Anda di halaman 1dari 10

Supplement Article

The Path to Perfect Pediatric Posology — The Journal of Clinical Pharmacology


2018, 58(S10) S48–S57
Drug Development in Pediatrics 
C 2018, The American College of

Clinical Pharmacology
DOI: 10.1002/jcph.1081

Joan M. Korth-Bradley, PharmD, PhD, FCP, FAAPS

Abstract
Reluctance to enroll pediatric subjects in clinical trials has left gaps in information about dosing, safety, and efficacy of medications. Pharmacotherapeutic
information for pediatric patients may be available for only a small range of ages and may be deficient, as children respond differently as they grow and
mature from prematurity to adolescence. Current regulations, however, require early planning for the participation of children in drug development, as
pediatric plans must be submitted at the end of phase 1 (European Union) or the end of phase 2 (United States). These plans are extensive, outlining
planned studies, subjects to be enrolled, dose and dosage form justification, planned observations, and statistical analysis as well as planned modeling,
simulation, and extrapolation analyses. The extent to which efficacy information in adults can be extrapolated to children depends on how similar the
disease is in adults and each of the 5 pediatric age groups. Extrapolation may not be possible for conditions that do not occur in adults, requiring a
complete development plan in adults, or extrapolation may be complete because of similar pathology and response to treatment. Pharmacokinetic and
safety information cannot be extrapolated and must be collected in children of all ages, unless a waiver is granted. Physiologically based pharmacokinetic
modeling, optimal design, population pharmacokinetics, and scavenged samples are all examples of new methodologies being used to study pediatric
therapeutics. Clinicaltrials.gov and EU Clinical Trials registry are good sources of results of pediatric trials, although sponsors are also working toward
prompt publication of study results in peer-reviewed journals.

Keywords
clinical research, pharmacodynamics, pharmacokinetics, physiologically based pharmacokinetic models, neonates

It is not enough to simply tailor information available who sometimes died with a few hours of onset of
in adults to fit children. In the not-too-distant past, the distress. The collection of symptoms was referred to
use of Clark’s rule and other formulae assumed children as gray syndrome because of the ashen appearance of
were small or young adults and scaled accordingly.1 the skin. Assay of 200-μL blood samples, collected
Doses for children were determined by multiplying using finger- and heel-sticks, in affected infants showed
the adult dose by the ratio of the child’s weight to their chloramphenicol concentrations to be unusually
150 pounds, for example. Studies to verify that doses high. A series of focused clinical studies subsequently
so calculated were appropriate, safe, and effective were conducted in groups of 3 to 5 patients showed
rarely conducted, in part because of the hesitancy to greatly prolonged half-lives (26 hours) in the very
subject children to the demands of participation in clin- young children who were 1 to 2 days of age, compared
ical trials. Information about the stability, palatability, with half-lives of 10 hours in babies who were 10 to
or acceptability of either the commercially available or 16 days of age. Ultimately, it was determined that
extemporaneously prepared dosage forms was limited. slow conversion of chloramphenicol to metabolites
Unfortunately, we have learned from therapeutic and immature renal tubular excretion were likely
misadventure that pediatric-specific pharmacokinetic, responsible for the high concentrations. It was
safety, and efficacy data must be collected across the subsequently recommended that full-term infants
age and developmental spectrum so that clinicians can receive 50 mg/kg/day and premature infants receive
use medication optimally and avoid potentially severe 25 mg/kg/day for the first week of life. The report
adverse reactions. It is useful to consider an example summarizing the investigation,2 published almost
to understand why pediatric subjects must be included 60 years ago, encapsulates solutions to many of the
in drug development. Chloramphenicol at doses of
100 mg/kg/day in divided doses was administered to Clinical Pharmacology, Global Product Development, Pfizer Inc., Col-
neonates to treat or prevent infections in hospital legeville, PA, USA
nurseries.2 Cases were reported of babies who had Submitted for publication 6 October 2017; accepted 21 December 2017.
been treated for 3 or 4 days and were then observed Corresponding Author:
to have abdominal distention with or without emesis, Joan M. Korth-Bradley, PharmD, PhD, Pfizer Inc, 500 Arcola Road,
progressive pallid cyanosis, and vasomotor collapse, Collegeville, PA 19426
frequently accompanied by irregular respiration and Email: joan.korth-bradley@pfizer.com
Korth-Bradley S49

challenges of determining the best dose in young Table 1. Pediatric Age Ranges Used for Labeling
infants and even recommends the use of therapeutic European
drug monitoring for prolonged therapy. Medicines Agency
It is now expected that medications come to market and International
with directions for safe and effective use for patients US Food and Drug Conference on
Classification Administration Harmonisation
of all ages or, alternatively, with statements that use
in certain age ranges is not indicated for the approved Preterm newborn <37 weeks of age = PMAa
indications. The directions, including contraindications infant
and cautions, should be based on data collected in Neonate (full-term Birth to 1 month 0 to 27 days
newborn)
appropriately designed studies or robust documenta-
Infant (toddler) 1 month to 2 years 1 to 23 months
tion from epidemiological and toxicology studies. It is Children 2 to 12 years 2 to 11 years
unfortunate that previous hesitancy to include children Adolescent 12 to <17 years 12 to <18 years
in clinical trials has resulted in gaps in the informa- a
PMA is postmenstrual age.
tion available. Current drug development practices will
hopefully make these deficiencies a thing of the past.
If we agree that all drugs that are developed should required either for the label itself or by other disciplines,
include children of all ages in clinical trials, let us so that they may perform their studies. Drug develop-
first look at the drug development process in general. ment is highly regulated, and so the data required for
The potential list of pharmaceutical products that approval and, in turn, the studies required to provide
could be developed is almost endless. With thousands much of the data are described in regulatory guidance
of diseases already described, continuing investigation statements and regulations, including the definition of a
into pathophysiology and identification of potential child.
targets that could be manipulated to remediate or
even cure and huge expansion of types of medicines Definition of Pediatric Age Groups
that can be synthesized, there is a very long list of To be sure to gather data across the spectrum of pe-
potential products that could be developed. Choices diatric development milestones, children are separated
about which particular projects to advance are based into 5 age groups, shown in Table 1, and each age group
on many factors. What therapeutic areas is the sponsor must be considered in clinical development. Jurisdic-
(company, manufacturer) of the development project tions other than the United States3 and the European
already working in? It is easier to develop and market Union4 may have slightly different limits to the age
products to an existing community. What are the largest ranges. In some development programs, there may be
threats to public health or well-being that demand further subdivision of the 2- to 12-year age group of
immediate attention and are likely to be supported those younger than than 6 years and those at least 6
by payers? Big, messy problems, such as Alzheimer’s years old. Specific developmental milestones or conven-
disease, may be solved more readily if there are many tions in medical practice may impose other restrictions
different sponsors working on the problem. Ongoing on age. For example, tetracycline causes discoloration
collaboration between academic researchers and pri- in developing teeth, and so is not recommended for
vate industry may also suggest emphasis on particular children younger than 8 years old.5 Hemophilia re-
diseases. placement products include children aged 12 years
Drug development begins with the end in mind, and older in the clinical trials of adults.6 These age
by making a mockup of what would be the ideal groups are not simply arbitrary divisions, but were
product labeling, including the indication, posology developed to map onto stages of development that
for all age groups, and the anticipated adverse reac- coincide with stages of maturity and milestones in
tions and precautions required for the most effective development. Those who care for children recognize
and safe use. With ideal product labeling outlined, that there are different body systems influencing re-
a development plan is prepared that includes all the sponses to pharmacotherapy and that these systems
clinical and nonclinical studies required to provide mature at different rates with added variability because
information needed to support the specific language in of genetic, environmental, and social influences.7 In
the label. Each discipline, including discovery, clinical addition to the obvious differences in weight between
and nonclinical pharmacology, clinical and medical different age groups, there may also be differences
research, product formulation, toxicology, safety and in gastric pH, body composition, cytochrome P450
epidemiology, among other functions, then expands metabolic capacity, transporters, renal function, and
its portion of the development plan to assess what is hepatic function. For example, neonates have elevated
already known about the new chemical entity and what intragastric pH and lower total volume of gastric secre-
studies need to be performed to gather new information tions and immature conjugation and biliary function.8
S50 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

As therapeutic proteins become common, more will be Recently enacted legislation, the Pregnancy and
learned about the ontogeny of target engagement and Lactation Labeling Rule (PLLR),11 which requires
the elimination of monoclonal antibodies and fusion inclusion of information about the safety and
proteins, among others.9 Children may also differ in pharmacokinetics of medications in pregnant and
the signs, symptoms, and pathology of diseases from lactating women, is being implemented and will be
children in different age groups or adults with similar largely completed by 2019. Studies of pregnant and
disease. Some conditions are present at birth but may be lactating women are not part of drug development
delayed in manifesting pathology or may not respond in children, and further discussion is beyond the
to delayed treatment. By including children from each scope of this article, but implementation of the PLLR
of these age categories in drug development, it is hoped provides important information about indirect dose
that there will be adequate information available when administration to infants.
a drug is first available on the market that will enable In the European Union, the Paediatric Regulation,
prescribers caring for children who may be maturing at which was enacted in 2007, resulted in the establish-
different rates or who are smaller or larger than average ment of the Paediatric Committee (PDCO). The main
to make good choices of doses to use. responsibility of the PDCO is to determine the studies
that sponsors must conduct in children as part of
Paediatric Investigation Plans (PIPs). Because the PIP
Regulatory Guidance is rate limiting among these various legislations, clinical
Drug development has evolved such that inclusion of development plans for children will frequently begin
pediatric subjects in clinical development or, at a min- with it. PIPs are the development plans describing the
imum, justification for why their involvement should necessary data to be obtained through studies in chil-
be deferred or waived, is now a legal requirement. dren that will support the approval of a medicine, in the
Most potential drug products are developed with the European Union. All applications for new medicines
goal of global registration in mind, and so clinical have to include the results of studies as described in an
development programs are planned to include elements agreed-on PIP, unless the medicine is exempt because of
that will satisfy the requirements of the major markets a deferral or waiver. This requirement also applies when
of the United States, Europe, and Japan and be in a sponsor wants to add a new indication, a new dosage
accordance with recommendations of the International form, or a route of administration for a medicine that
Conference on Harmonisation. The key regulations is already approved. PIPs are rate limiting because they
and summary documents that provide a framework on must be submitted to the PDCO no later than the end
how pediatric trials will be incorporated into clinical of the first phase 1 studies to assess pharmcokinetics
development include an alphabet soup of the Food, in healthy adult subjects or patients.12 They cannot be
Drug and Cosmetics (FD&C) Act in the United submitted after the initiation of pivotal trials, confir-
States, the Food and Drug Administration Safety and matory, phase 3 trials, or trials conducted in children.
Innovation Act, enacted in 2012, in which regulations It is important to note that the PIP must be agreed to
originally approved in the Best Pharmaceuticals for by the European Medicines Agency (EMA) and must
Children Act (BPCA, 2002) and the Pediatric Research be fulfilled by the sponsor, unless an amendment is also
Equity Act (PREA, 2003) are included. Under the agreed to by the EMA. Failure to complete the PIP to
BPCA, sponsors of certain applications could obtain the satisfaction of the EMA will result in the sponsor
an additional 6 months of exclusivity if the sponsor being unable to file the application for the applicable
submits information responding to a written request new pharmaceutical product.13
relating to the use of a drug in the pediatric population. As noted previously, drug development in the United
All new medications including both drug and biological States is governed by the FD&C Act. One of the
products are covered, although those with “orphan important milestones is the end of the phase 2 meeting.
drug” designation are exempt from PREA regulations. This meeting is held between the Food and Drug
Under the PREA, studies in pediatric patients are Administration (FDA) staff in the division that is
required for the indication that will be part of the new reviewing the ongoing development and the sponsor.
drug application unless granted a waiver or deferral. The meeting’s purpose is to review the safety informa-
Waivers may be issued if the indication is not present in tion that has been gathered in phase 1 and phase 2
pediatrics, would not represent a significant therapeutic studies, to evaluate the phase 3 plan and protocols, to
advantage, or poses a significant safety concern. identify additional information necessary to support a
Studies in rare diseases in pediatric subjects may also marketing application, and to review plans for studies
be eligible for the added incentive of a priority review in pediatric subjects. The initial pediatric study plan
voucher. Priority review vouchers may be transferred (iPSP)14 describing these studies and/or justification for
to other programs or sold to other sponsors.10 waivers and deferrals should be discussed with the FDA
Korth-Bradley S51

Queson 1: Do children has similar disease progression and response to treatment as adults? No extrapolaon
(1) Conduct a dose-ranging study in children to establish dose
If no – then proceed to ‘no extrapolaon’, if yes, proceed to queson 2.
(2) Conduct safety and efficacy studies at established dose in children
Queson 2: Is it likely that similar exposure-responses
will be observed in children and adults?

Paral extrapolaon
If no – proceed to queson 4, if yes, proceed to queson 3.
(1) Conduct dose-ranging study in children to idenfy dose
that achieves the target PD effect
Queson 3: Does drug/metabolite concentraon predict clinical response?
(2) Conduct safety study at idenfied dose
If no – proceed to queson 4, if yes, proceed to ‘full extrapolaon’

Queson 4: Is there a PD measurement that will predict efficacy in children? Full extrapolaon
(1) Conduct PK study in children to idenfy dose
If no – proceed to ‘no extrapolaon’; if yes, proceed to ‘paral extrapolaon’ that achieves similar exposure to adults
(2) Conduct safety study at the idenfied dose
Figure 1. Algorithm to determine extent of extrapolation.
Figure 2. Studies required by degree of extrapolation.

prior to the end of phase 2, and the PSP should be pharmacokinetic and safety data need to be collected
submitted within 60 days of the end of the phase 2 in children, whereas partial extrapolation will mean a
meeting. All marketing applications including a new dose-finding study is required, and no extrapolation
active ingredient, new indication, new dosage form, of information from adults means a sponsor is obliged
new dosing regimen, or new route of administration to conduct separate dose-finding, safety, and efficacy
are required to submit an iPSP. iPSPs are reviewed by studies in children.15 It must be remembered that the 4
the Pediatric Review Committee, which will provide questions must be answered for each of the 5 pediatric
comments to the sponsor and ultimately issue the age categories, and different answers to the questions
agreed initial PSP. The goal of the PSP process is to may result in different degrees of extrapolation. For
identify the necessary pediatric studies so that they example, drugs used to treat infections are examples of
can be completed prior to the submission of the new medication for which full extrapolation is likely.16 It is
drug application or biologics license application in the reasonable to assume that if similar concentrations of
case of biological therapeutics. For most sponsors, the an antibiotic are achieved in children, whether neonates
iPSP is modeled on the PIP, which is required earlier in or adolescents, then the infection should respond in a
development. similar way, but as described in the chloramphenicol
example, pharmacokinetic data and safety data cannot
Pediatric Studies be extrapolated and must be collected in all age groups.
As noted previously, the road map to clinical An example of an indication that does not al-
development is the clinical plan, which is mapped out low extrapolation is treatment for retinopathy of
prior to administration of the very first dose to a human prematurity.17 This condition does not occur in adults,
subject and is discussed with regulatory authorities in and so all data supporting the safety, efficacy, and phar-
meetings prior to submission of the investigational new macokinetics must be collected in children, specifically
drug. The pediatric components may not be completely in those who are premature infants. Bevacizumab is
described at the pre—investigational new drug meeting, an antivascular endothelial growth factor monoclonal
but as noted above, a PIP is required soon after the antibody approved for the treatment of adults with
completion of the first-in-human study conducted and metastatic colorectal cancer, nonsquamous, non–small
the iPSP prior to the end of phase 2, so there will be cell lung cancer, metastatic breast cancer, and glioblas-
some vagueness in the initial plans, although the general toma. After experimental use in adults with intraocular
outline of what studies will be performed in what age neovascular diseases without severe adverse reactions,
groups is expected. To determine the degree to which investigators explored its use in combination with stan-
adult data may be used to support pediatric indications, dard nonpharmacological therapy in an 8-week-old
an algorithm, shown in Figure 1, that includes 4 key infant who was born at 25 weeks’ gestation.18 Then,
questions has been developed. Depending on the a large prospective, randomized trial was conducted
responses to the 4 questions, sponsors will be able in 150 infants 33 to 35 weeks postmenstrual age, who
to assess the extent of extrapolation possible for were treated with 0.625 mg administered by intravitreal
adult data. Neither pharmacokinetic nor safety injection into each eye.19 Efficacy was based on pho-
observations can be extrapolated, and some data must tographs taken at 54 weeks’ postmenstrual age that were
be collected in every age group for which labeling is evaluated for recurrence of retinopathy, again unique
expected. As shown in Figure 2, full extrapolation to this condition. Because the dosing was empiric,
of efficacy information from adults means that only additional trials are ongoing to assess the effectiveness
S52 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

of lower doses.17 The safety profile of systemically General Study Considerations for
administered bevacizumab was well described from Pediatric Subjects
studies in adults being treated for cancer but was not
The age range in a dedicated pediatric pharmacokinetic
applicable to infants treated with intravitreal injections
or pharmacokinetic/pharmacodynamic study should
who would receive 1 or 2 doses in each affected eye, with
be consistent with the pediatric age range for the
minimal anticipated systemic availability.
intended population of treatment. Clinical trials in
In addition to medications developed with either
pediatric populations are conducted in those individ-
no extrapolation or full extrapolation of efficacy from
uals who stand to potentially benefit from the treat-
adults are intermediate cases, for which information
ment. Healthy children participate in studies such as
obtained in adults can be supplemented with efficacy
immunization trials against diseases that they might
data from children. Adult to adolescent extrapolation is
reasonably be expected to contract22 or postapproval
relatively common.20 As noted above, in rare diseases,
assessment of nutritional supplements.23 Reasonably
such as hemophilia, adolescents are enrolled in adult
stable children with the target disease participate in
studies without distinction because of their age,6 and
clinical pharmacology studies as well as in clinical
for some conditions, such as migraine headache and
studies of medications used to treat diseases that occur
schizophrenia,15 there is a continuity of both disease
only in children.24 Legal regulations and social conven-
and response to treatment between adults and pediatric
tions assert that children lack the autonomy and life
populations. Adolescents are similar in size to adults
experience to judge the risk and benefit of participating
and have mature physiology. Their quest for autonomy
in a clinical trial. Institutional review boards and ethics
and potential lack of compliance may make participa-
committees reviewing and approving studies being con-
tion in clinical trials challenging but is needed to vali-
ducted in children must include adequately trained and
date assumptions of similar pharmacokinetics, safety,
experienced clinicians and researchers who are able to
and efficacy.21 Extrapolation of efficacy and response
assess the risks and benefits on behalf of the vulnerable
data from adults to children younger than 12 years
pediatric population. In addition to the consent to
of age may be more difficult than for adolescents and
participate given by parents or guardians, it is also good
becomes progressively more difficult in the youngest
practice to provide age-appropriate information about
age groups because of immature renal function and
the clinical trial and obtain assent from the potential
metabolic activity, differences in body composition, and
participants if their maturity and condition allow.
challenges in drug administration. For some, a waiver
Because there are fewer pediatric patients available to
is preferred because the disease does not manifest
participate in clinical trials and because children are a
itself or the assessment of response is not possible,
vulnerable population, study-site selection is important
such as allergic conjunctivitis in children younger than
to ensure that the clinical sites have the infrastructure
2 years of age, for example.15 Onset of disease may
in place to meet the protocol requirements, especially if
be indicative of a different pathology than disease that
special techniques or assessments are required. Fortu-
usually occurs in adults. For example, congestive heart
nately, as pediatric studies have become a regular part
failure in children is more likely from congenital heart
of drug development, expertise is becoming more avail-
disease, whereas in adults, ischemic heart disease is the
able. Sponsors have formed internal pediatric councils,
culprit.15 Partial extrapolation is possible when there is
among whose members are toxicologists, pathologists,
uncertainty in the assumption that the disease and the
clinical pharmacologists, formulators, assay specialists,
response to treatment are similar in adults and children.
statisticians, researchers, and clinicians with expertise
Either a single, adequate, well-controlled trial proving
in pediatric drug development. These councils review
efficacy in addition to safety and pharmacokinetics
clinical development plans and study protocols prior
can be performed, or when there is less uncertainty,
to submission to regulatory authorities to ensure high-
a study underpowered to demonstrate efficacy can be
quality pediatric development programs that are deliv-
conducted. Juvenile rheumatoid arthritis (JRA) is an
ered in a timely fashion while attending to the safety
example of partial extrapolation as well as evolution
of participants in the trials. In addition to internal pe-
of requirements as more children participate in clinical
diatric councils, pediatric research networks have been
studies.15 For symptomatic relief, when there was an ex-
expanded to include sponsors in addition to academic
tensive understanding of mechanism of action, extrap-
research centers.25 Research networks improve the ef-
olation was permitted, or at least inclusion of different
ficiency of drug development by providing education
subtypes in the same study. For disease-modifying
and certification for investigators that can be recog-
drugs, the FDA requires independent confirmation of
nized by different sponsors. Through their experience,
efficacy in pediatric populations in each of the subtypes
they can offer direction for improving trial design,26
of JRA.15
developing and validating standard processes for
Korth-Bradley S53

subject enrollment, dosing, collection of observations, genetically determined occur more commonly in boys,
including blood sampling,27 and information about the such as hemophilia; at some ages, such as apnea of
natural history of disease, growth, and development. prematurity; or predominately in some races, such as
Networks can be organized to span different countries sickle cell disease. Cultural sensitivity is needed when
and regions to promote the highest standards of clinical enrolling girls who could potentially have achieved
research, including attention and support of pediatric menarche, and older children who may be participating
subjects and their families to minimize the burdens of in sexual activity or illicit drug use. Thoughtful
participating in clinical trials. Networks vary in their explanation of the testing necessary to protect the
size, diseases, and populations of interest and funding safety of the children, as well as the validity of the
sources. Examples of pediatric research networks are interpretation of the study results, is important to the
the Children’s Oncology Group,28 the European Net- conduct of the study. Beyond general considerations
work of Paediatric Research at the European Medicines of designing clinical trials in children, 3 common
Agency, the Pediatric Trials Network,29 the Institute elements, dose, observations, and analysis, require
for Advanced Clinical Trials for Children, and the particular attention. These will now be considered in
Paediatric Trials Network Australia. turn.
The decision to conduct a study as an inpatient or
an outpatient trial is based on considerations similar
to those in adults and include the age of the child, Pediatric Dose
the illness, the treatment required, the observations To optimize the benefit:risk ratio in trials in patients,
being made, and the location of staff and equipment. investigators try to avoid both ineffective doses and
Studies of premature infants are typically inpatient doses higher than required during dose-finding or phar-
studies. Children in stable condition may be able to macokinetic studies and include only doses predicted
participate as outpatients or with only a limited number to achieve concentrations that have been shown in
of overnight stays. Accommodation must be made for nonclinical studies or simulations to be efficacious
parents or guardians who may need to stay with the and tolerable. When the clearance of a drug can be
child and provide transportation of the child to the re- predicted in children, then the dose required to achieve
search unit. Timing of the activities in studies must also a target concentration can be calculated, albeit with
consider school schedules and immunization schedules, sometimes large variability. Differences in volume of
which may make participation in trials difficult for some distribution must also be considered but are usually of
age groups. concern only when dosing is intermittent or a particular
Knowledge of the impact of coadministration of target concentration must be achieved immediately on
food with the study drugs is important in the design starting treatment.
of clinical trials in children.30 Healthy adult volunteers The most common method of determining clearance
and many adult patients can tolerate administration in in pediatric subjects is weight-based allometric scaling.
fasted conditions or administration with a standardized Clearance is assumed to be a function of weight at
meal. It is difficult to be confident of compliance on three-quarters power.32 However, because age-related
the part of children. Infants and young children are maturation is not included in calculations, allometric
fed 4 to 6 times daily, consuming breast milk, formula, scaling is not reliable for young children, especially
or semisolid food, such as cereal, yogurt, and pureed those younger than 2 years,7 because of continuing
vegetables, fruits, and meats. Children may refuse to ongoing maturation of metabolic pathways. Segmented
consume standard meals or perhaps even food of any allometric models, which implement different expo-
kind, particularly if they are feeling unwell. When nents for different ages, have been developed to accom-
the effect of food is either unknown or a source of modate different age groups33 and dosage forms such
variability in clinical response, it is important to collect as extended-release tablets that support only limited
information about the time and composition of the flexibility in dosing.34
last meal consumed prior to dose administration to Physiologically based pharmacokinetic (PBPK)
help to interpret the results. The effect of food on models may be able to overcome some of the limitation
pharmacokinetics or efficacy and safety can also be of allometric scaling. PBPK models integrate phys-
incorporated into the study design by allowing children iologic information, such as age-specific blood flow,
to consume one of several meals or remain fasting and protein concentration, and enzyme and transporter
noting their feeding status as a covariate.31 ontogeny with pharmacokinetic parameters such
The decision to include children of different as percent of dose eliminated by renal clearance or
races/ethnicities and sexes is made in a similar metabolized by particular pathways and can be used
manner as in adults. No group of children with the to predict the disposition in children of various levels
target disease is excluded, but some diseases often of maturity.35,36 Advances in software, the increased
S54 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

availability of physiological and tissue distribution data tablets that may be more stable to be used in cases in
have resulted in more frequent use of PBPK models which some flexibility in dosing is permissable.35,40
for drugs in development. Models have been developed The relative bioavailability between the different
for both small molecules and biologics. Edington and dosage forms must be known so that as children are
colleagues37 extended preexisting PBPK models for switched, the required dosing is maintained. Stabil-
acetaminophen, alfentanil, morphine, theophylline, ity information is also required for all dosage forms
and levofloxacin to include data for children and and understanding of the impact of excursions in
evaluated the performance of the models in predicting temperature, such as when doses are not refrigerated.
pediatric plasma profiles. Eighty-three percent, 97%, Dissolution testing, which is an important metric for
and 87% of the predicted plasma concentrations, the development of oral dosing forms, needs to be
volumes of distribution, and elimination half-lives, evaluated using a relevant dissolution media if the
respectively, were within 50% of values reported. PBPK product will be used in very young children whose
models are then used to help to design dosing schemes, physiology differs from older children and adults.41
PK sampling times, and times for other observations Beyond oral formulations, differences between
in clinical trials in pediatric subjects. For example, adults and children must also be considered.42 For
sirolimus has not been investigated in children yunger example, medications that are injected will require
than 13 years old, so Emoto and colleagues used PBPK concentrations that are neither too potent, when it is
methods to develop a model for sirolimus to make difficult to accurately administer the very small doses
predictions for children aged 1 month to 2 years who required for premature infants, or too dilute, which
were to be enrolled in a phase 2 efficacy and safety trial would require large injection volumes that would not
for treatment of vascular anomalies.38 Regardless of be tolerated for subcutaneous injection. There are
the method used to select the doses to be evaluated in many different ways of administering medication by
protocols, it is a best practice to include a maximum aerosol that vary in the effectiveness in delivery as well
dose to avoid overdosing large or heavy children. as degree of cooperation required of the patient and
skill in use by research staff.43 Drugs administered as
eye drops are also challenging for treating conditions
in children. Because of the small volume administered,
Dosage Forms the concentration of active ingredients is high. The eye
Important to accurate, precise, and consistent dose of a newborn is approximately two-thirds of the adult
administration is the availability of an appropriate size and does not reach adult size until age 3 to 4 years.
dosage form for the intended population. Although Ocular dosing is not weight or size adjusted, and so
there are many different ways that medication can be children may receive much higher doses than adults.44
administered and all can be adapted for different age Protocols and supplementary dose administration
groups, the most common route of administration is instructions need to be clear so that the doses are
oral, and the most common oral dosage forms used administered as consistently and accurately as possible.
in children are liquids and solids, including powders,
sprinkles, dissolving tablets, minitablets, and tablets.
Discussion of dosage forms is an integral part of Observations
PIPs, and so each of the following must be described Technical challenges in collection, standardization,
in the PIP document4,35 : the specific formulation, phar- and interpretation have limited the use of potentially
maceutical form, strength and route of administration less invasive matrices45 such as saliva, urine, and hair,
for each of the pediatric age groups, potential issues and so blood samples are collected in almost every type
concerning excipients and their predicted exposures at of pediatric study in drug development, whether for as-
the anticipated dose, administration of doses to the sessment of safety, efficacy, or pharmacokinetics, even
different pediatric age groups, including acceptability, in critically ill and very small patients. To characterize
use of administration devices, ability to mix with milk, pharmacokinetic parameters in individual subjects,
formula, or food, precision of the dose delivery and/or conventional pharmacokinetic sampling requires 10 to
accuracy of the dosage form for each of the pedi- 12 samples collected over a dosing interval in multiple-
atric age groups, and the anticipated time frame for dose studies or more than 3 to 5 half-lives in single-
development of the formulations and dosage forms. dose studies. A review46 of guidelines on sampling
Dosage forms used with children must be palatable and permitted in pediatric studies reported a maximum
must permit flexible dosing to accommodate reasonable cumulative volume of blood of 3–7 mL/kg collected
precision over the large range of doses that are required. over 8 weeks. Faced with the necessity of including
Use of dosing bands, by assigning children over a range baseline and end-of-study safety assessments of serum
of weights to the same dose, allows use of capsules or chemistry, hepatic, renal, and hematology values,
Korth-Bradley S55

pharmacokinetic sampling in young children and the limited number of samples collected, individual
infants, by necessity, is sparse, representing only a determination of pharmacokinetic parameters for each
subset of conventional sampling. Optimal sampling subject may be only post hoc estimates based on a
designs may be used in which blood samples are population pharmacokinetic model. Naive pooled esti-
collected at the times that are most likely to provide mates may also be used to determine pharmacokinetic
information about the likely model47 as well as times parameters,51 although care must be taken to normalize
linked to particular safety or efficacy signals, such as for dose administered and size of the sample of the
likely peaks and troughs. Simulations of the anticipated subjects studied.
concentration–time profiles are helpful to avoid
sampling times that are likely to be uninformative
because the concentrations fall below the limits of
quantification of the assay. Disclosure/Publication of Study Results
In addition to minimizing the volume of blood Important to the protection of subjects who participate
collected, investigators must also minimize the number in clinical trials are the processes requiring quick, com-
of collection times. Opportunistic study designs, also plete disclosure. All interventional studies conducted
known as scavenged sampling, which schedule research in the United States must be registered on the website
sample collection to coincide with sample collection www.clinicaltrials.gov. The clinicaltrials.gov informa-
for routine laboratory draws,48 may be used in studies tion resource was initiated as a result of the Food
in children. Authors have suggested that opportunistic and Drug Administration Modernization Act of 1997.
study designs are useful for further investigation of The registry was established by the National Institutes
medications that are already prescribed to children of Health for experimental treatments for serious or
rather than investigational drug products. The decision life-threatening diseases or conditions and was initially
to prescribe the medication is made outside the clinical intended to provide patients searching for available
trial, but once the order has been written, enrollment in trials a central place to look. It was expanded in 2007
the study occurs, with consent focused on the collection to require additional types of trials to be registered
of extra blood volume to answer the research question. as well as reporting summary results, including ad-
The limitations of varied timing and number of samples verse events. Under most circumstances for studies in
make opportunistic sampling strategies difficult to use children, results must be posted within 12 months of
in drug development. the date of final data collection for the prespecified
Monitoring for safety and efficacy in pediatric sub- primary outcome measures.52 European regulations
jects is incorporated into research protocols in much established Article 46 of Regulation (EC) 1901/2006,53
the same manner as for adults. Baseline assessment under which studies, of products that are registered in
takes place prior to administration of the study drug the European Union, enrolling pediatric subjects must
to verify that subjects are eligible to enroll, do not provide a completed clinical study report to the EMA
have any exclusion characteristics, and will be able within 6 months of study completion. The requirements
to cooperate with study assessments. For the study apply to all studies with pediatric subjects, even if not
results to be meaningful, assessments must be age included in a PIP. The EMA will in turn make all
appropriate and capable of detecting safety signals results available through the EU Clinical Trials Register
unique to children, such as interference with growth49 (www.clinicaltrialsregister.eu).
and development. Specific systems may be more (or Publication of study results in a peer-reviewed jour-
less) tolerant of drug effect as a result of a combination nal is the final step in making information available
of differences in physiology and body composition. For to the public. Guidelines54 have been developed en-
example, higher brain penetration of medications in couraging prompt and balanced reporting of all com-
neonates, both human and animal, may be the result of pleted clinical trials, regardless of whether they were
active transport systems evolved to transport nutrient positive. Because of the difficulty in enrolling pediatric
and other molecules needed for development of the subjects, it is particularly important that these data be
central nervous system, as well as differences in volume presented and made available to add to the literature.
of distribution related to body composition, reduced The primary publication is the initial publication of a
blood plasma protein-binding capacity, and reduced clinical trial and should be submitted for publication
renal flow.50 ideally within 12 months (18 months at the latest) of the
completion of the clinical trials.54 Although data are
not available specifically for trials of pediatric subjects
Data Analysis or of clinical pharmacology studies, approximately
Data analysis methods used for pediatric studies are 90% of studies registered at clinicaltrials.gov have been
similar to those used for adult studies. Because of published.55
S56 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

Next Steps and Remaining Questions 10. US Department of Health and Human Services Food and
Drug Administration Center for Drug Evaluation and Research
With the inclusion of pediatric subjects as a standard (CDER), Center for Biologics Evaluation and Research (CBER)
part of clinical development, more data are being and Office of Orphan Products Development (OOPD). Rare Pe-
collected so that drug development in children has ex- diatric Disease Priority Review Vouchers, Guidance for Industry.
panded far beyond simple description of a small group 2014.
11. US Department of Health and Human Services Food and
of heterogeneous subjects. Beyond studying children of Drug Administration Center for Drug Evaluation and Re-
all ages, the problem of the correct dose for obese56 search (CDER). Pregnancy, Lactation, and Reproductive Po-
or underweight children is beginning to get attention. tential: Labeling for Human Prescription Drug and Biological
Perhaps more importantly, the advances that are being Products—Content and Format Guidance for Industry. 2014.
made to support drug development work in children, 12. European Medicines Agency. Paediatric Investigation Plans:
Questions and Answers. http://www.ema.europa.eu/ema/
including in fragile newborns, can be extended to the index.jsp?curl=pages/regulation/q_and_a/
study of other vulnerable populations such as pregnant q_and_a_detail_000015.jsp&mid=WC0b01ac0580925cc7.
and lactating women.57 The goal of perfect pediatric Accessed September 10, 2017
posology may indeed be at hand. 13. European Medicines Agency. Questions and answers on
the procedure of PIP compliance verification at EMA,
and on paediatric rewards. EMA/PDCO/179892/2011
Declaration of Conflicting Interests Rev. 2, 2014. http://www.ema.europa.eu/docs/en_GB/
Joan M. Bradley is a Senior Director, Clinical Pharma- document_library/Regulatory_and_procedural_guideline/2009/
cology, Pfizer Inc. She is an employee and shareholder 09/WC500003916.pdf. Accessed October 1, 2017.
14. US Department of Health and Human Services, Food and
of the company. Drug Administration, Center for Drug Evaluation and Research
(CDER), Center for Biologics Evaluation and Research (CBER).
References Pediatric study plans: Content of and process for submitting
initial pediatric study plans and amended initial pediatric study
1. Subrahmanya NK, Patel KS, Kori VK, Shrikrishna R. Role plans. Guidance for Industry (draft guidance). 2016.
of Kasahara Dashemani Vati in Kasa and Vyadhikshamatva in 15. Dunne J, Rodriguez WJ, Murphy MD, et al. Extrapolation
children with special reference to recurrent respiratory tract of adult data and other data in pediatric drug-development
infections. Ayu. 2013;34:281–287. programs. Pediatrics. 2011;128:e1242–e1249.
2. Weiss CF, Glazko AJ, Weston JK. Chloramphenicol in the 16. Nikanjam M, Kabamba D, Cressey TR, et al. Nevirapine expo-
newborn infant. A physiologic explanation of its toxicity sure with WHO pediatric weight band dosing: enhanced thera-
when given in excessive doses. N Engl J Med 1960;262:787– peutic concentrations predicted based on extensive international
794. pharmacokinetic experience. Antimicrob Agents Chemother.
3. US Department of Health and Human Services Food and 2012;56:5374–5380.
Drug Administration Center for Drug Evaluation and Research 17. Wallace DK, Kraker RT, Frreman SF, et al. Assessment of lower
(CDER). General Clinical Pharmacology Considerations for Pe- doses of intravitreous bevacizumab for retinopathy of prematu-
diatric Studies for Drugs and Biological Products. Guidance for rity. A phase 1 dosing study. JAMA Ophthalmol. 2017;135:654–
Industry. 2014. 656.
4. European Commission. Guideline on the format and content of 18. Chung EJ, Kim JH, Ahn HS, Koh HJ. Combination of laser
applications for agreement or modification of a paediatric photocoagulation and intravitreal bevacizumab (Avastin R
) for
investigation plan and requests for waivers or deferrals aggressive zone I retinopathy of prematurity. Graefe’s Arch Clin
and concerning the operation of the compliance check Exp Ophthalmol. 2007;245:1727–1730.
and on criteria for assessing significant studies. Official J 19. Mintz-Hittner HA, Kennedy KA, Chuang AZ. Efficacy of
of EU Union. 2014/C 338/01. https://ec.europa.eu/health/ intravitreal bevacizumab for stage 3+ retinopathy of prematurity.
/sites/health/files/files/eudralex/vol-1/2014_c338_01/ N Engl J Med. 2011;364:603–615.
2014_c338_01_en.pdf. Accessed September 10, 2017. 20. Momper JD, Mulugeta Y, Green DJ, et al. Adolescent dosing
5. Vennila V, Madhu V, Rajesh R, Ealla KKR, Velidandla SR, and labeling since the Food and Drug Administration Amend-
Santoshi S. Tetracycline-induced discoloration of deciduous ments Act of 2007. JAMA Pediatr. 2013;167:926–932.
teeth: Case series. J Int Oral Health. 2014;6:115–119. 21. Kapogiannis BG, Mattison DR. Adolescents in clinical trials.
6. Committee for medicinal products for human use (CHMP), Eu- Clin Pharm Ther. 2008;84:655–659.
ropean Medicines Agency. Guideline on the clinical investigation 22. Huang L-M, Puthanakit T, Cheng-Hsun, C, et al. Sustained
of recombinant and human plasma-derived factor VIII products. immunogenicity of 2-dose human papillomavirus 16/18 ASO4-
EMA/CHMP/BPWP/144533/2009. 2011. adjuvanted vaccine schedules in girls aged 9–14 years: a random-
7. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, ized trial. J Infect Dis. 2017;215:1711–1719.
Leeder JS, Kauffman RE. Developmental pharmacology–drug 23. Aglipay M, Birken CS, Parkin PC, et al. Effect of high-dose vs
disposition, action, and therapy in infants and children. N Engl standard-dose wintertime vitamin D supplementation on viral
J Med. 2003;349:1157–1167. upper respiratory tract infections in young healthy children.
8. Nicholas J-M, Bouzom F, Hugues C, Ungell A-L. Oral drug JAMA. 2017;318:245–254.
absorption in pediatrics: the intestinal wall, its developmental 24. Koren G, Kearns GL, Reed M, Pons G. Use of healthy children
changes and current tools for predictions. Biopharm Drug Dispos. as volunteers in drug studies: The ethical debate. Clin Pharm
2017;38:209–230. Ther. 2003;73:147–152.
9. Zhang Y, Wei X, Bajaj G, et al. Challenges and considerations 25. Turner MA, Attar S, deWildt SN, Vassal G, Mangiarini L,
for development of therapeutic proteins in pediatric patients. J Giaquinto C. Roles of clinical research networks in pediatric
Clin Pharm. 2015;55(S3):S103–S115. drug development. Clin Ther. 2017;in press.
Korth-Bradley S57

26. Laughon MM, Benjamin DK, Capparelli EV, et al. Innovative 41. Batchelor HK, Fotaki N, Klein S. Paediatric oral biopharmaceu-
clinical trial design for pediatric therapeutics. Expert Rev Clin tics: key considerations and current challenges. Adv Drug Deliv
Pharmacol. 2011;4:643–652. Rev. 2014;73:102–106.
27. Hawcutt DB, Rose AC, Nunn T, Turner MA. NIHR Medicines 42. Batchelor HK, Marriott JF. Formulations for children:
for Children Research Network (MCRN): Points to consider problems and solutions. Br J Clin Pharmacol. 2015;79:405–
when planning the collection of blood samples in clinical trials 418.
of investigational medicinal products. 2008. 43. Ari A. Drug delivery interfaces: a way to optimize inhalation
28. Henderson TO, Parsons SK, Wroblewski KE, et al. Outcomes therapy in spontaneously breathing children. World J Clin Pe-
in adolescents and young adults with Hodgkin lymphoma diatr. 2016;5:281–287.
treated on US Cooperative Group protocols: an adult intergroup 44. Farkouh A, Frigo P, Czejka M. Systemic side effects of
(E2496) and Children’s Oncology Group (COG AHOD0031) eye drops: a pharmacokinetic perspective. Clin Ophthalmol.
comparative analysis. Cancer. 2018;24:136–144. 2016;10:2433–2441.
29. England A, Wade K, Smith PB, Berezny K, Laughon M. Opti- 45. Wille SMR, Baumgartner MR, Di Fazio V, Samyn N, Kraemer
mizing operational efficiencies in early phase trials: the Pediatric T. Trends in drug testing in oral fluid and hair as alternative
Trials Network experience. Contemp Clin Trials. 2015;47:376– matrices. Bioanalysis. 2014:6:2193–2209.
382. 46. Howie SRC. Blood sample volumes in child health research:
30. Martir J, Flanagan T, Mann J, Fotaki N. Recommended strate- review of safe limits. Bull World Health Organ. 2011;89:46–
gies for the oral administration of paediatric medicines with food 53.
and drinks in the context of their biopharmaceutical properties: 47. Perera V, Bies RR, Mo G, et al. Optimal sampling of an-
a review. J Pharm Pharmacol. 2016;69:384–397. tipsychotic medicines: a pharmacometric approach for clinical
31. Borrmann S, Sallas WM, Machevo S, et al. The effect of food practice. Br J Clin Pharmacol. 2014;78:800–814.
consumption on lumefantrine bioavailability in African chil- 48. Gonzalez D, Melloni C, Yogev R, et al. Use of opportunistic
dren receiving artemether—lumefantrine crushed or dispersible clinical data and a population pharmacokinetic model to support
tablets (Coartem R
) for acute uncomplicated Plasmodium falci- dosing of clindamycin for premature infants to adolescents. Clin
parum malaria. Trop Med Int Health. 2010;15:434–441. Pharmacol Ther. 2014;96:429–437.
32. Anderson BJ, Holford NHG. Mechanism-based concepts of size 49. Skoner DP. Inhaled corticosteroids. Effects on growth and
and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. bone health. Ann Allergy Asthma Immunol. 2016;117:595–
2008;48:303–332. 600.
33. Mahood I. Dosing in children: a critical review of the pharma- 50. Schmitt G, Parrott N, Prinssen E, Barrow P. The great barrier
cokinetic allometric scaling and modelling approaches in paedi- belief: the blood-brain barrier and considerations for juvenile
atric drug development and clinical settings. Clin Pharmacokinet. toxicity studies. Repro Tox. 2017;72:129–135.
2014;53:327–346. 51. Pouplin T, Bang ND, Toi PV, et al. Naı̈ve-pooled pharma-
34. Boni J, Korth-Bradley JM, Martin P, et al. Pharmacokinetics cokinetic analysis of pyrazinamide, isoniazid and rifampicin
of etodolac in patients with stable juvenile rheumatoid arthritis. in plasma and cerebrospinal fluid of Vietnamese children with
Clin Ther. 1999;21:1715–1724. tuberculous meningitis. BMC Infect Dis. 2016;16:144.
35. Johnson TN, Rostami-Hodjgan A, Tucker GT. Prediction of the 52. Zarin DA, Tse T, Williams RJ, Carr S. Trial reporting in Clin-
clearance of eleven drugs and associated variability in neonates, icalTrials.gov — the Final Rule. N Engl J Med. 2016;375:1998–
infants and children. Clin Pharmacokinet. 2006;45:931–956. 2004.
36. Jones HM, Mayawala K, Poulin P. Dose selection based on phys- 53. European Medicines Agency. Submission of Article-46
iologically based pharmacokinetic (PBPK) approaches. AAPS J. paediatric studies: questions and answers. Rev. December
2013;15:377–387. 2014. http://www.ema.europa.eu/ema/index.jsp?curl=pages/
37. Edginton AN, Schmitt W, Willmann S. Development and evalu- regulation/q_and_a/q_and_a_detail_000044.jsp&mid=
ation of a generic physiologically based pharmacokinetic model WC0b01ac0580023e7f. Accessed October 3, 2017.
for children. Clin Pharmacokinet. 2006;48:1013–1034. 54. Battisti WP, Wager E, Baltzer L, et al. Good Publication Practice
38. Emoto C, Fukuda T, Johnson TN, Adams DM, Vinks AA. for Communicating Company-Sponsored Medical Research:
Development of a pediatric physiologically based pharmacoki- GPP3. Ann Intern Med. 2015;163:461–464.
netic model for sirolimus: applying principles of growth and 55. Phillips AT, Desai NR, Krumholz HM, Zou CX, Miller JE,
maturation in neonates and infants. CPT Pharmacometics Syst Ross JS. Association of the FDA Amendment Act with trial reg-
Pharmacol. 2015;4;e17. istration, publication, and outcome reporting. Trials. 2017;18:
39. Van Riet-Nales DA, Kozarewicz P, Aylward B, et al. Paediatric 333.
drug development and formulation design—a European perspec- 56. Hawcutt DB, Bellis J, Price V, et al. Growth hormone prescribing
tive. AAPS Pharm Sci Tech. 2017;18:241–262. and initial BMI SDS: Increased biochemical adverse effects and
40. Sevinsby H, Zaru L, Wang R, et al. Pharmacokinetics and costs in obese children without additional gain in height. PLoS
pharmacodynamics of atazanavir in HIV-1-infection children One. 2017;12:e0181567.
treated with atazanavir powder and ritonavir: combined analysis 57. Zimmerman K, Gonzalez D, Swamy GK, Cohen-Wolkowiez
of the PRINCE-1 and -2 studies [published online ahead of print M. Pharmacologic studies in vulnerable populations: using the
2017]. Ped Infect Dis J. pediatric experience. Semin Perinatol. 2015;39:532–536.

Anda mungkin juga menyukai