Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Supplement Article

Optimizing Antibiotic Drug Therapy The Journal of Clinical Pharmacology


2018, 58(S10) S108–S122
in Pediatrics: Current State and Future 
C 2018, The American College of

Clinical Pharmacology
DOI: 10.1002/jcph.1128
Needs

Jennifer Le, PharmD, MAS, BCPS-ID, FIDSA, FCCP, FCSHP1


and John S. Bradley, MD, FAAP, FIDSA, FPIDS2,3

Abstract
The selection of the right antibiotic and right dose necessitates clinicians understand the contribution of pharmacokinetic variability stemming from
age-related physiologic maturation and the pharmacodynamics to optimize drug exposure for clinical response. The complexity of selecting the right
dose arises from the multiplicity of pediatric age groups, from premature neonates to adolescents. Body size and age (which relate to organ function)
must be incorporated to optimize antibiotic dosing in this vulnerable population. In the effort to optimize and individualize drug dosing regimens, clinical
pharmacometrics that incorporate population-based pharmacokinetic modeling, Bayesian estimation, and Monte Carlo simulations are utilized as a
quantitative approach to understanding and predicting the pharmacology and clinical and microbiologic efficacy of antibiotics. In addition, opportunistic
study designs and alternative blood sampling strategies can serve as practical approaches to ensure successful conduct of pediatric studies. This review
article examines relevant literature on optimization of antibiotic pharmacotherapy in pediatric populations published within the last decade. Specific
pediatric antibiotic data, including beta-lactam antibiotics, aminoglycosides, and vancomycin, are critically evaluated.

Keywords
Pharmacokinetic, pharmacodynamic, pediatrics, neonates, antimicrobials, vancomycin, Monte Carlo simulation, beta-lactams, vancomycin, aminoglyco-
sides, antibiotics, Bayesian methods, opportunistic study, ceftaroline

Effective pharmacotherapy in pediatric patients with independent of year of publication, were selected to
infection requires the integration of pertinent data highlight important attributes of pediatric pharmacol-
on the antibiotic, bacterial pathogen, and patient. In ogy. Recent articles with new information related to
addition to appropriate antibiotic selection based on pediatric pharmacology and specific antibiotics, par-
the susceptibility of the pathogen and the site of ticularly focusing on literature within the past decade,
infection, optimizing antibiotic therapy to achieve the were critically reviewed and are presented in this review.
desired outcome in pediatric patients necessitates the
understanding of growth and development related to Pharmacokinetics: Alterations Stemming
age for both physiologic and immunologic processes From Growth and Development
that govern drug absorption, distribution, metabolism,
The two key factors stemming from physiologic mat-
and elimination, as well as maturation of granulocyte,
uration that contribute to PK alterations in pediatric
lymphocyte, and humoral immune function. Physi-
patients are body size and organ function (which is
ologic development may alter the pharmacokinetics
defined by age). The complexity of PK variability arises
(PK) and impact the pharmacodynamics (PD) of drugs.
In selecting the right antibiotic and dose, clinicians
must understand the contribution of PK variability
1 SkaggsSchool of Pharmacy and Pharmaceutical Sciences, University of
stemming from age-related physiologic maturation and
California at San Diego, La Jolla, CA, USA
the PD to optimize antibiotic exposure for clinical 2 Department of Pediatrics, Division of Infectious Diseases, University of
response.1 While the immature nature of the immune California at San Diego, La Jolla, CA, USA
system in neonates and infants may potentially affect 3 Rady Children’s Hospital San Diego, San Diego, CA, USA

the PD of antibiotics, this article focuses on the effect of Submitted for publication 29 November 2017; accepted 1 March 2018.
PKPD alterations stemming from physiologic changes
Corresponding Author:
that occur with age on antibiotic therapy. A PubMed Jennifer Le, PharmD, University of California San Diego, Skaggs School of
search for publications up to November 2017was con- Pharmacy and Pharmaceutical Sciences, 9500 Gilman Drive, MC 0714, La
ducted using key terms relevant to pediatric PKPD and Jolla, CA 92093-0714
specific antibiotics reviewed in this article. Key articles, Email: jenle@ucsd.edu
Le and Bradley S109

from the multiple pediatric age groups, from premature Table 1. Maturational Physiologic Changes Affecting Drug Pharmacoki-
neonates to adolescents. “Pediatric patients” denotes all netics in Pediatric Patients13
pediatric age groups from birth to 18 years, including Age Group
premature neonate (born before 37 weeks’ gestation), Reaching Adult
neonates (within first month of life), infants (1 to 12 Variable Neonatea Valueb
months), children (1 to 12 years), and adolescents (13 Absorption
to 18 years). The neonatal and infant periods com- Gastric pH ↑ Child
prise the most dramatic maturational developments Gastric and intestinal emptying ↑ Child
that affect a drug’s PK. While age-related physiologic time
Biliary function, pancreatic ↓ Infant
maturation continues into childhood, the most promi-
function, and gut microbial
nent differences, compared with adults, are evident in colonization
neonates and infants. Table 1 describes the maturational Intramuscular absorption ↓ Child
processes that affect a drug’s absorption, distribution, Skin permeability and ↑ Child
metabolism, and renal elimination in pediatric patients percutaneous absorption
Distribution
compared with adults. The age group in which the
Total body water and ↑ Late childhood
maturational process reaches adult values are also extracellular water
provided. Total body fat and muscle ↓ Child
mass
Total plasma binding proteins ↓ Early childhood
Hepatic distribution and elimination
Body Size CYP1A2 ↓ Early childhood
Drug dosing in pediatric patients should be scaled to CYP2C9 ↓ (30% of adult Infant, with peak
body size, expressed as milligram per kilogram. Total activity) activity in
body weight is a common and appropriate measure to childhood
use for drug dosing in normal-sized children. However, CYP2C19 ↓ (30% of adult Infant, with peak
activity) activity in
pediatric drug doses cannot be normalized directly childhood
from an adult dose using total body weight (ie, adult CYP2D6 ↓ (30% of adult Infant
dose in milligram per kilogram) when pediatric dosing activity)
data are unavailable. This is because drug elimination, CYP2E1 No data, with ↓ Late childhood
particularly via the renal route, is nonlinear to weight.2 in infant
CYP3A4 ↓ (30%-40% of ↑ in early
Two contrasting consequences arise from this nonlinear adult activity) childhood, with
relationship: the underestimation of drug clearance progressive decline
(CL) in children and its overestimation in adults.2 CYP3A7103 ↑ Low to
While total body weight is an inadequate body size undetectable in
descriptor to apply in pediatric drug dosing that is early childhood
Uridine 5-diphosphate ↓ Early childhood
derived from adult data, a child’s body size can be glucuronyltransferase
referenced to a 70-kg adult using allometric scaling— N-acetyl transferase 2 ↓ Early childhood
a coefficient of 1 for volume distribution (Vd; weight1 ) Methyltransferase ↑ Infant
and 0.75 for CL (weight0.75 ).3 This allometric approach Sulfotransferase ↓ Adolescent, with ↑
to the prediction of drug CL has been applied in in infant and child
Renal elimination
children older than 5 years, including children with Glomerular filtration ↓ Infant
cystic fibrosis (CF) and, possibly, obesity (albeit other Tubular secretion and ↓ Infant
factors play a role in children who are obese).4–6 reabsorption
The exponent of 0.75 has been used to allometrically a
↓ = decreased, ↑ = increased activity compared with adults.
scale weight to describe the maturation of glomerular b
Pediatric age groups include premature neonate (born before 37 weeks
filtration rate (GFR) in pediatric patients, including gestation), neonates (within first month of life), infants (1 to 12 months),
neonates.7 The 0.75 allometric scaling was superior to children (1 to 12 years), and adolescents (13 to 18 years).
body surface area (which uses an exponent of 0.67)
in measuring GFR, liver and kidney volumes, and
cardiac output.2 However, for neonates and infants, this Age
exponent may not be applicable for predicting the CL Similar to weight, age is another significant factor to
of some drugs, especially those that have elimination integrate when optimizing drug dosing in neonates,
outside of glomerular filtration.8 Ideally, to optimize infants, and young children because it correlates well
age-specific dosing across the pediatric age spectrum, with organ function responsible for the maturation of
the exact value used for allometric scaling should be CL, both renal and hepatic. In the healthy pediatric
derived for each individual drug through PK studies. population, age defines organ function. The presence
S110 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

of diseases, such as CF, may alter organ function. Maturation of hepatic pathways that affect drugs
Relative to body mass, the size and weight of organs, undergoing metabolic transformation or elimination
specifically the kidneys and liver, which are responsible occurs at diverse rates at different ages, ranging
for drug CL, are several-fold greater in younger from the first months to years of life. The body’s
children compared with adults. Clearance accounts primary organ for drug metabolism, the liver, produces
for both metabolism and elimination of a drug from microsomal cytochrome P450 enzymes at birth.
the body and largely determines the maintenance The activity of most of these enzymes is reduced
dose. Renal elimination, including reduced GFR, during infancy, with maturation occurring from
tubular secretion, and reabsorption, is immature in infancy into late childhood, depending on the specific
neonates and infants. Because of this, the CL of enzyme (Table 1).2 Consequently, the metabolic CL,
antibiotics that undergo elimination primarily via particularly the glucuronidation reaction, is lower in
GFR (including certain beta-lactams, aminoglycosides, neonates and infants than in adults. Two exceptions
and vancomycin) are decreased in neonates and infants to the underdeveloped liver function in neonates and
relative to older pediatric age groups. This may lead to infants are methyltransferase and sulfotransferase,
increased drug exposure in neonates and infants and respectively, for which enhanced activity is observed.
potentially increases the risk for toxicity, particularly The different rates of hepatic maturation, along with
nephrotoxicity for aminoglycosides, if the dosing is not the complexity of the hepatic CL of drugs that may
adjusted.9 use different mechanisms at varying degrees, precludes
Within the first 2 weeks of life, a rapid surge in renal accurate prediction of drug dosages. Enzymatic matu-
function is observed that is due to an increase in renal ration has been described using the sigmoidal Hill equa-
blood flow.10,11 This marks the most prominent change tion and coefficients that incorporate postmenstrual
within the first year of life and occurs nonlinearly with age and the maturation half-time (TM50 ).2 In neonates
age during this interval. The nonlinear relationship be- and infants, calculations based on postmenstrual age
tween age and renal function follows an evident plateau are more accurate than calculations based on other
of drug dosing, expressed as milligram per kilogram. types of age because it accounts for maturation that
Consequently, increasing drug dosage is necessary up occurs during gestation.
to a certain age, when dosage then decreases to that In addition to renal and hepatic functions that
appropriate for adults. contribute to a drug’s metabolism and elimination (ie,
An in-depth discussion of age (usually referred as CL) and therefore dosing, age also influences drug
postnatal age or chronologic age) is crucial with respect distribution through body water or fat composition and
to neonates because nephrogenesis, which is respon- plasma protein binding. Body composition and protein
sible for the increase in the GFR in utero, requires binding change with age. Drug distribution, which is
completion of 36 weeks’ gestation.10,11 By definition, represented by Vd, equates to the total amount of
gestational (or menstrual) age is the time between the drug distributed within the blood and tissues. Drugs
first day of the last normal menstrual period and that remain in the circulation and distribute mainly
birth; postnatal age accounts for the duration of life in the extracellular fluid compartment (eg, hydrophilic
after birth; and postmenstrual age incorporates both drugs, including aminoglycosides and linezolid) usually
gestational and postnatal age. For premature neonates possess a relatively low Vd. In contrast, drugs that
born before 36 weeks’ gestation, a drug’s PK may distribute intracellularly or well in tissues generally have
depend on more than postnatal (or chronologic) age a relatively high Vd. Because physiologic maturation
because nephrogenesis has not occurred. The use of that occurs with age alters body composition and
postnatal age compared with postmenstrual age as an plasma protein binding, the Vd of drugs in pediatric
indicator for the maturation of drug elimination may, patients changes accordingly (Table 1). For example,
therefore, be suboptimal for premature neonates.3 In the augmented total body and extracellular water con-
fact, postmenstrual age was more useful than post- tent evident in neonates and young infants translates
natal age in describing GFR in premature and full- to enhanced Vd of aminoglycosides and linezolid and,
term neonates.7 In a large pediatric study of pooled consequently, the need for increased loading doses
data from 923 participants ranging from 22 weeks’ (milligram per kilogram) to achieve adequate drug
postmenstrual age to 31 years, allometric scaling was exposure (ie, usually by peak concentration).9,12 The
applied to predict and compare GFR between pediatric reverse is observed for highly lipophilic drugs such as
participants and adults. Interestingly, half of the adult clindamycin.13
GFR (ie, approximately 60 mL/min per 70 kg) was Another important factor that affects drug distri-
reached at 48 weeks of postmenstrual age and 90% of bution is plasma protein binding. The decrease in
the adult GFR, at 1 year of age.7 Age becomes less both the concentration and affinity of plasma binding
influential in late childhood and adolescence. proteins such as albumin and alpha-1-acid glycoprotein
Le and Bradley S111

in neonates, infants, and young children alters the distri- Vd and CL, that determine drug dosing.19 The incorpo-
bution of drugs, resulting in an increase in the unbound ration of covariates, particularly weight and age, as ex-
moiety (which is the free, pharmacologically active plained above, is integral to optimizing the use of drugs
drug) (Table 1). For example, vancomycin demonstrates in pediatric patients because this approach can address
partial binding to albumin, with a reported range of the wide age spectrum across the diverse pediatric age
30% to 60% in adults. Compared with adults, the de- groups.20 The use and acceptance of PopPK (which
crease in albumin binding evident in pediatric patients integrates nonlinear mixed-effects modeling) continues
results in a significant increase in the unbound van- to improve our understanding of PKPD in children.21
comycin fraction, to approximately 80%.14 In addition, Furthermore, PopPK can be used to individualize drug
a recent study showed that the Vd of clindamycin dosing directly in the patient care setting to improve
in children who are non-obese and children who are the precise achievement of a therapeutic goal that min-
obese was proportional to concentrations of alpha-1- imizes drug toxicity while maximizing clinical benefit.22
acid glycoprotein and albumin because it binds highly Bayesian estimation can be incorporated into
to both proteins.15 PopPK modeling to enhance prediction of PK param-
Plasma membrane transporters mediate the absorp- eters and measures of drug exposure, including drug
tion, distribution, and excretion of drugs. The activities concentrations and area-under-time-concentration
of these transporters are affected by age. Data on curves.23 The use of the Bayesian approach facilitates
the ontogeny of these transporters and their clinical derivation of more accurate predictions for PK, as well
relevance are limited. The two major transporters are as the PD of drugs.24 The Bayesian approach assumes
adenosine triphosphate (ATP)-binding cassette (ABC) that covariates of interest are random and introduces
and the solute carrier (SLC) transporters. In general, uncertainty, which can be better understood with prior
the expression of drug transporters appears low during knowledge of drug behavior in that population. The
the fetal and neonatal periods, but augments from 7 “Bayesian prior” is the construction of the probable
years of age onward.16 Data on age-related maturation PK parameters for a patient, obtained from prior PK
from infancy to early childhood are currently limited. studies in the relevant age-specific population, that can
One of the most studied transporters is ABCB1, be applied to that patient. Combining this Bayesian
which supports renal CL of daptomycin. Interestingly, prior with the patient’s measured drug concentrations,
the development of ABCB1 is also dependent on the the estimation and prediction of PopPK parameters
organ type, with stable expression in the intestines of are greatly enhanced. Population-based PK modeling
adults and pediatric patients, low hepatic expression in with Bayesian estimation is accepted by the US Food
neonates and infants until 12 months of age, and low and Drug Administration as an approach to drug
renal expression until 3 to 6 months of age.16,17 The evaluation in pediatric patients.25
renal CL of daptomycin is higher in neonates and in-
fants than in adults due to the enhancement of ABCB1
expression that mediates renal tubular secretion despite Pharmacodynamics
their immature GFR.16 Evaluation of the PD allows us to understand the rela-
tionship between drug exposure at the site of infection
and the onset, intensity, and duration of pharmacologic
Population-Based Pharmacokinetic effect on the pathogen.26 Along with the PK, the
Modeling and Bayesian Estimation PD property of an antimicrobial agent can be used
While extrapolating pharmacologic data from adults to maximize clinical response and prevent toxicities
may produce meaningful predictions in older chil- related to therapy. To treat or cure an infection, the
dren and adolescents, age-related maturational changes antibiotic exposure should be maximized to inhibit (or
in neonates, infants, young children, and individuals eradicate) the pathogen and suppress the development
with pediatric-predominant diseases (including CF and of resistance.27 The PD exposure target must integrate
Kawasaki disease) require PKPD studies be conducted a feature for pathogen susceptibility known as the min-
directly in these pediatric groups to serve the unmet imum inhibitory concentration, or MIC.28 In addition
needs.18 In the effort to optimize and individualize to the MIC, the drug concentration of interest, which
drug dosing regimens, pharmacometrics is utilized as should reflect the active unbound moiety, can be the
a quantitative approach to understand and predict the peak, trough, or average concentrations or exposure
PK and PD of a drug. Pharmacometric modeling, over a period of time (AUC over 24 hours).
specifically population-based PK (PopPK), allows us to The exposure-response relationship for an antimi-
integrate different factors (or covariates) to ultimately crobial agent is more predictable in adults than in
characterize a population average, with intersubject and pediatric patients. A standard antibiotic dose will gen-
intrasubject variabilities of PK parameters, including erally provide a baseline exposure to elicit a predictable
S112 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

clinical response in adults. However, one standard dose Table 2. Antibiotics With Predictably Good Penetration Into the
cannot be applied in pediatric patients because sub- Central Nervous System
stantial variability exists in exposure to an antibiotic Antibiotic
across the pediatric age spectrum. Once the age-specific
Penicillins, especially aqueous formulation
exposure is known in pediatric patients to achieve an
Aminopenicillins, particularly ampicillin and parenteral (not oral)
equivalent exposure to that demonstrated in adults, amoxicillina
the exposure that yields the desired microbiologic and First- and second-generation cephalosporins (excluding cefuroxime)
clinical cure in adults is then assumed to be similar Third- and fourth-generation cephalosporins (ceftriaxone, cefotaxime,
in that age group. Notably, the age-specific exposure ceftazidime, cefixime, and cefepime)b
Carbapenems, including meropenem and imipenemb
in pediatric patients, particularly in neonates due to
Aztreonam
their immature immune function, may differ from the Chloramphenicol
exposure needed to achieve microbiologic and clin- Sulfamethoxazole/trimethoprim
ical cure in adults; in other words, the drug expo- Fosfomycin
sure required in the immune-compromised neonate for Metronidazole
Rifampin
cure may be greater than that associated with cure in
Antituberculosis agents (ie, isoniazid and pyrazinamide)b
adults. Currently, no prospective data exist in neonates Daptomycin
to validate the exposures required for microbiologic Fluoroquinolones
and clinical cure. In addition, underlying genetic and Tetracyclines
clinical factors that may affect response may also be a
Nafcillin and piperacillin have been studied in pediatric patients but penetra-
explored to optimize therapy in both pediatric patients tion is erratic.
and adults. b
Studies have documented penetration specifically in children.
An important component of PD for successful treat-
ment of an infection is antibiotic penetration. The
degree of antibiotic penetration must be accounted for For pneumonia caused by extracellular pathogens,
to ensure adequate drug exposure at the site of infection epithelial lining fluid (ELF) has been used as the target
to maximize therapeutic response. The MIC provided site to determine adequacy of drug exposure. Currently,
by clinical microbiology depicts susceptibility of a the literature on ELF-to-plasma ratios is limited to a
pathogen based on achievable serum concentration. In small number of antibiotics, all of which were evaluated
the scenario where a pathogen is reported resistant in adults, and comprises beta-lactams (ranging from
to a certain antibiotic, that same antibiotic may still 0.21 to 1.04, depending on the specific beta-lactam),
successfully treat the infection if the resistance (assessed vancomycin (0.18 to 0.50), aminoglycosides (highly
by the actual MIC) is not profound and complete and variable hysteresis depending on sampling time and
adequate drug exposure can be achieved (eg, treating a arising from the hydrophilic property of these antibi-
urinary tract infection using a drug that highly concen- otics), and fluoroquinolones (>1.0, which represents
trates in the kidneys). In contrast, even if an antibiotic extensive lung penetration).30 The presence of hystere-
is reported as susceptible, therapeutic response may sis on a concentration-effect plot implies a delay in time
not be achieved if drug exposure is inadequate in between measured concentration and response, as well
specific tissue sites, particularly for serious infections, as an associated lower peak concentration. Notably,
including meningitis and pneumonia, or undrained these studies of ELF do not account for redistribution
abscesses. of drugs from the ELF back to plasma because only
Appropriate management of central nervous system specific time points after dose administration (rather
(CNS) infections requires that antibiotics penetrate the than over the entire dosing interval) were evaluated.
blood–brain barrier to attain concentrations at the site Nonetheless, for all of these distinct infection types,
of infection for eradication of the infecting pathogen. adequate antibiotic exposure implies the need to use
Independent of age, the physicochemical property of free drug concentrations in PKPD studies to account
a drug is the main factor that determines drug distri- for antibiotic penetration at the site of infection.
bution into the CNS. In fact, small-sized and highly
lipophilic drugs with low protein binding penetrate the Monte Carlo Simulation
blood–brain barrier well. The distribution pathways To identify the best dosing regimen that achieves
that are affected by growth and development, and the microbiologic and clinical cure (and even resistance
presence of meningeal inflammation, are also factors suppression), a PD exposure target or index should be
that affect overall CNS drug penetration. Antibiotics explicitly defined and incorporated (along with age-
with good CNS penetration are provided in Table 2. 29 specific PK data) to conduct Monte Carlo simulations.
Penetration of these antibiotics is enhanced when the The PD indices that vary with antibiotic class must
meninges are inflamed. integrate pathogen susceptibility and antibiotic
Le and Bradley S113

exposure, depicted by free drug peak concentration over Beta-lactams


MIC (Cmax /MIC, associated with aminoglycosides), Because of their long-standing safety profile and their
AUC24 /MIC associated with fluoroquinolones, or broad spectrum of antibacterial activity, beta-lactam
the time that free drug concentrations remain above antibiotics present the most extensively studied class
the MIC (fT>MIC), associated with beta-lactams. evaluated for their PKPD properties in pediatric pa-
Currently, the PD exposure targets for antibiotics have tients. Because this class of antibiotics exhibits time-
originated primarily from preclinical in vitro models, dependent bactericidal but no post-antibiotic effects,
with very limited validation in clinical studies.27 Fur- the use of beta-lactams can be optimized by increasing
thermore, these limited human clinical investigations the infusion time or the dosing frequency, particularly
were conducted retrospectively in adult patients. As to treat pathogens with higher MICs (Table 3). These
such, there is an evident demand for well-designed two approaches align with maximal use of the time-
studies in pediatric participants, preferably conducted dependent PD killing of beta-lactams. Nonetheless,
prospectively, to validate PD targets of antimicrobial extending the infusion time (which includes prolonged
agents that were derived from preclinical and adult and continuous infusions) has been evaluated more
data. Despite improvement in the number of pediatric frequently than has increasing dosing frequency (ie,
PK studies in recent decades, the need for PD studies in from every 6 to every 4 hours) for its feasible application
pediatric participants should be underscored to ensure in the patient care setting. This is particularly valuable
the optimization of antibiotic dosing required for the in the care of pediatric patients with certain medi-
cure of different infections over the entire pediatric cal conditions (eg, prematurity, CF, or hematologic/
age spectrum. In addition, protein binding, drug oncologic disorders) that necessitate the use of multiple
penetration to the site of infection, and immunologic concurrent medications.
competence that changes with age are important To determine the probability of target attainment
factors that should be evaluated in PD studies. (PTA) against Pseudomonas aeruginosa with varying
susceptibilities, Monte Carlo analyses in children
between 1 and 12 years of age were conducted
Clinical Pharmacology Optimizing using different regimens of cefepime, ceftazidime,
Bedside Care of Pediatric Infections imipenem/cilastatin, meropenem, and piperacillin/
Safe and effective antibiotic use in all pediatric age tazobactam.33–35 The PD target to achieve bactericidal
groups, including neonates, requires thorough under- activity used in these studies was fT>MIC 40%
standing and application of the quantitative clinical for carbapenems and 50% for penicillins and
pharmacology that encompasses PKPD.20 Pharmacol- cephalosporins. For bacteriostatic effect, PD targets
ogists, pharmacists, and physicians must collectively be 20% fT>MIC for carbapenems and 30% fT>MIC
equipped to support and integrate PopPK modeling, for penicillins and cephalosporins were used. Collec-
Bayesian estimation, and simulation methods in the tively from these studies, prolongation of infusion from
clinical decision-making process that is integral to ther- 30 minutes to 3 hours and continuous infusion signifi-
apeutic drug monitoring and management to optimize cantly improved the PTA to >90% to achieve bacterici-
exposure-response and clinical outcome relationships dal activity for all antibiotics that were evaluated.33–35
across the pediatric age spectrum.18 This is a clinically relevant finding that can be applied
To demonstrate the application of clinical pharma- to critically ill children to achieve optimal PTA against
cology to the optimization of pharmacotherapy and moderately susceptible strains of P aeruginosa.
its positive impact on the care of pediatric patients For less susceptible pathogens, increasing the dose
with infections, specific antibacterial agents (including alone, without optimizing the PD property of fT>MIC
beta-lactams, aminoglycosides, and vancomycin) are by increasing infusion time, may not always achieve
presented in the subsequent sections. The selection adequate drug exposure. For example, at one institution
of these antibiotics was based on the availability of with elevated MICs among the P aeruginosa isolates,
studies in pediatric patients and on their PD prop- increasing the meropenem dose from 20 mg/kg to 40
erties. Antibiotics with unique PD features (including mg/kg, administered every 8 hours using 30-minute
concentration-dependent, time-dependent, and post- infusions, resulted in a low, inadequate PTA of 58%.35
antibiotic effects) and, more importantly, antibiotics This illustrates that increasing the dose alone without
with a sufficient number of PKPD evaluations in pe- extending the infusion time of beta-lactams may not be
diatric patients are presented. Table 3 describes the PD sufficient to adequately treat serious antibiotic-resistant
properties of antibiotics, including those with limited infections.
PKPD studies in pediatric patients. Antifungal agents Emerging interest in defining age-related devel-
were excluded from this article, but two reviews have opmental exposures has prompted population-based
been published.31,32 PKPD evaluations in preterm neonates.36–41 To
S114 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

Table 3. Pharmacodynamic Properties of Antimicrobial Classes in Pediatric Patients104

Antibiotic Class Mechanism Index Dose Optimization

Aminoglycosides Concentration-dependent, with Cmax /MIC, AUC24 /MIC Increase dose to maximize Cmax or
moderate post-antibiotic effect AUC24
β-lactams Time-dependent, with no to minimal fT>MIC Increase duration of infusion or
post-antibiotic effect frequency of administration
Amphenicols (chloramphenicol) Time-dependent, with some AUC24 /MIC Increase dose, frequency of
post-antibiotic effect against administration, or duration of infusion
Gram-positive bacteria
Fluoroquinolones and quinolones Concentration-dependent, with Cmax /MIC, AUC24 /MIC Increase dose to maximize Cmax or
moderate post-antibiotic effect AUC24
Lincosamides (clindamycin) Time-dependent, with moderate to AUC24 /MIC Increase dose, frequency of
prolonged post-antibiotic effect administration, or duration of infusion
Macrolides and azalides Time-dependent, with moderate to AUC24 /MIC Increase dose, frequency of
prolonged post-antibiotic effect administration, or duration of infusion
Nitroimidazoles (metronidazole) Concentration-dependent, with AUC24 /MIC Increase dose to maximize Cmax or
moderate post-antibiotic effect AUC24
Nitrofurantoin Time-dependent for E coli; fT>MIC, AUC24 /MIC Increase dose, frequency of
concentration-dependent for S administration, or duration of infusion
saprophyticus and E faecium
Oxazolidinones (linezolid) Time-dependent, with moderate to AUC24 /MIC, fT>MIC Increase dose, frequency of
prolonged post-antibiotic effect administration, or duration of infusion
Rifamycins Concentration-dependent, with Cmax /MIC, AUC24 /MIC Increase dose to maximize Cmax or
moderate post-antibiotic effect at ³ 16 AUC24
× MIC
Sulfonamides and trimethoprim Concentration-dependent, with Cmax /MIC, AUC24 /MIC Increase dose to maximize Cmax or
moderate post-antibiotic effect AUC24
Tetracyclines and glycylcyclines Time-dependent, with moderate to AUC24 /MIC Increase dose, frequency of
(tigecycline) prolonged post-antibiotic effect administration, or duration of infusion
Glycopeptides Time-dependent, with moderate to AUC24 /MIC Increase dose, frequency of
prolonged post-antibiotic effect administration, or duration of infusion

Abbreviations: AUC24, area under the concentration–time curve over a 24-hour period; Cmax , maximum serum concentration; MIC, minimum inhibitory
concentration; fT>MIC, fraction or percentage of the dosing interval above the MIC.

adequately design dosing strategies that reflect The PKPD of meropenem was evaluated for the
maturational development, including nephrogenesis, treatment of meningitis and intra-abdominal infec-
both gestational and postnatal ages (resulting in tions in neonates.39 For meningitis, meropenem con-
postmenstrual age) were used in these studies. To centrations in the cerebrospinal fluid (CSF) that were
achieve antibiotic microbiologic and clinical efficacy measured at various time periods varied from 4.1 to
and account for deficiencies in the immune response 34.6 µg/mL, which represent an average of 70% (range
system expected in neonates, higher PD targets of of 5% to 148%) penetration. For suspected or compli-
>60% fT>MIC for meropenem and >75% fT>MIC cated intra-abdominal infections, meropenem 20 to 30
for ampicillin and piperacillin were employed in these mg/kg, depending on gestational age and postnatal age,
studies. was administered every 8 or 12 hours. Most (85% of
Meropenem was evaluated in preterm and full- 200) of these infants experienced clinical therapeutic
term neonates <2 months old with gestational ages success and tolerated meropenem well.38,39
of 23 to 42 weeks.36,41 Important covariates for CL Similar to meropenem, piperacillin/tazobactam was
were postmenstrual age, weight, and serum creatinine. also studied in premature and term infants 90 days
Meropenem CL increased in older neonates, defined with suspected or complicated intra-abdominal infec-
by gestational age and chronologic age. Meropenem tions. The CL of piperacillin improved with increasing
20 mg/kg every 8 hours, infused over 30 min- gestational age at birth, but decreased by 60% when
utes, achieved >90% PTA for its bactericidal effect serum creatinine was 1.2 mg/dL.42 Piperacillin 80
against nosocomial Gram-negative pathogens.36 How- to 100 mg/kg infused every 8 hours did not meet
ever, meropenem 40 mg/kg every 8 hours infused over PD efficacy target for P aeruginosa in 70% of in-
4 hours was recommended for less susceptible bac- fants, implying that further studies of higher doses
teria, including P aeruginosa, with MIC of 4 to 8 or more frequent or more prolonged infusions are
mcg/mL.36,41 warranted.42
Le and Bradley S115

A phase 1, single-dose study of doripenem was ill.33,48 This increased Vd may potentially result from
conducted in 52 infants up to 12 weeks of postnatal underlying disease states affecting the interstitial fluid,
age.43 Displaying linear PK, neonates <4 weeks had where increased extracellular fluids observed in third-
decreased CL and increased mean AUC exposure. The fluid spacing increases the Vd.
PK of doripenem infused at 5 to 8 mg/kg over 1 hour The optimal PD target for immune-compromised
in these term and preterm infants was similar to those neonates and children is not well-defined. However,
previously reported for other carbapenems in neonates one investigator has proposed an aggressive and un-
and young infants. validated PD target to combat serious P aeruginosa
Similar to premature infants, children with CF have infections in patients with immune-compromised sta-
altered PK that may necessitate different dosing strate- tus and maintain free drug concentrations at 100%
gies. Increased CL (eg, aztreonam 100 mL/min in chil- fT >6 times the MIC.49,50 Studies in adults have
dren with CF versus 76 mL/min in healthy participants, evaluated a PD target of 100% fT>MIC to im-
P < .01), without a significant change in Vd, has been prove clinical cure and microbiologic eradication
reported.4,44 To prevent subtherapeutic dosing that may using cefepime, piperacillin/tazobactam, meropenem,
result from the increased CL, the PD property of an- and ceftobiprole.51,52 Using this PD target in ado-
tibiotics should be maximized to optimize dosing in this lescents with febrile neutropenia, a loading dose
population. One study showed significant improvement of 100 mg/kg followed by continuous infusion of
in achieving the PTA of 65% fT>MIC with the piperacillin/tazobactam at 350 to 400 mg/kg/day
use of 4- to 5-hour prolonged or continuous infusions achieved >99% PTA at an MIC of 4 mg/L.49 The
of ceftazidime 6 grams per day (weight-adjusted for standard dose of 300 mg/kg/day achieved 85% PTA,
70 kg). Extended or continuous infusions increased the which the authors considered inappropriate for pa-
PTA for pathogens with elevated MIC, even as high tients with recent intensive chemotherapy, predicted
as 8 to 12 μg/mL.4 This is invaluable in patients with prolonged and profound neutropenia, or fever >39°C
CF because they usually have a history of previous because such patients are at increased risk for serious
exposure to antibiotics for both acute and chronic infections.
respiratory infections.45,46 Likely due to their repeated In conclusion, studies in the different pediatric age
antibiotic exposures, reported antibiotic resistance rates groups serve as evidence for the use of various dosing
are higher in children with CF. strategies for beta-lactams to enhance their PD target
Ceftaroline is the first beta-lactam antibiotic with attainment. Strategies for the optimization of beta-
activity against methicillin-resistant Staphylococcus au- lactam use consist of frequent dosing and extended
reus (MRSA) and, as such, may play a significant role in or continuous infusions that may improve PTA in
patients with CF. A case report of a ceftaroline-resistant certain pediatric populations, including individuals
MRSA strain from a girl with CF was reported after who are critically ill, immunocompromised (including
22 ceftaroline treatment courses using standard adult premature neonates), have CF, or are infected with
dosing that achieved suboptimal antibiotic exposure, pathogens exhibiting high MICs. Outside these specific
possibly arising from increased CL and Vd in this medical conditions, the PD-guided dosing strategies,
patient. This case report prompted a study evaluating as compared to traditional dosing, may not provide
the single-dose PK and safety of ceftaroline in children additional clinical benefit to patients. Based on a
with CF for staphylococcal pneumonia. In this phase systematic review of limited studies conducted in
1, open-label, single-dose, prospective study using pediatric patients, the routine use of extended or
10 mg/kg (up to 600 mg) in 20 participants between 6 continuous infusion of beta-lactam antibiotics was not
and 18 years of age, PopPK modeling showed that chil- supported by current evidence, as excessive treatment
dren with CF have increased ceftaroline CL compared failures have not been reported.53 Nonetheless, more
with published data from children without CF. As such, well-designed prospective trials in pediatric patients,
higher dosages are likely to be required in children with particularly those with underlying diseases that may
CF to achieve adequate exposure for cure and possibly benefit from PD-guided dosing strategies, are necessary
delay the emergence of resistance.47 to confirm the clinical value of these dosing strategies
In addition to neonates and children with CF, criti- to directly affect patient care.
cally ill children may have experienced alteration in PK
parameters necessitating changes in dosing strategies. Aminoglycosides
Increased Vd of certain antibiotics, particularly those Based on studies in adults using traditional multiple-
with high water-solubility, has been reported in criti- daily dosing of tobramycin and gentamicin, clinical
cally ill children. For example, the Vd of piperacillin response to these antibiotics is maximized when the
is 0.51 L/kg in critically ill young children versus Cmax /MIC ratio is 8 to 10 (Table 3).54,55 This traditional
0.28 L/kg in young children who are not critically dosing has been associated with decreased resistance
S116 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

patterns, but suppression of P aeruginosa requires a for these reasons, extended-interval dosing provides
much higher Cmax /MIC target of 30.56,57 To optimize a rational strategy to employ in these patients to
the aminoglycosides’ PD features and potentially maximize clinical benefit.
minimize adaptive resistance, the once-daily (or Pharmacodynamically, once-daily dosing can
extended-interval) dosing strategy consolidates multi- achieve antibiotic exposures that are effective
ple doses into one large daily dose to achieve high peak against less susceptible bacteria. In fact, tobramycin
concentrations to effectively eradicate certain bacteria, 10 mg/kg/day (which achieves Cmax of 20 to 40 μg/mL,
including those with reduced susceptibility and an trough concentrations <1 μg/mL, and AUC24 of 60
MIC >1 μg/mL.58,59 With their post-antibiotic effect, to 120 mg-h/L) and once-daily amikacin 30 to 35
once-daily aminoglycoside dosing provides a drug-free mg/kg/day (which achieves Cmax of 80 to 120 μg/mL,
interval where serum concentrations can fall below the trough concentrations <1 μg/mL, and AUC24 of 235 ±
MIC of susceptible organisms, yet retain inhibition of 110 mg-h/L) can reach the PD index of Cmax /MIC 10
bacterial growth. As an additional clinical benefit, the for less susceptible pathogens, specifically those with
extended-interval strategy may reduce nephrotoxicity MICs up to 2 μg/mL for tobramycin and 4 μg/mL for
because it allots sufficient time for complete elimination amikacin.57 Because of the PD property of aminogly-
of the drug prior to re-administration.59,60 While cosides, the routine use of extended-interval dosing is
Cmax /MIC is the PD index used for aminoglycoside supported by the Cystic Fibrosis Foundation.63
efficacy, the clinical significance of AUC monitoring re- Gentamicin is bactericidal against Gram-negative
quires further exploration. Current dose-fractionation pathogens and has been used widely in neonates for
studies reveal that AUC/MIC may be correlated to the decades. Multiple elements affect the PK of gentamicin
development of P aeruginosa resistance.55,56 in neonates, including postconceptional age, weight,
The efficacy and safety of extended-interval dosing and renal function. The PD index usually targeted in
was compared to traditional thrice-daily dosing of this population is Cmax of 5 to 10 μg/mL for efficacy
tobramycin at 10 or 15 mg/kg/day in a meta-analysis (assuming MIC 1 μg/mL) and trough concentrations
of four randomized, controlled studies that evaluated <1 to 2 μg/mL to minimize toxicity.60 The high variabil-
both pediatric and adult participants with CF.61 No ity and unpredictability of gentamicin’s PK reinforces
significant differences in clinical outcomes (including the value of individualized dosing and therapeutic
pulmonary function, nutritional status, time to first drug monitoring, particularly in premature neonates.64
pulmonary exacerbation after treatment course, and In fact, a multicenter study demonstrated the rapid
ototoxicity) were reported between these dosing strate- achievement of therapeutic concentrations using an
gies. However, the advantage of the extended-interval individualized nomogram-based dosing strategy, with
dosing strategy was decreased nephrotoxicity, defined less need for measuring drug concentrations and dosing
by serum creatinine and urinary renal biomarkers. adjustments.65
Gentamicin, tobramycin and, to some extent, With an evidently prolonged half-life due largely to
amikacin have been thoroughly examined in pediatric the neonate’s immature renal function and, to a lesser
patients. While tobramycin and amikacin are the pri- extent, increased Vd, once- daily dosing is reasonable
mary aminoglycosides used to treat pulmonary infec- and, especially in extreme preemies, dosing every 36 to
tions caused by P aeruginosa in CF, gentamicin has 48 hours may be necessary.62 Based on a comparative
been more widely used for decades for the treatment of meta-analysis of 574 neonates who were <28 days of
suspected or proven bacterial sepsis caused by enteric life, once-daily dosing, as compared with multiple-daily
bacilli in neonates. Currently, this PD-guided, once- dosing, improved attainment of peak concentrations
daily strategy is standard of care for adults and has 5 μg/mL and trough concentrations <2 μg/mL.60
been evaluated in children with CF, children with febrile Efficacy and toxicity, including ototoxicity and nephro-
neutropenia, and neonates.55,57,60–62 toxicity (assessed by changes in serum creatinine and
Children with CF commonly receive multiple renal biomarkers) were comparable between these dos-
courses of aminoglycoside therapy because they ing strategies. Accordingly, neonates who receive once-
have pulmonary infections caused by P aeruginosa daily gentamicin dosing and for a duration that is
throughout their lifetime. As a result, the infecting less than 48 to 72 hours may not routinely require
bacteria often have decreased antibiotic susceptibility the therapeutic monitoring of drug concentrations or
owing to frequent previous antibiotic exposure.57 assessments of renal function for toxicity.
Furthermore, adequate antibiotic concentrations are Children may experience febrile neutropenia as
difficult to achieve in the ELF and mucosal surfaces a result of cancer chemotherapy or transplantation
in these infected children. In addition, patients with immunosuppressive therapy. Depending on patient
CF tend to have increased drug Vd and CL, which age, gentamicin or tobramycin administered at 6 to
necessitates higher antibiotic doses. Collectively, 10.5 mg/kg/day achieves Cmax /MIC 10, which is
Le and Bradley S117

necessary to treat infections caused by P aeruginosa differs from adult response and adult data may not
in these children.66–68 Age-related differences in renal always be applicable. Furthermore, there are insuffi-
CL necessitate a higher milligram per kilogram dose of cient PD data to demonstrate improved outcomes with
gentamicin for younger children with febrile neutrope- attainment of vancomycin AUC/MIC 400 in pediatric
nia. In addition, once-daily administration is preferred patients and the data come primarily from retrospective
to thrice-daily administration to maximize efficacy analyses.76–79 Prospective, comparative PD studies in
and minimize nephrotoxicity and has the concomitant pediatric patients are necessary to validate the PD
benefit of less nursing time for drug administration. target that so far has been evaluated only in adults.
With minimal post-antibiotic leukocyte enhancement Optimal vancomycin use requires incorporating
because of their neutropenia, loading doses to achieve both age-related PK developmental changes and MIC
therapeutic peak concentrations early in the course of data. PK studies in pediatric participants suggest lower
therapy may be advantageous in these patients. trough concentrations will achieve AUC/MIC 400
Aminoglycosides may cause a concerning adverse but PD outcomes data are limited by inconclusive
effect that limits their use, nephrotoxicity. Prospective retrospective studies. In the largest PopPK study in
randomized trials have documented the advantage of pediatric participants >3 months (N = 702), the inte-
once-daily over multiple-daily dosing in reducing the gration of weight, age, and serum creatinine (which,
occurrence of nephrotoxicity.55,69,70 In proximal renal in aggregate, reflects developmental renal maturation)
tubular epithelial cells, aminoglycosides can accumulate was necessary to estimate vancomycin CL.75 Using
and thereby result in nephrotoxicity. Once-daily dosing AUC/MIC 400 as the PD target index due to lack of
exposes the renal cells to high, intermittent concen- current pediatric studies, increased vancomycin doses at
trations that saturate drug uptake and consequently 60 to 80 mg/kg/day (depending on age, serum creatinine,
lead to an increase in drug excretion.55,60 Further- and MIC by Etest) was necessary to achieve adequate
more, once-daily dosing facilitates sufficient time for PTA. Notably, vancomycin AUC/MIC, compared with
elimination of the drug to negligible concentrations trough concentrations, was a more achievable and,
before the subsequent dose is administered, thereby hence, realistic target in children. Higher doses of 90 to
preventing nephrotoxicity. In a study of neonates, once- 100 produced excessively high AUCs of 840 to 940 mg-
daily gentamicin dosing resulted in reduction in the h/L, with trough concentrations exceeding 20 μg/mL.75
ratio of urinary N-acetyl-βD-glucosaminidase to cre- This excessive exposure may potentially place patients
atinine, which appears to be a sensitive marker for at unnecessary and increased risk of nephrotoxicity, as
nephrotoxicity.69 concluded by multiple studies in children with trough
The other significant adverse effect of amingoglyco- concentrations 15 μg/mL, AUC >800 mg-h/L, and
sides is ototoxicity, in which free radicals irreversibly doses 10 mg/kg.80–82
damage the cochlear and vestibular hair cells. The Because vancomycin is eliminated primarily via
occurrence of ototoxicity has not been associated with GFR with some active tubular secretion, dosing at
frequency of aminoglycoside administration.23,55 15 mg/kg every 8 hours (ie, 45 mg/kg/day) may be most
appropriate empirically for older children with acute re-
Vancomycin nal insufficiency.83,84 Furthermore, children with acute
Vancomycin exhibits time-dependent bactericidal activ- renal insufficiency generally do not have chronic disease
ity, with some post-antibiotic effect lasting 0.7 to 2.6 and, hence, may require close monitoring because re-
hours for Staphylococcus aureus and 4.3 to 6.5 hours covery of renal function may occur during vancomycin
for Staphylococcus epidermidis.71 Based on adult PKPD therapy, with markedly improved CL.
studies, the efficacy surrogate for vancomycin in treat- Population-based PK studies with Bayesian estima-
ing invasive MRSA infections is achieving AUC/MIC tion in preterm and full-term neonates have evaluated

ratio 400, with the MIC determined by Etest
R
vancomycin doses ranging from 15 to 60 mg/kg/day
(Biomérieux; Marcy-l’Étoile, France). An AUC/MIC administered intermittently every 8 to 24 hours or as
ratio 400 corresponds to a minimum concentration continuous infusions.85–88 Notably, these studies did
of 15 to 20 μg/mL when the MIC is 1 μg/mL in not evaluate PTA by AUC/MIC 400, which is the PD
adults.72,73 However, in infants and children >3 months target best linked to successful treatment outcomes in
of age, this PD index may be achieved with lower adults. However, one study evaluated the association
trough concentrations of 8 to 10 μg/mL when the between AUC and trough concentrations in 240
MIC is 1 μg/mL (evaluated using broth microdilution neonates, with 1702 vancomycin concentrations.89 This
and Etest methods).74,75 This crucial observation was PopPK study derived a model for vancomycin CL that
based on two independent PK studies that incorporated was based on weight, postmenstrual age, and serum cre-
robust Bayesian estimation and Monte Carlo simu- atinine; using Monte Carlo simulations with Bayesian
lations, emphasizing the fact that pediatric response estimation, trough concentrations ranging from 7 to
S118 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

11 μg/mL were highly predictive of an AUC24 of >400 in the pediatric setting remains unacceptably high. Fur-
μg-h/mL in at least 90% of neonates. Doses to achieve thermore, the integration of PKPD into clinical prac-
this PKPD target ranged from 15 to 20 mg/kg every 8 to tice still needs improvement. The link between in vitro
12 hours, depending on postmenstrual age and serum and in vivo PKPD properties and their extrapolation to
creatinine. The incidence of vancomycin-associated the clinical use of drugs in pediatric patients needs to
nephrotoxicity reported in neonates has been low, be communicated clearly to clinicians in a manner that
ranging from 1% to 9%.90 The high variability of changes practice.26 One approach may be to address the
vancomycin’s PK in preterm and full-term neonates quality of PKPD studies by developing an evidence-
necessitates therapeutic drug monitoring, preferably based grading scheme that enables clinicians, many
with AUC estimation, to ensure adequate therapeutic of whom are not pharmacologists, to appreciate the
exposure.29 strength of the evidence.21 In addition, new evidence in
Recent PK data highlighting differences in trough pediatric patients, including those who are obese, often
concentrations required to achieve the PD vancomycin emerges post-marketing and should be integrated into
exposure target of AUC/MIC 400 in children and clinical practice by updating drug formularies.
adults prompts the need to investigate the optimal Despite the successful use of allometric weight in
monitoring strategy that can be integrated into clinical pediatric patients to estimate PK parameters, errors
practice.74,75 The minimal PD evidence in children, may still occur, especially for drugs undergoing hepatic
coupled with the differences in trough concentrations, metabolic CL. Physiologically based pharmacokinetic
supports the monitoring of vancomycin exposure by models are emerging to facilitate simulations of PK
AUC because it is a more relevant and achievable profiles under varying physiological conditions.94 These
PD target. Furthermore, AUC targeting and moni- models integrate data that are traditionally not in-
toring may prevent excessive dosing, frequent dosing tegrated into PK modeling, including drug proper-
adjustments, and potentially adverse effects. A PopPK ties, physiological changes, and biological parameters,
study with Bayesian estimation in 138 pediatric par- which may differ between individuals due to age and
ticipants demonstrated improved the accuracy and disease states. The biological and mechanistic basis of
precision of estimating AUC using peak and trough PBPK models closes the gap between tissue concen-
concentrations compared with trough concentrations trations and observed therapeutic or toxic effects. A
alone.91 However, one study in adults suggests that a unique approach that combines the full PBPK and stan-
single sample may be adequate to predict appropriate dard PopPK analyses, which has been termed as “semi-
dosing.92 physiological-based PK,” has been used to characterize
and predict the developmental changes of important
elimination pathways, like GFR, across the human life
Current Challenges With Potential span, from neonates to adults. This system-based phar-
Solutions macometric methodology permits the development of
Antimicrobial agents are widely used in pediatric pa- one model that simultaneously describes multiple drugs
tients, especially neonates and young infants. Despite (eg, aminoglycosides and vancomycin) because they
extensive use of antibiotics, the PKPD in this popu- undergo similar elimination.95
lation are limited, arising from its historical exclusion The ethics and logistics of blood sampling (both
from clinical trials. Legislation via the Best Pharma- by number and volume) in pediatric patients present
ceuticals for Children Act (BPCA) and the Pediatric a challenge to conducting PKPD studies with enough
Research Equity Act (PREA) support and mandate pe- samples to accurately estimate PK parameters. Tra-
diatric research to improve drug product labeling.93 The ditionally, a whole blood volume up to 5 mL and
PREA legislation authorizes the National Institutes sampling 5 to 8 times within a dosing interval were
of Health to implement research programs through necessary to accurately describe PK parameters in
funding clinical trials to study off-patent drugs in all pediatric patients. Innovative strategies to overcome
children, including neonates and young infants. BPCA this challenge have been attempted and include sparse
provides economic incentives to companies in exchange sampling, microsampling, dried blood spot technique,
for important clinical data that address an important and opportunistic study design.
medical need in pediatric patients. Continued efforts are Sparse sampling is more practical than the tradi-
necessary to promote the need for PKPD evaluations of tional intensive approach and has been increasingly
drugs (both generic and new) across the broad pediatric appearing in published PK studies evaluating pediatric
age spectrum. populations.40,42,75,91,96 Sparse sampling provides one
While quantitative pharmacology is increasingly em- major advantage: it reduces the number of blood draws
ployed to evaluate the PKPD of drugs in pediatric and, when applied in population-based modeling, pro-
patients, the off-label use of drugs without PKPD data vides estimation for both individual and population
Le and Bradley S119

PK parameters with inter-subject, intra-subject, and sampling in providing definitive dosing recommenda-
“unexplained” residual variabilities.21 To accommodate tions may be drug dependent and needs to be further
the reduced number of blood samples, a larger partic- explored.42 Opportunistic studies have been used to
ipant sample size (often >100) must be used to pro- extract important findings for the preliminary construct
duce meaningful results in PopPK modeling. Similar of phase 1 through 3 trials and enhance the dosing
to sparse sampling, microsampling reduces the overall of currently marketed antimicrobial agents in pediatric
blood volume needed: whereas sparse sampling reduces populations.75,101,102
the frequency, microsampling reduces the actual vol-
ume drawn during sampling. In fact, microsampling Conclusions
requires a volume <50 μL, which can be collected by
Children of all ages may experience variability in
skin prick.97
response to antimicrobial agents, thereby potentially
Dried blood spot is a sampling technique that utilizes
leading to undesired subtherapeutic or unanticipated
an ultra-low volume (ie, 30 μL of whole blood, which
toxic effects when PKPD data are unavailable. This
is 20 times lower than traditional venous or arterial
underscores the importance of thoroughly understand-
samples) to evaluate the PK of drugs. It is stable and
ing the age-related physiologic changes that occur with
reproducible on paper filters. In addition, dried blood
normal human growth and development. Regulatory
spot sampling eliminates the need for centrifugation or
agencies through PREA are requiring PK and efficacy
freezing of samples and measurement of drug concen-
data to optimize pharmacotherapy in pediatric pop-
tration in whole blood. As such, the advantages of dried
ulations, including neonates. While the availability of
blood spot sampling are low sample volume, minimal
PK data in these populations has improved over the
personnel training, no sample processing, room tem-
past decades, PKPD outcome assessments remain in-
perature storage, and simple bioanalytical analysis.98
finitesimal. Rational antibiotic dosing can only improve
Using this technique, the estimates and precision of
with more well-designed PKPD pediatric studies in the
metronidazole PK parameters for premature infants in
future.
one small study were similar between plasma and dried
blood spot samples.99 Of concern though, metronida-
zole concentration in the dried blood spot samples was Author Disclosure
15% lower than in the plasma samples. Limited pedi- Drs Bradley and Le have no personal financial involve-
atric PK studies currently utilize this technique.99,100 ment with any company that creates, develops, markets, or
Future studies should explore this sampling strat- sells antimicrobial agents. Their employer, the University of
egy and identify class- and drug-specific concentra- California, has contracts with many companies to develop
tion differences between dried blood spot and plasma and test antimicrobial agents. Dr Le has received research
samples. support from NICHD and NIAID (1U54HD090259 and
The opportunistic study design capitalizes on stan- 1K23AI089978) and subagreement from Duke University
dard clinical care to perform clinical research. This (HHSN27500027). Dr Bradley has received research support
design is perceived as minimal risk, diminishes research- from NICHD (1U54HD090259).
related costs, and enhances parental consent. While the
opportunistic design does not permit administration
of study drug to patients because the drug is already
prescribed as standard of care, it provides four main
References
advantages and therefore is attractive to use in pediatric 1. Bradley JS, Garonzik SM, Forrest A, Bhavnani SM. Pharma-
research. First, this design evaluates a drug that is cokinetics, pharmacodynamics, and Monte Carlo simulation:
selecting the best antimicrobial dose to treat an infection.
already prescribed by clinical indication. Second, it Pediatr Infect Dis J. 2010;29(11):1043–1046.
optimizes the collection for PK samples by coinciding 2. Anderson BJ, Holford NH. Understanding dosing: children are
timing with routine labs to minimize pricking. Third, small adults, neonates are immature children. Arch Dis Child.
for drugs with active therapeutic drug monitoring, data 2013;98(9):737–744.
on drug concentrations are already measured by the 3. Anderson BJ, Holford NH. Mechanism-based concepts of
size and maturity in pharmacokinetics. Annu Rev Pharmacol
clinical laboratory and available for use in PK mod- Toxicol. 2008;48:303–332.
eling. Finally, the opportunistic study design utilizes 4. Bulitta JB, Landersdorfer CB, Huttner SJ, et al. Population
scavenged samples to maximize testing on drawn blood pharmacokinetic comparison and pharmacodynamic break-
that would otherwise be left unused. points of ceftazidime in cystic fibrosis patients and healthy
Scavenged PK sampling is a minimal-risk approach volunteers. Antimicrob Agents Chemother. 2010;54(3):1275–
1282.
that can provide meaningful information related to 5. Mahmood I. Prediction of drug clearance in children: impact of
the development of PK models but cannot inform allometric exponents, body weight, and age. Ther Drug Monit.
dosing recommendations. The utility of scavenged 2007;29(3):271–278.
S120 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

6. Natale S, Bradley J, Nguyen WH, et al. Pediatric obesity: phar- and Medical Informatics. Advanced Information and Knowledge
macokinetic alterations and effects on antimicrobial dosing. Processing. London, England: Springer-Verlag; 2005:351–369.
Pharmacotherapy. 2017;37(3):361–378. 25. Food and Drug Administration. Guidance for industry popula-
7. Rhodin MM, Anderson BJ, Peters AM, et al. Human renal tion pharmacokinetics. https://www.fda.gov/downloads/Drugs/
function maturation: a quantitative description using weight .../Guidances/UCM072137.pdf. Accessed October 3, 2017.
and postmenstrual age. Pediatr Nephrol. 2009;24(1):67–76. 26. Gabrielsson J, Green AR. Quantitative pharmacology or phar-
8. Mahmood I. Prediction of drug clearance in children from macokinetic pharmacodynamic integration should be a vital
adults: a comparison of several allometric methods. Br J Clin component in integrative pharmacology. J Pharmacol Exp
Pharmacol. 2006;61(5):545–557. Ther. 2009;331(3):767–774.
9. Nielsen EI, Sandstrom M, Honore PH, Ewald U, Friberg LE. 27. Drusano GL. Pharmacokinetics and pharmacodynamics of
Developmental pharmacokinetics of gentamicin in preterm and antimicrobials. Clin Infect Dis. 2007;45(1 suppl):S89–S95.
term neonates: population modelling of a prospective study. 28. Michalopoulos AS, Falagas ME. Colistin: recent data on
Clin Pharmacokinet. 2009;48(4):253–263. pharmacodynamics properties and clinical efficacy in critically
10. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, ill patients. Ann Intensive Care. 2011;1(1):30.
Leeder JS, Kauffman RE. Developmental pharmacology–drug 29. Sullins AK, Abdel-Rahman SM. Pharmacokinetics of antibac-
disposition, action, and therapy in infants and children. N Engl terial agents in the CSF of children and adolescents. Pediatr
J Med. 2003;349(12):1157–1167. Drugs. 2013;15(2):93–117.
11. van den Anker JN, Schwab M, Kearns GL. Developmental 30. Onufrak NJ, Forrest A, Gonzalez D. Pharmacokinetic and
pharmacokinetics. Handb Exp Pharmacol. 2011;205:51–75. pharmacodynamic principles of anti-infective dosing. Clin
12. Vinita BP, Nahata MC. Drug dosing in pediatric patients. In: Ther. 2016;38(9):1930–1947.
Murphy JE, ed. Clinical Pharmacokinetics. 5th ed. Bethesda, 31. Autmizguine J, Guptill JT, Cohen-Wolkowiez M, Benjamin
MD: American Society of Health-System Pharmacists; DK Jr, Capparelli EV. Pharmacokinetics and pharmacody-
2012. namics of antifungals in children: clinical implications. Drugs.
13. Pai VB, Nahata MC. Dosing in pediatric patients. In: Murphy 2014;74(8):891–909.
JE, ed. Clinical Pharmacokinetics. 6th ed. Bethesda, MD: 32. Stockmann C, Constance JE, Roberts JK, et al. Pharmacoki-
American Society of Health-System Pharmacists; 2017:65–76. netics and pharmacodynamics of antifungals in children and
14. Oyaert M, Spriet I, Allegaert K, et al. Factors impacting their clinical implications. Clin Pharmacokinet. 2014;53(5):429–
unbound vancomycin concentrations in different patient popu- 454.
lations. Antimicrob Agents Chemother. 2015;59(11):7073–7079. 33. Cies JJ, Shankar V, Schlichting C, Kuti JL. Population phar-
15. Smith MJ, Gonzalez D, Goldman JL, et al. Pharmacokinetics macokinetics of piperacillin/tazobactam in critically ill young
of clindamycin in obese and nonobese children. Antimicrob children. Pediatr Infect Dis J. 2014;33(2):168–173.
Agents Chemother. 2017;61(4). 34. Courter JD, Kuti JL, Girotto JE, Nicolau DP. Optimizing
16. Mooij MG, Nies AT, Knibbe CA, et al. Development of human bactericidal exposure for beta-lactams using prolonged and
membrane transporters: drug disposition and pharmacogenet- continuous infusions in the pediatric population. Pediatr Blood
ics. Clin Pharmacokinet. 2016;55(5):507–524. Cancer. 2009;53(3):379–385.
17. Lam J, Baello S, Iqbal M, et al. The ontogeny of P-glycoprotein 35. Ellis JM, Kuti JL, Nicolau DP. Use of Monte Carlo simulation
in the developing human blood-brain barrier: implication for to assess the pharmacodynamics of beta-lactams against Pseu-
opioid toxicity in neonates. Pediatr Res. 2015;78(4):417–421. domonas aeruginosa infections in children: a report from the
18. Vinks AA, Emoto C, Fukuda T. Modeling and simulation OPTAMA program. Clin Therapeut. 2005;27(11):1820–1830.
in pediatric drug therapy: application of pharmacometrics 36. Bradley JS, Sauberan JB, Ambrose PG, Bhavnani SM, Ras-
to define the right dose for children. Clin Pharmacol Ther. mussen MR, Capparelli EV. Meropenem pharmacokinet-
2015;98(3):298–308. ics, pharmacodynamics, and Monte Carlo simulation in the
19. Ette EI, Williams PJ. Population pharmacokinetics I: neonate. Pediatr Infect Dis J. 2008;27(9):794–799.
background, concepts, and models. Ann Pharmacother. 37. Cohen-Wolkowiez M, Ouellet D, Smith PB, et al. Population
2004;38(10):1702–1706. pharmacokinetics of metronidazole evaluated using scavenged
20. Samardzic J, Allegaert K, Wilbaux M, Pfister M, van den Anker samples from preterm infants. Antimicrob Agents Chemother.
JN. Quantitative clinical pharmacology practice for optimal use 2012;56(4):1828–1837.
of antibiotics during the neonatal period. Expert Opin Drug 38. Cohen-Wolkowiez M, Poindexter B, Bidegain M, et al. Safety
Metab Toxicol. 2016;12(4):367–375. and effectiveness of meropenem in infants with suspected or
21. Barker CI, Standing JF, Turner MA, McElnay JC, Shar- complicated intra-abdominal infections. Clin Infect Diseases.
land M. Antibiotic dosing in children in Europe: can we 2012;55(11):1495–1502.
grade the evidence from pharmacokinetic/pharmacodynamic 39. Smith PB, Cohen-Wolkowiez M, Castro LM, et al. Population
studies - and when is enough data enough? Curr Opin Infect Dis. pharmacokinetics of meropenem in plasma and cerebrospinal
2012;25(3):235–242. fluid of infants with suspected or complicated intra-abdominal
22. Jelliffe RW, Schumitzky A, Van Guilder M, et al. Individualiz- infections. Pediatr Infect Dis J. 2011;30(10):844–849.
ing drug dosage regimens: roles of population pharmacokinetic 40. Tremoulet A, Le J, Poindexter B, et al. Population pharmacoki-
and dynamic models, Bayesian fitting, and adaptive control. netics of ampicillin in neonates using an opportunistic study
Ther Drug Monit. 1993;15(5):380–393. design. Antimicrob Agents Chemother. 2014;58(6):3013–3020.
23. Beaubien AR, Desjardins S, Ormsby E, et al. Incidence 41. van den Anker JN, Pokorna P, Kinzig-Schippers M, et al.
of amikacin ototoxicity: a sigmoid function of total drug Meropenem pharmacokinetics in the newborn. Antimicrob
exposure independent of plasma levels. Am J Otolaryngol. Agents Chemother. 2009;53(9):3871–3879.
1989;10(4):234–243. 42. Cohen-Wolkowiez M, Benjamin DK Jr, Ross A, et al.
24. Lunn DJ. Bayesian analysis of population pharmacoki- Population pharmacokinetics of piperacillin using scavenged
netic/pharmacodynamic models. In: Husmeier D, Dybowski samples from preterm infants. Ther Drug Monit.
R, Roberts S, eds. Probabilistic Modelling in Bioinformatics 2012;34(3):312–319.
Le and Bradley S121

43. Cirillo I, Vaccaro N, Castaneda-Ruiz B, et al. Open-label study 59. Drusano GL, Louie A. Optimization of aminoglycoside
to evaluate the single-dose pharmacokinetics, safety, and toler- therapy. Antimicrob Agents Chemother. 2011;55(6):2528–
ability of doripenem in infants less than 12 weeks in chrono- 2531.
logical age. Antimicrob Agents Chemother. 2015;59(8):4742– 60. Rao SC, Srinivasjois R, Hagan R, Ahmed M. One dose per day
4749. compared to multiple doses per day of gentamicin for treatment
44. Vinks AA, van Rossem RN, Mathot RA, Heijerman HG, Mou- of suspected or proven sepsis in neonates. Cochrane Database
ton JW. Pharmacokinetics of aztreonam in healthy subjects and Syst Rev. 2011(11):CD005091.
patients with cystic fibrosis and evaluation of dose-exposure 61. Smyth AR, Bhatt J. Once-daily versus multiple-daily dosing
relationships using Monte Carlo simulation. Antimicrob Agents with intravenous aminoglycosides for cystic fibrosis. Cochrane
Chemother. 2007;51(9):3049–3055. Database Syst Rev. 2012;2:CD002009.
45. Ashish A, Shaw M, Winstanley C, Ledson MJ, Walshaw 62. Mohamed AF, Nielsen EI, Cars O, Friberg LE.
MJ. Increasing resistance of the Liverpool Epidemic Strain Pharmacokinetic-pharmacodynamic model for gentamicin
(LES) of Pseudomonas aeruginosa (Psa) to antibiotics in cystic and its adaptive resistance with predictions of dosing
fibrosis (CF)–a cause for concern? J Cyst Fibros. 2012;11(3): schedules in newborn infants. Antimicrob Agents Chemother.
173–179. 2012;56(1):179–188.
46. Spencker FB, Staber L, Lietz T, Schille R, Rodloff AC. Devel- 63. Flume PA, Mogayzel PJ Jr, Robinson KA, et al. Cystic fibrosis
opment of resistance in Pseudomonas aeruginosa obtained from pulmonary guidelines: treatment of pulmonary exacerbations.
patients with cystic fibrosis at different times. Clin Microbiol Am J Respir Crit Care Med. 2009;180(9):802–808.
Infect. 2003;9(5):370–379. 64. Roberts JK, Stockmann C, Constance JE, et al. Pharmacoki-
47. Le J, Bradley JS, Hingtgen S, et al. Pharmacokinetics of single- netics and pharmacodynamics of antibacterials, antifungals,
dose ceftaroline fosamil in children with cystic fibrosis. Pediatr and antivirals used most frequently in neonates and infants. Clin
Pulmonol. 2017;52(11):1424–1434. Pharmacokinet. 2014;53(7):581–610.
48. Reed MD, Goldfarb J, Yamashita TS, Lemon E, Blumer 65. Glover ML, Shaffer CL, Rubino CM, et al. A multicenter
JL. Single-dose pharmacokinetics of piperacillin and tazobac- evaluation of gentamicin therapy in the neonatal intensive care
tam in infants and children. Antimicrob Agents Chemother. unit. Pharmacotherapy. 2001;21(1):7–10.
1994;38(12):2817–2826. 66. Dupuis LL, Sung L, Taylor T, et al. Tobramycin pharmacoki-
49. Delvallee M, Mazingue F, Abouchahla W, et al. Optimiza- netics in children with febrile neutropenia undergoing stem cell
tion of continuous infusion of piperacillin-tazobactam in transplantation: once-daily versus thrice-daily administration.
children with fever and neutropenia. Pediatr Infect Dis J. Pharmacotherapy. 2004;24(5):564–573.
2013;32(9):962–964. 67. Inparajah M, Wong C, Sibbald C, et al. Once-daily gen-
50. Kuti JL, Nightingale CH, Quintiliani R, Nicolau D. Phar- tamicin dosing in children with febrile neutropenia resulting
macodynamic profiling of continuously infused piperacillin/ from antineoplastic therapy. Pharmacotherapy. 2010;30(1):43–
tazobactam against Pseudomonas aeruginosa using Monte 51.
Carlo analysis. Diagn Microbiol Infect Dis. 2002;44(1):51–57. 68. Sung L, Dupuis LL, Bliss B, et al. Randomized controlled trial
51. Tam VH, McKinnon PS, Akins RL, Rybak MJ, Dru- of once- versus thrice-daily tobramycin in febrile neutropenic
sano GL. Pharmacodynamics of cefepime in patients with children undergoing stem cell transplantation. J Natl Cancer
Gram-negative infections. J Antimicrob Chemother. 2002;50(3): Inst. 2003;95(24):1869–1877.
425–428. 69. de Alba Romero C, Gomez Castillo E, Manzanares Secades
52. Zelenitsky SA, Ariano RE, Zhanel GG. Pharmacodynamics C, Rodriguez Lopez J, Arreaza Lopez L, Saenz Valiente P.
of empirical antibiotic monotherapies for an intensive care Once daily gentamicin dosing in neonates. Pediatr Infect Dis J.
unit (ICU) population based on Canadian surveillance data. 1998;17(12):1169–1171.
J Antimicrob Chemother. 2011;66(2):343–349. 70. van Lent-Evers NA, Mathot RA, Geus WP, van Hout BA,
53. Walker MC, Lam WM, Manasco KB. Continuous and ex- Vinks AA. Impact of goal-oriented and model-based clin-
tended infusions of beta-lactam antibiotics in the pediatric ical pharmacokinetic dosing of aminoglycosides on clini-
population. Ann Pharmacother. 2012;46(11):1537–1546. cal outcome: a cost-effectiveness analysis. Ther Drug Monit.
54. Moore RD, Lietman PS, Smith CR. Clinical response to 1999;21(1):63–73.
aminoglycoside therapy: importance of the ratio of peak con- 71. Lowdin E, Odenholt I, Cars O. In vitro studies of pharmacody-
centration to minimal inhibitory concentration. J Infect Dis. namic properties of vancomycin against Staphylococcus aureus
1987;155(1):93–99. and Staphylococcus epidermidis. Antimicrob Agents Chemother.
55. Pagkalis S, Mantadakis E, Mavros MN, Ammari C, Falagas 1998;42(10):2739–2744.
ME. Pharmacological considerations for the proper clinical use 72. Kullar R, Davis SL, Levine DP, Rybak MJ. Impact of
of aminoglycosides. Drugs. 2011;71(17):2277–2294. vancomycin exposure on outcomes in patients with methicillin-
56. Tam VH, Ledesma KR, Vo G, Kabbara S, Lim TP, Nikolaou resistant Staphylococcus aureus bacteremia: support for
M. Pharmacodynamic modeling of aminoglycosides against consensus guidelines suggested targets. Clin Infect Dis.
Pseudomonas aeruginosa and Acinetobacter baumannii: iden- 2011;52(8):975–981.
tifying dosing regimens to suppress resistance development. 73. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic
Antimicrob Agents Chemother. 2008;52(11):3987–3993. monitoring of vancomycin in adult patients: a consensus review
57. Young DC, Zobell JT, Stockmann C, et al. Optimiza- of the American Society of Health-System Pharmacists, the
tion of anti-pseudomonal antibiotics for cystic fibrosis pul- Infectious Diseases Society of America, and the Society of
monary exacerbations: V. Aminoglycosides. Pediatr Pulmonol. Infectious Diseases Pharmacists. Am J Health Syst Pharm.
2013;48(11):1047–1061. 2009;66(1):82–98.
58. Downes KJ, Hahn A, Wiles J, Courter JD, Vinks AA. 74. Frymoyer A, Guglielmo BJ, Hersh AL. Desired vancomycin
Dose optimisation of antibiotics in children: application of trough serum concentration for treating invasive methicillin-
pharmacokinetics/pharmacodynamics in paediatrics. Int J An- resistant Staphylococcal infections. Pediatr Infect Dis J.
timicrob Agents. 2014;43(3):223–230. 2013;32(10):1077–1079.
S122 The Journal of Clinical Pharmacology / Vol 58 No S10 2018

75. Le J, Bradley JS, Murray W, et al. Improved vancomycin dosing the concentration-time curve in neonates. Antimicrob Agents
in children using area under the curve exposure. Pediatr Infect Chemother. 2014;58(11):6454–6461.
Dis J. 2013;32(4):e155–163. 90. Lestner JM, Hill LF, Heath PT, Sharland M. Vancomycin
76. Hahn A, Frenck RW Jr, Allen-Staat M, Zou Y, Vinks AA. toxicity in neonates: a review of the evidence. Curr Opin Infect
Evaluation of target attainment of vancomycin area under Dis. 2016;29(3):237–247.
the curve in children with methicillin-resistant Staphylococcus 91. Le J, Ngu B, Bradley JS, et al. Vancomycin monitoring
aureus bacteremia. Ther Drug Monit. 2015;37(5):619–625. in children using Bayesian estimation. Ther Drug Monit.
77. McNeil JC, Kaplan SL, Vallejo JG. The influence of the route 2014;36(4):510–518.
of antibiotic administration, methicillin susceptibility, van- 92. Neely MN, Youn G, Jones B, et al. Are vancomycin trough
comycin duration and serum trough concentration on outcomes concentrations adequate for optimal dosing? Antimicrob Agents
of pediatric Staphylococcus aureus bacteremic osteoarticular Chemother. 2014;58(1):309–316.
infection. Pediatr Infect Dis J. 2017;36(6):572–577. 93. Ren Z, Zajicek A. Review of the Best Pharmaceuticals for
78. McNeil JC, Kok EY, Forbes AR, et al. Healthcare-associated Children Act and the Pediatric Research Equity Act: what can
Staphylococcus aureus bacteremia in children: evidence for the obstetric community learn from the pediatric experience?
reverse vancomycin creep and impact of vancomycin trough Semin Perinatol. 2015;39(7):530–531.
values on outcome. Pediatr Infect Dis J. 2016;35(3):263–268. 94. Espie P, Tytgat D, Sargentini-Maier ML, Poggesi I, Watelet JB.
79. Welsh KJ, Abbott AN, Lewis EM, et al. Clinical character- Physiologically based pharmacokinetics (PBPK). Drug Metab
istics, outcomes, and microbiologic features associated with Rev. 2009;41(3):391–407.
methicillin-resistant Staphylococcus aureus bacteremia in pe- 95. De Cock RF, Allegaert K, Brussee JM, et al. Simultaneous
diatric patients treated with vancomycin. J Clin Microbiol. pharmacokinetic modeling of gentamicin, tobramycin and van-
2010;48(3):894–899. comycin clearance from neonates to adults: towards a semi-
80. Le J, Ny P, Capparelli E, et al. Pharmacodynamic charac- physiological function for maturation in glomerular filtration.
teristics of nephrotoxicity associated with vancomycin use in Pharm Res. 2014;31(10):2643–2654.
children. J Pediatric Infect Dis Soc. 2015;4(4):e109–116. 96. Meibohm B, Laer S, Panetta JC, Barrett JS. Population phar-
81. McKamy S, Hernandez E, Jahng M, Moriwaki T, Deveikis macokinetic studies in pediatrics: issues in design and analysis.
A, Le J. Incidence and risk factors influencing the develop- AAPS J. 2005;7(2):E475–487.
ment of vancomycin nephrotoxicity in children. J Pediatr. 97. Dorofaeff T, Bandini RM, Lipman J, Ballot DE, Roberts JA,
2011;158(3):422–426. Parker SL. Uncertainty in antibiotic dosing in critically ill
82. Myers AL, Gaedigk A, Dai H, James LP, Jones BL, Neville neonate and pediatric patients: can microsampling provide the
KA. Defining risk factors for red man syndrome in children and answers? Clin Ther. 2016;38(9):1961–1975.
adults. Pediatr Infect Dis J. 2012;31(5):464–468. 98. Laughon MM, Benjamin DK, Jr., Capparelli EV, et al.
83. Jacqz-Aigrain E, Zhao W, Sharland M, van den Anker JN. Use Innovative clinical trial design for pediatric therapeutics. Expert
of antibacterial agents in the neonate: 50 years of experience Rev Clin Pharmacol. 2011;4(5):643–652.
with vancomycin administration. Semin Fetal Neonatal Med. 99. Cohen-Wolkowiez M, Sampson M, Bloom BT, et al. Deter-
2013;18(1):28–34. mining population and developmental pharmacokinetics of
84. Le J, Vaida F, Nguyen E, et al. Population-based pharmacoki- metronidazole using plasma and dried blood spot samples
netic modeling of vancomycin in children with renal insuffi- from premature infants. Pediatr Infect Dis J. 2013;32(9):956–
ciency. J Pharmacol Clin Toxicol. 2014;2(1):1017–1026. 961.
85. Mehrotra N, Tang L, Phelps SJ, Meibohm B. Evalua- 100. Suyagh M, Collier PS, Millership JS, et al. Metronidazole
tion of vancomycin dosing regimens in preterm and term population pharmacokinetics in preterm neonates using dried
neonates using Monte Carlo simulations. Pharmacotherapy. blood-spot sampling. Pediatrics. 2011;127(2):e367–374.
2012;32(5):408–419. 101. Tremoulet A, Le J, Poindexter B, et al. Characterization of the
86. Patel AD, Anand D, Lucas C, Thomson AH. Continu- population pharmacokinetics of ampicillin in neonates using
ous infusion of vancomycin in neonates. Arch Dis Child. an opportunistic study design. Antimicrob Agents Chemother.
2013;98(6):478–479. 2014;58(6):3013–3020.
87. Zhao W, Kaguelidou F, Biran V, et al. External evalua- 102. Wade KC, Wu D, Kaufman DA, et al. Population pharma-
tion of population pharmacokinetic models of vancomycin in cokinetics of fluconazole in young infants. Antimicrob Agents
neonates: the transferability of published models to different Chemother. 2008;52(11):4043–4049.
clinical settings. Br J Clin Pharmacol. 2013;75(4):1068–1080. 103. Giebel NL, Shadley JD, McCarver DG, et al. Role of chromatin
88. Zhao W, Lopez E, Biran V, Durrmeyer X, Fakhoury M, Jacqz- structural changes in regulating human CYP3A ontogeny. Drug
Aigrain E. Vancomycin continuous infusion in neonates: dosing Metab Dispos. 2016;44(7):1027–1037.
optimisation and therapeutic drug monitoring. Arch Dis Child. 104. Downes KJ, Hahn A, Wiles J, Courter JD, Vinks AA.
2013;98(6):449–453. Dose optimisation of antibiotics in children: application of
89. Frymoyer A, Hersh AL, El-Komy MH, et al. Association pharmacokinetics/pharmacodynamics in paediatrics. Int J An-
between vancomycin trough concentration and area under timicrob Agents. 2014;43(3):223–230.

You might also like