pharmacokinetics of pantoprazole in populations with various CYP2C19 metabolic activities. A comprehensive investigation of previous reports and drug databases was conducted to collect the clinical pharmacogenomic data, physicochemical data, and disposition properties of pantoprazole, and the collected data were used for model establishment. The model was evaluated by comparing the predicted plasma concentration–time profiles and/or pharmacokinetic parameters (AUC and Cmax) with the clinical observation results. The predicted plasma concentration–time profiles in different CYP2C19 phenotypes properly captured the observed profiles. All fold error values for AUC and Cmax were included in the two-fold range. Consequently, the minimal PBPK model for pantoprazole related to CYP2C19 genetic polymorphism was properly established and it can predict the pharmacokinetics of pantoprazole in different CYP2C19 phenotypes. The present model can broaden the insight into the individualized pharmacotherapy for pantoprazole.
Article
Full-text available
Tirzepatide is an emerging hypoglycemic agent that has been increasing used in adults, yet its pharmacokinetic (PK) behavior and dosing regimen in pediatric population remain unclear. This study aimed to employ the physiologically based pharmacokinetic (PBPK) model to predict changes of tirzepatide exposure in pediatric population and to provide recommendations for its dose adjustments. A PBPK model of tirzepatide in adults was developed and verified by comparing the simulated plasma exposure with the observed data using PK-Sim&MoBi software. This model was then extrapolated to three specific age subgroups, i.e., children (10–12 years), early adolescents (12–15 years), and adolescents (15–18 years). Each subgroup included healthy and obese population, respectively. All known age-related physiological changes were incorporated into the pediatric model. To identify an appropriate dosing regimen that yielded PK parameters which were comparable to those in adults, the PK parameters for each aforementioned subgroup were predicted at pediatric doses corresponding to 87.5%, 75%, 62.5%, and 50% of the adult reference dose. According to the results of simulation, dose adjustments of tirzepatide are necessary for the individuals aged 10–12 years, as well as those aged 12–15 years with healthy body weights. In conclusion, the adult PBPK model of tirzepatide was successfully developed and validated for the first time, and the extrapolated pediatric model could be used to predict pediatric dosing regimen of tirzepatide, which will provide invaluable references for the design of future clinical trials and its rational use in the pediatric population.
Chapter
Physiologically based pharmacokinetic (PBPK) models, with integrating the structural, in silico , and in vitro physicochemical data of drugs and the physiological and anatomical features of the body, provide a realistic characterization of the systemic disposition of drugs. Therapeutic monoclonal antibodies (mAbs), as the fastest growing class of new therapeutic molecules, hold great promise for the treatment of a variety of diseases. This chapter first presents the background and history of PBPK models, and then details the principles and methods of PBPK modelling for mAbs. A number of factors should be particularly considered for antibodies in developing PBPK models: distribution space, extravasation, lymphatic distribution, and specific target binding. Then, the chapter discusses the challenges in PBPK modelling, by considering the physiological parameters, extravasation mechanisms, and FcRn function. The chapter also highlights two situations where the minimal PBPK model enacts target‐mediated drug disposition (TMDD) in either plasma or interstitial space.
Chapter
In this study, we present the application of fractional calculus (FC) in biomedicine. We present three different integer order pharmacokinetics models which are widely used in cancer therapy with two and three compartments and we solve them numerically and analytically to demonstrate the absorption, distribution, metabolism, and excretion (ADME) of drug or nanoparticles (NPs) in different tissues. Since tumor cells interactions are systems with memory, the fractional order framework is a better approach to model the cancer phenomena rather than ordinary and delay differential equations. Therefore, the nonstandard finite difference analysis or NSFD method following the Grünwald-Letinkov discretization may be applied to discretize the model and obtain the fractional order form to describe the fractal processes of drug movement in body. It will be of great significance to implement a simple and efficient numerical method to solve these fractional order models. Therefore, numerical methods using finite difference scheme has been carried out to derive the numerical solution of fractional order two and tri-compartmental pharmacokinetics models for oral drug administration. This study shows that the fractional order modeling extends the capabilities of integer order model into the generalized domain of fractional calculus. In addition, the fractional order modeling gives more power to control the dynamical behaviors of (ADME) process in different tissues because the order of fractional derivative may be used as a new control parameter to extract the variety of governing classes on the non local behaviors of a model, however, the integer order operator only deals with the local and integer order domain. As a matter of fact, NSFD may be used as an effective and very easy method to implement for this type application, and it provides a convenient framework for solving the proposed fractional order models.
Article
Full-text available
Purpose A mechanism-based PK-PD model was developed to predict the time course of dopamine D2 receptor occupancy (D2RO) in rat striatum following administration of olanzapine, an atypical antipsychotic drug. Methods A population approach was utilized to quantify both the pharmacokinetics and pharmacodynamics of olanzapine in rats using the exposure (plasma and brain concentration) and D2RO profile obtained experimentally at various doses (0.01–40 mg/kg) administered by different routes. A two-compartment pharmacokinetic model was used to describe the plasma pharmacokinetic profile. A hybrid physiology- and mechanism-based model was developed to characterize the D2 receptor binding in the striatum and was fitted sequentially to the data. The parameters were estimated using nonlinear mixed-effects modeling . Results Plasma, brain concentration profiles and time course of D2RO were well described by the model; validity of the proposed model is supported by good agreement between estimated association and dissociation rate constants and in vitro values from literature. Conclusion This model includes both receptor binding kinetics and pharmacokinetics as the basis for the prediction of the D2RO in rats. Moreover, this modeling framework can be applied to scale the in vitro and preclinical information to clinical receptor occupancy.
Article
Full-text available
Model-based drug development (MBDD) is an approach that is used to organize the vast and complex data streams that feed the drug development pipelines of small molecule and biotechnology sponsors. Such data streams are ultimately reviewed by the global regulatory community as evidence of a drug's potential to treat and/or harm patients. Some of this information is captured in the scientific literature and prescribing compendiums forming the basis of how new and existing agents will ultimately be administered and further evaluated in the broader patient community. As this data stream evolves, the details of data qualification, the assumptions and/or critical decisions based on these data are lost under conventional drug development paradigms. MBDD relies on the construction of quantitative relationships to connect data from discrete experiments conducted along the drug development pathway. These relationships are then used to ask questions relevant at critical development stages, hopefully, with the understanding that the various scenarios explored represent a path to optimal decision making. Oncology, as a therapeutic area, presents a unique set of challenges and perhaps a different development paradigm as opposed to other disease targets. The poor attrition of development compounds in the recent past attests to these difficulties and provides an incentive for a different approach. In addition, given the reliance on multimodal therapy, oncological disease targets are often treated with both new and older agents spanning several drug classes. As MBDD becomes more integrated into the pharmaceutical research community, a more rational explanation for decisions regarding the development of new oncology agents as well as the proposed treatment regimens that incorporate both new and existing agents can be expected. Hopefully, the end result is a more focussed clinical development programme, which ultimately provides a means to optimize individual patient care.
Article
Full-text available
Physiologically based pharmacokinetic (PBPK) modeling and simulation is a tool that can help predict the pharmacokinetics of drugs in humans and evaluate the effects of intrinsic (e.g., organ dysfunction, age, genetics) and extrinsic (e.g., drug–drug interactions) factors, alone or in combinations, on drug exposure. The use of this tool is increasing at all stages of the drug development process. This report reviews recent instances of the use of PBPK in decision-making during regulatory review. The examples are based on Center for Drug Evaluation and Research reviews of several submissions for investigational new drugs (INDs) and new drug applications (NDAs) received between July 2008 and June 2010. The use of PBPK modeling and simulation facilitated the following types of decisions: the need to conduct specific clinical pharmacology studies, specific study designs, and appropriate labeling language. The report also discusses the challenges encountered when PBPK modeling and simulation were used in these cases and recommends approaches to facilitating full utilization of this tool.
Article
Full-text available
Whether children incur different risks from xenobiotics than adults will depend on the exposure, biokinetics, and dynamics of compound. In this paper, current knowledge on developmental physiology and possible effects on biokinetics are evaluated and the role of biokinetics in risk assessment both for drugs and chemicals is discussed. It is concluded that most dramatic age-related physiological changes that may affect biokinetics occur in the first 6–12 months of age. The difference in internal exposure between children and adults can generally be predicted from already known developmental physiological differences. However, for risk assessment it will also be necessary to determine whether internal exposure is within the drug’s therapeutic window or if it will exceed the NOAEL of a chemical. Furthermore, the effects of internal exposure of potentially harmful compounds on developing organ systems is of utmost importance. However, knowledge on this aspect is very limited. Risk assessment in children could be improved by: (1) application of pediatric PBPK-models in order to gain insight into internal exposure in children, (2) studies in juvenile animals for studying effects on developing systems, and (3) extrapolation of knowledge on the relationship between internal exposure and dynamics for drugs to other chemicals.
Article
Full-text available
Background: Physiologically based pharmacokinetic (PBPK) modelling can assist in the development of drug therapies and regimens suitable for challenging patient populations such as very young children. This study describes a strategy employing PBPK models to investigate the intravenous use of the neuraminidase inhibitor oseltamivir in infants and neonates with influenza. Methods: Models of marmoset monkeys and humans were constructed for oseltamivir and its active metabolite oseltamivir carboxylate (OC). These models incorporated physicochemical properties and in vitro metabolism data into mechanistic representations of pharmacokinetic processes. Modelled processes included absorption, whole-body distribution, renal clearance, metabolic conversion of the pro-drug, permeability-limited hepatic disposition of OC and age dependencies for all of these processes. Models were refined after comparison of simulations in monkeys with plasma and liver concentrations measured in adult and newborn marmosets after intravenous and oral dosing. Then simulations with a human model were compared with clinical data taken from intravenous and oral studies in healthy adults and oral studies in infants and neonates. Finally, exposures after intravenous dosing in neonates were predicted. Results: Good simulations in adult marmosets could be obtained after model optimizations for pro-drug conversion, hepatic disposition of OC and renal clearance. After adjustment for age dependencies, including reductions in liver enzyme expression and renal function, the model simulations matched the trend for increased exposures in newborn marmosets compared with those in adults. For adult humans, simulated and observed data after both intravenous and oral dosing showed good agreement and although the data are currently limited, simulations in 1-year-olds and neonates are in reasonable agreement with published results for oral doses. Simulated intravenous infusion plasma profiles in neonates deliver therapeutic concentrations of OC that closely mimic the oral profiles, with 3-fold higher exposures of oseltamivir than those observed with the same oral dose. Conclusions: This work exemplifies the utility of PBPK models in predicting pharmacokinetics in the very young. Simulations showed agreement with a wide range of observational data, indicating that the processes determining the age-dependent pharmacokinetics of oseltamivir are well described.
Article
Full-text available
The concept of physiologically based pharmacokinetic (PBPK) modeling was introduced years ago, but it has not been practiced significantly. However, interest in and implementation of this modeling technique have grown, as evidenced by the increased number of publications in this field. This paper demonstrates briefly the methodology, applications, and limitations of PBPK modeling with special attention given to discuss the use of PBPK models in pediatric drug development and some examples described in detail. Although PBPK models do have some limitations, the potential benefit from PBPK modeling technique is huge. PBPK models can be applied to investigate drug pharmacokinetics under different physiological and pathological conditions or in different age groups, to support decision-making during drug discovery, to provide, perhaps most important, data that can save time and resources, especially in early drug development phases and in pediatric clinical trials, and potentially to help clinical trials become more "confirmatory" rather than "exploratory".
Article
Background: Liver cirrhosis is a progressive disease characterized by loss of functional hepatocytes with concomitant connective tissue and nodule formation in the liver. The morphological and physiological changes associated with the disease substantially affect drug pharmacokinetics. Whole-body physiologically based pharmacokinetic (WB-PBPK) modelling is a predictive technique that quantitatively relates the pharmacokinetic parameters of a drug to such (patho-)physiological conditions. Objective: To extend an existing WB-PBPK model, based on the physiological changes associated with liver cirrhosis, which allows for prediction of drug pharmacokinetics in patients with liver cirrhosis. Methods: The literature was searched for quantitative measures of the physiological changes associated with the presence of Child-Pugh class A through C liver cirrhosis. The parameters that were included were the organ blood flows, cardiac index, plasma binding protein concentrations, haematocrit, functional liver volume, hepatic enzymatic activity and glomerular filtration rate. Predictions of pharmacokinetic profiles and parameters were compared with literature data for the model compounds alfentanil, lidocaine (lignocaine), theophylline and levetiracetam. Results: The predicted versus observed plasma concentration-time profiles for alfentanil and lidocaine were similar, such that the pharmacokinetic changes associated with Child-Pugh class A, B and C liver cirrhosis were adequately described. The theophylline elimination half-life was greatly increased in Child-Pugh class B and C patients compared with controls, as predicted by the model. Levetiracetam urinary excretion was consistently reduced with disease progression and very closely resembled observed values. Conclusion: Consideration of the physiological differences between healthy individuals and patients with liver cirrhosis was important for the simulation of drug pharmacokinetics in this compromised group. The WB-PBPK model was altered to incorporate these physiological differences with the result of adequate simulation of drug pharmacokinetics. The information provided in this study will allow other researchers to further validate this liver cirrhosis model within a WB-PBPK model.
Chapter
Pediatric research and development is typically poorly funded in both academic and industrial settings creating greater incentive for optimized trial designs with high degree of technical success. Likewise, the application of modeling and simulation approaches is extremely valuable in the evaluation of pediatric trial design. Beyond bridging adult dose-exposure relationships, pediatric clinical trial simulation models must accommodate relevant developmental, maturational, and size relationships on both PK and PD expressions. Design considerations that address sample size per age strata, the probability of achieving adult exposure-response targets, and assumptions regarding PD response vs. outcomes are especially valuable in the support of pediatric drug development and/or dosing guidance.
Article
During regulatory review of clinical pharmacology data in new drug applications and biologics license applications, questions are routinely asked about how intrinsic factors (e.g., organ dysfunction, age, and genetics) and extrinsic factors (e.g., drug-drug interactions) might influence dose-response and exposure-response and about the impact of these individual factors on the efficacy and safety of the candidate compound. Physiologically based pharmacokinetic (PBPK) modeling and simulation is one of the tools that can be used to address these critical questions.
Article
Recent legislation in the United States and Europe has resulted in an increased number of clinical trials of pharmaceutical agents in children. Creating a well-designed clinical trial that can be successfully completed is a challenging task, particularly as the study population includes younger and smaller children. Although there are some established principles for initially estimating appropriate doses of pharmaceutical agents in children based on known effective doses in adults, these rules are inadequate as the sole basis for designing a clinical trial in children. Factors such as maturation of metabolizing enzymes, relative physical maturation of the child, and altered absorption because of physiological differences in adults and children may contribute to alterations in the dose-exposure relationship. To account for the impact of these potential factors on a clinical trial, the use of modeling and simulation is necessary to anticipate the influence these variables can have on the desired clinical question to be addressed. The examples presented in this article highlight the principle that modeling and simulation is critical for adequately designing pediatrics trials.