You are on page 1of 31

Clin Pharmacokinet (2014) 53:581–610

DOI 10.1007/s40262-014-0147-0

REVIEW ARTICLE

Pharmacokinetics and Pharmacodynamics of Antibacterials,


Antifungals, and Antivirals Used Most Frequently in Neonates
and Infants
Jessica K. Roberts • Chris Stockmann • Jonathan E. Constance •

Justin Stiers • Michael G. Spigarelli • Robert M. Ward •


Catherine M. T. Sherwin

Published online: 29 May 2014


Ó Springer International Publishing Switzerland 2014

Abstract Antimicrobials and antivirals are widely used activity is maximized when the plasma concentration
in young infants and neonates. These patients have his- exceeds the minimum inhibitory concentration for a pro-
torically been largely excluded from clinical trials and, as a longed period, suggesting that more frequent dosing may
consequence, the pharmacokinetics and pharmacodynam- optimize b-lactam therapy. Aminoglycosides are typically
ics of commonly used antibacterials, antifungals, and an- administered at longer intervals with larger doses in order
tivirals are incompletely understood in this population. to maximize exposure (i.e., area under the plasma con-
This review summarizes the current literature specific to centration–time curve) with gestational age and weight
neonates and infants regarding pharmacokinetic parameters strongly influencing the pharmacokinetic profile. None-
and changes in neonatal development that affect antimi- theless, safety concerns necessitate therapeutic drug mon-
crobial and antiviral pharmacodynamics. Specific drug itoring across the entire neonatal and young infant
classes addressed include aminoglycosides, aminopenicil- spectrum. Vancomycin, representing the glycopeptide class
lins, cephalosporins, glycopeptides, azole antifungals, of antibacterials, has a long history of clinical utility, yet
echinocandins, polyenes, and guanosine analogs. Within there is still uncertainty about the optimal pharmacody-
each drug class, the pharmacodynamics, pharmacokinetics, namic index in neonates and young infants. The high
and clinical implications and future directions for proto- degree of pharmacokinetic variability in this population
typical agents are discussed. b-Lactam antibacterial makes therapeutic drug monitoring essential to ensure
adequate therapeutic exposure. Among neonates treated
with the triazole agent fluconazole, it has been speculated
that loading doses may improve pharmacodynamic target
J. K. Roberts  C. Stockmann  J. E. Constance  attainment rates. The use of voriconazole necessitates
M. G. Spigarelli  R. M. Ward  C. M. T. Sherwin (&)
therapeutic drug monitoring and dose adjustments for
Division of Clinical Pharmacology, Department of Pediatrics,
University of Utah School of Medicine, University of Utah patients with hepatic dysfunction. Neonates treated with
Health Sciences Center, 295 Chipeta Way, Salt Lake City, lipid-based formulations of the polyene amphotericin B
UT 84108, USA may be at an increased risk of death, such that alternative
e-mail: catherine.sherwin@hsc.utah.edu
antifungal agents should be considered for neonates with
C. Stockmann invasive fungal infections. Alternative antifungal agents
Division of Infectious Diseases, Department of Pediatrics, such as micafungin and caspofungin also exhibit unique
University of Utah School of Medicine, Salt Lake City, pharmacokinetic considerations in this population. Neo-
UT, USA
nates rapidly eliminate micafungin and require nearly three
C. Stockmann  M. G. Spigarelli times the normal adult dose to achieve comparable levels
Department of Pharmacology/Toxicology, University of Utah of systemic exposure. Conversely, peak caspofungin con-
College of Pharmacy, Salt Lake City, UT, USA centrations have been reported to be similar among neo-
nates and adults. However, both of these drugs feature
J. Stiers  R. M. Ward
Division of Neonatology, Department of Pediatrics, University favorable safety profiles. Recent studies with acyclovir
of Utah School of Medicine, Salt Lake City, UT, USA have suggested that current dosing regimens may not result
582 J. K. Roberts et al.

in therapeutic central nervous system concentrations and antifungals, echinocandins, polyenes, and guanosine ana-
more frequent dosing may be required for neonates at later logs. A prototypic drug or drugs were chosen for each drug
postmenstrual ages. Though ganciclovir and valganciclovir class and is discussed in detail. As a consequence of leg-
demonstrate excellent activity against cytomegalovirus, islative advancements, a large body of evidence is avail-
they are associated with significant neutropenia. In sum- able for newer drugs, which are frequently investigated in
mary, many pharmacokinetic and pharmacodynamic stud- neonatal clinical trials and featured prominently in this
ies have been conducted in this vulnerable population; review. In addition, a larger emphasis was placed on the
however, there are also substantial gaps in our knowledge known pharmacokinetics of each drug class. A literature
that require further investigation. These studies will be search was conducted using PubMed where the anti-in-
invaluable in determining optimal neonatal dosing regi- fectives specifically addressed in this review in conjunction
mens that have the potential to improve clinical outcomes with key words, such as ‘neonate’, ‘infant’, ‘pharmacody-
and decrease adverse effects associated with antimicrobial namics’, and ‘pharmacokinetics’, were used to identify
and antiviral treatments. relevant literature. Additional references were identified
from the reference lists of published articles. Pharmaco-
kinetics studies that did not stratify their analyses to enable
our evaluation of patients less than 1 year of age were not
Key Points
included in this review.
Though many antibacterials have been around for
decades, variable pharmacokinetics of many drug
2 Antibacterials
classes requires therapeutic drug monitoring in
neonates and infants.
Neonatal sepsis has declined over the last 75 years, but still
Antifungal drugs display a safer toxicity profile in remains the leading cause of mortality in neonates in
neonates and infants than in older children and developed countries [4, 5]. Sepsis can be defined as early
adults. onset (\7 days after birth confirmed by blood culture),
Minimal studies have been published with neonatal usually acquired from the mother, or late onset ([7 days
and infant antiviral drug use and more work needs to after birth confirmed by blood culture) acquired nosoco-
be completed to correctly treat this population. mially [4]. The overall incidence of early-onset sepsis, as
reported by the National Institute of Child Health and
Human Development Neonatal Research Network, is esti-
mated to be 0.98/1,000 births, although late-onset sepsis
occurs most commonly in pre-term very low weight infants
1 Introduction and is inversely related to gestational age (GA) [6]. Ami-
noglycosides, aminopenicillins, glycopeptides, and cepha-
Pharmacokinetic and pharmacodynamic data for the neo- losporins are commonly used to treat sepsis in neonates and
nate and infant populations are increasing, but remain infants (pharmacodynamics in Table 1; pharmacokinetics
minimal for specific drugs and specific populations of in Table 2).
patients. Historically, neonates and infants have been
excluded from clinical trials for a variety of reasons, 2.1 Aminoglycosides: Gentamicin
including their small blood volume, lack of validated
endpoints, and ethical concerns related to frequent blood 2.1.1 Pharmacodynamics
draws [1]. Some of these limitations have been overcome
with the recent use of scavenged blood samples and dry Gentamicin is primarily used for the treatment of Gram-
blood spot analysis. More recently, federal regulations negative sepsis [7]. Its mechanism of action is to bind
have promoted the study of drugs in newborns, which is irreversibly to the 30S subunit of bacterial ribosomes,
often required to account for developmental and matura- which inhibits protein synthesis, and results in cell death
tional processes that occur throughout the first year of life [8]. Resistance is largely due to transfer of plasmids and
[2, 3]. This review attempts to incorporate the pharmaco- the expression of aminoglycoside-modifying enzymes [9].
kinetic and pharmacodynamic data available in the litera- Gentamicin resistance occurs in a wide range of microbes
ture on the treatment of neonates and infants 1 year of age including Escherichia coli, Klebsiella, Pseudomonas,
and younger, using the following representative antibac- Staphylococcus aureus, coagulase-negative staphylococci,
terial, antifungal, and antiviral classes: aminoglycosides, and Enterobacter [10–16]. Gentamicin penetrates several
aminopenicillins, glycopeptides, cephalosporins, azole tissues of the newborn, with the kidney and lung having the
Antimicrobial/Antiviral PK–PD in Neonates and Infants 583

Table 1 Antibacterial pharmacodynamics


Class Drug Mechanism of action Resistant microbes Tissue Adverse events References
penetration

Aminoglycosides Gentamicin Bind irreversibly to Escherichia coli, Kidney and Nephrotoxicity and [7–19, 206]
30S subunit of Klebsiella, lung [ heart otoxicity
bacterial ribosomes, Pseudomonas, and
disrupting protein Staphylococcus muscle [ brain
synthesis aureus, coagulase-
negative
staphylococci,
Enterobacter
Aminopenicillins Ampicillin Irreversibly inhibits Enterobacter cloacae, Penetrates most Erythematous rashes, [109, 138,
transpeptidase, an Enterococcus faecium tissues; poor vomiting, diarrhea, 207, 208]
essential enzyme for CSF hepatic impairment
bacterial cell wall penetration with chronic use,
fission hypersensitivity
reaction, seizures,
pseudomembranous
enterocolitis,
impaired platelet
function
Glycopeptides Vancomycin Inhibition of cell wall Vancomycin-resistant Penetrates most Nephrotoxicity, Red [209–211]
biosynthesis by Enterococcus (VRE) tissues; CSF man’s syndrome,
blocking spp., vancomycin- penetration is ototoxicity,
incorporation of N- intermediate increased by neutropenia
acetylmuramic acid Staphylococcus inflammation
and N- aureus (VISA), of the
acetylglucosamine vancomycin-resistant meninges
peptide subunits S. aureus (VRSA)
Cephalosporins Cefotaxime Inhibits penicillin- Enterobacter spp., Brain Hypersensitivity and [180–188,
binding proteins Ib Serratia spp., and gastrointestinal 212]
and III, inhibiting Citrobacter freundii
bacterial wall
synthesis
CSF cerebrospinal fluid

highest concentrations, followed by the heart and muscle, which is consistent with the larger extracellular fluid
with very low levels in the brain [17]. Nephrotoxicity, compartment and volume of distribution (Vd) reported
though not as common in the neonate, has been correlated among pre-term infants. However, beyond the first week of
with high trough levels above 2 lg/mL, and ototoxicity has life, pre-term infants had a higher Cmax (4.74 lg/mL) than
been associated with peak serum concentrations[12 lg/mL full-term infants (3.00 lg/mL). Among full-term infants,
[18, 19]. The pharmacodynamic parameter that has been higher birthweights associated with a decreased Vd were
most strongly associated with bactericidal activity is the correlated with higher serum gentamicin levels during the
area under the plasma concentration–time curve (AUC), first 2 h after administration. In babies under 1 week of
which is maximized by the use of larger doses, which are age, the elimination half-life (t‘) for gentamicin was also
administered infrequently [20]. affected by the baby’s weight: t‘ = 11.5 h for infants
under 1,500 g, 8 h for infants 1,500–1,999 g, and 4.25–5 h
2.1.2 Pharmacokinetics for infants over 2,000 g. When patients were stratified by
PNA, pre-term infants less than one week of age had a t‘
In 1972, the US FDA approved a pediatric formulation of of 6.25 h, where all other infants ([1 week, \1 month)
gentamicin [21]. McCracken and Jones [22] evaluated ranged from 3.25 to 4.5 h, possibly due to more mature
gentamicin use in 42 neonates, where dosage, weight, GA, kidney function in the older infants. In three infants treated
and postnatal age (PNA) were taken into consideration as for meningitis, cerebral spinal fluid (CSF)/serum ratios
factors affecting pharmacokinetic parameters. At a dose of ranged from 10 to 120 %, with higher ratios correlating
1.5 mg/kg every 8–12 h, researchers reported a higher with higher cell counts and protein in the CSF. A follow-up
maximum (peak) concentration (Cmax) in full-term infants study evaluated the role of intramuscular (IM) dosing,
(2.72 lg/mL) than pre-term infants (\35 weeks; 2.60 lg/mL), serum creatinine, and weight on urinary clearance in 32
584

Table 2 Antibacterial pharmacokinetics


Class Drug Recommended dosing Therapeutic range Metabolism Clearance References

Aminoglycosides Gentamicin PNA B14 days, 5 mg/kg/q48 h IV Peak 5–10 lg/mL NA Kidney, [7–19, 206]
\1 kg unchanged
PNA B7 days, 5 mg/kg/q48 h IV
1–2 kg
PNA 15–28 days, 4–5 mg/kg/q24–48 h IV
\1 kg
PNA 8–28 days, 4–5 mg/kg/q24–48 h IV Trough Below 2 lg/mL
C1–2 kg
PNA B7 days, 4 mg/kg/q24 h IV
[2 kg
PNA 8–28 days, 4 mg/kg/q12–24 h IV
[2 kg
Aminopenicillins Ampicillin PNA B7 days, 50 mg/kg q12 h IV, IM NA Hepatic (10 %) Kidney [109, 138,
\2 kg (25–40 %) 207, 208]
PNA B7 days, 50 mg/kg q8 h IV, IM
C2 kg
PNA [7 days, 50 mg/kg q8 h IV, IM
\2 kg
PNA [7 days, 50 mg/kg q6 h IV, IM
C2 kg
Glycopeptides Vancomycin PMA B29 weeks, 10–15 mg/kg q18 h, IV Peak 30–40 mg/L NA Kidney, [209–211]
PNA 0–14 days infusion 1 h (PNA [14, unchanged
q12 h)
PMA 30–36 weeks, 10–15 mg/kg q12 h, IV
PNA 0–14 days infusion 1 h (PNA [14, q8 h)
PMA 37–44 weeks, 10–15 mg/kg q12 h, IV Trough 5–15 mg/mL, 15–20 mg/mL
PNA 0–7 days infusion 1 h (PNA [7, q8 h) for invasive infection
PMA C45 weeks 10–15 mg/kg q6 h, IV infusion
1h
Cephalosporins Cefotaxime Neonates \1 week 50 mg/kg q12 h IV NA Liver— Kidney [180–188,
Neonates [1 week 50 mg/kg q8 h IV desacetylcefotaxime 212]
(active metabolite)
Older infants 50 mg/kg q 6h IV
IM intramuscular, IV intravenous, NA not available, PMA postmenstrual age, PNA postnatal age, qx h every x hours
J. K. Roberts et al.
Antimicrobial/Antiviral PK–PD in Neonates and Infants 585

neonates [23]. In this study, excretion of gentamicin was Giacoia and Schentag [87] evaluated multiple daily dosing
directly correlated with PNA and creatinine clearance versus continuous infusion and found that infants who
(increased PNA correlated with increased clearance). received gentamicin via continuous infusion had a larger
Gentamicin is primarily eliminated via the kidneys and its Vd, a larger AUC, a longer terminal t‘, and slower clear-
clearance is closely correlated with creatinine clearance; as ance. Additionally, creatinine clearance was significantly
such, with increases in creatinine clearance, the t‘ lower in infants who received continuously infused gen-
decreases [24]. Subsequent studies evaluating different tamicin. The authors concluded that infants receiving
dosing regimens and very low birthweight (VLBW) infants gentamicin via continuous infusion were at higher risk for
(\1,500 g) support these pharmacokinetic findings [25– nephrotoxicity. Subsequently, Sherwin et al. [88] deter-
45]. Other groups found a greater correlation between mined that ELBW (\1,000 g) neonates have prolonged IV
postconceptional age and t‘ or clearance [19, 46, 47]. One gentamicin delivery due to slow IV flow rates and small
study suggested sex played a role on clearance, with girls administered volumes, which result in only 60 % of the
having a faster clearance than boys [48]. A study by Szefler drug delivered in 60 min. The authors suggested that bolus
et al. [49] suggested that a longer dosing interval of 18 h be doses be given to this population to ensure the total dose is
used in pre-term infants\35 weeks. To further simplify the delivered.
dosing interval, Hindmarsh et al. [50] suggested a higher Gentamicin renal toxicity had been well-documented in
dose every 24 h. Longer dosing intervals have subse- adults, which prompted Haughey et al. [89] to evaluate this
quently been supported by other studies, in which dosing further in pre-term infants. The researchers determined that
intervals ranged from 24 to 36 h [44, 51–78], though safety clearance in neonates was threefold higher than adults
of once daily dosing in extremely low birthweight (ELBW) when adjusted for body surface area, which has been
infants is not well-established. supported by other aminoglycoside work [90]. Utilizing a
The route of administration alters the pharmacokinetics two-compartment model to fit the data, neonates had a
of gentamicin in ways that are clinically important. Paisley twofold higher mean Vd. Aminoglycosides are reabsorbed
et al. [79] compared IM with intravenous (IV) adminis- by the proximal tubule. However, pre-term infants have
tration in 18 infants. Cmax concentrations for IM and IV immature renal proximal tubules and the authors hypoth-
were similar, but both the magnitude and the timing of esized that this protects neonates from renal toxicity. These
Cmax varied much more widely after IM administration. findings were supported by Landers et al. [91] who iden-
This difference has also been shown by others [80]. This tified a low incidence of nephrotoxicity in VLBW infants.
could be attributed to the pharmacodynamic observation Many neonatal patients receiving gentamicin therapy
that sick infants usually have decreased blood flow to their have additional co-morbidities affecting gentamicin phar-
extremities, which decreases the absorption of drug after macokinetics and renal function. Friedman et al. [92]
IM injection. Half-lives were similar for IM and IV, with evaluated gentamicin pharmacokinetics in asphyxiated
mean concentrations equal to 4 h. Additionally, this study newborns with impaired renal function and found that the
identified PNA as a significant pharmacokinetic variable, t‘ and clearance were prolonged in asphyxiated infants,
as the t‘ decreased significantly after 7 days of life. with clearance and urine output also correlating to mean
Overall, the authors found no difference in the pharmaco- arterial pressure. Part of this finding could be attributed to
kinetics of IM and IV gentamicin. The authors recom- hypothermia used to treat asphyxiated newborns to prevent
mended a dose of 2.5 mg/kg given every 8 h, though they further neurological injury, which also decreases glomer-
acknowledged the wide range of variability in their study ular filtration rate. This finding suggests that lower doses or
population, which supported therapeutic monitoring to longer dosing intervals may be needed in asphyxiated
guide clinical dosing. McCracken et al. [81] came to the infants. Bravo et al. [93] studied malnourished infants and
same conclusion in their study. Gentamicin exhibits con- reported that the Vd was 18.3 % higher in the malnourished
centration-dependent killing, emphasizing the importance population, resulting in a higher time to Cmax (tmax).
of Cmax values. To achieve therapeutic Cmax earlier, Wat- Watterberg et al. [94] determined that patients with patent
terberg et al. [82] evaluated the use of a 4 mg/kg loading ductus arteriosus (PDA) had a significantly increased Vd
dose. Ninety-two percent of the patients who received the and serum t‘. However, the Vd decreased after ductal
loading dose achieved gentamicin serum concentrations in closure. While the effect of PDA on gentamicin pharma-
the therapeutic window after the first dose, compared with cokinetics has been demonstrated in multiple studies, it has
only 45 % of patients who were given the usual dose of not been shown to have clinical relevance and dosing
2.5 mg/kg. There was no effect upon steady-state phar- adjustments based on the presence of a PDA are not nec-
macokinetics. The authors suggested a loading dose of essary [95, 96]. The effect of extracorporeal membrane
4 mg/kg in all neonates receiving gentamicin therapy, oxygenation (ECMO) on gentamicin pharmacokinetics has
which was later supported by additional studies [83–86]. been variable. Southgate et al. [97] found ECMO treatment
586 J. K. Roberts et al.

did not significantly affect Vd in infants, but increased the fewer failures to attain therapeutic peak and trough con-
elimination t‘ by more than 3 h. As a result, the authors centrations than multiple daily dosing (relative risk: 0.22;
suggest starting at the usual 2.5 mg/kg, but using an 95 % CI 0.11–0.47) [110]. Recently, in vitro time-kill
extended dosing interval of at least 18 h. Another study experiments and semi-mechanistic pharmacokinetic and
evaluating 18 infants compared individual patient phar- pharmacodynamic modeling studies have suggested that
macokinetic parameters before and after ECMO [98]. As due to gentamicin’s extended t‘ in neonates, extended
expected, this group found that patients had a higher Vd. interval dosing is not likely to promote the emergence of
However, a lower clearance and a prolonged t‘ while adaptive resistance to gentamicin [111].
undergoing ECMO necessitated a decrease in the dose by The pharmacokinetics and pharmacodynamics of gen-
25 % in addition to lengthening the dosing interval despite tamicin have been rigorously studied in pre-term and term
the increased Vd. This finding was also supported by Dodge neonates over the last 50 years. Appropriately, many of
et al. [99] using a population pharmacokinetic approach. these studies have focused on identifying patient charac-
Other groups, however, found no significant impact of teristics that influence the likelihood of achieving a good
ECMO on gentamicin pharmacokinetics [100, 101]. A clinical outcome. However, additional studies are needed
study by Lingvall et al. [102] examined Vd differences in unique neonatal subpopulations, including those with
between babies that were septic and those that were not. intra-uterine growth restriction and perinatal asphyxia, for
Septic babies had a 14 % increase in their Vd. Sherwin which data are scarce. Conditions such as these have been
et al. [103] evaluated whether this increase was due to IV reported to alter gentamicin pharmacokinetics and there-
volume expanders, but determined the Vd was not affected fore further study is needed to determine if alternative
by cumulative volume expanders. Neither UV light therapy dosing regimens are required to optimize the achievement
associated with jaundice [104] nor being large for GA of target peak and trough concentrations that are associated
[105] had a significant effect on the pharmacokinetic with therapeutic success.
properties of gentamicin.
2.2 Aminopenicillins: Ampicillin
2.1.3 Clinical Implications and Future Research
2.2.1 Pharmacodynamics
Gentamicin is one of the most commonly prescribed anti-
bacterials in the neonatal intensive care unit [106]. His- Ampicillin features activity against many of the most
torically, pharmacodynamic studies have demonstrated that common etiological agents of early-onset neonatal sepsis,
peak antibacterial activity occurs when the maximum including group B streptococci [112]. Ampicillin is dis-
blood concentration exceeds 5 lg/mL [107]. Additionally, tinguished from penicillin G by the presence of an amino
the risk of nephrotoxicity is minimized by targeting trough group, which aids in its penetration of Gram-negative cell
concentrations \2 lg/mL to prevent the accumulation of membranes [113]. The bactericidal activity of ampicillin
gentamicin in the proximal renal tubule [108]. For most stems from its irreversible inhibition of transpeptidase,
populations, trough monitoring for safety should be ade- which is an essential component required for bacterial cell
quate. However, for some special populations (e.g., renally wall synthesis and binary fission [114]. Ampicillin displays
impaired or cystic fibrosis) two post-distributive samples time-dependent bactericidal activity, which means that the
should be taken and the AUC should be used for adjusting pharmacodynamic parameter that has been most strongly
the dose. For neonates with varying degrees of renal correlated with maximal bacterial killing is the time above
maturity, optimal IV gentamicin dosing regimens incor- the minimum inhibitory concentration (MIC) [T [ MIC] of
porate the age and weight of the newborn, such that for the infecting organism [115, 116]. To achieve optimal
newborns \1 kg the recommended dosing regimen is bactericidal activity and to minimize the emergence of
5 mg/kg every 48 h for neonates B14 days of age, and microbial resistance, it is not necessary for ampicillin
4–5 mg/kg every 24–48 h for neonates 15–28 days of age concentrations to exceed the MIC for the entire dosing
[109]. Similarly, recommended gentamicin doses for interval [117, 118]. In adults, a T [ MIC of 30–40 % of the
infants 1–2 kg are 5 mg/kg every 48 h for neonates dosing interval has been strongly correlated with good
B7 days of age and 4–5 mg/kg every 24–48 h for neonates clinical outcomes, although studies have suggested that
8–28 days of age [109]. Finally, for infants [2 kg, the 40–50 % T [ MIC is required for neonates [115]. Ampi-
recommended dosing is 4 mg/kg every 24 h for neonates cillin concentrations that are much higher than the MIC do
B7 days of age and 4 mg/kg every 12–24 h for neonates not greatly enhance bactericidal activity [119].
8–28 days of age [109]. These extended interval and once- Although rare, several hypersensitivity reactions can
daily dosing regimens have been evaluated in a large sys- occur in ampicillin-treated infants younger than 6 months
tematic meta-analysis and were found to be associated with of age [120]. Patients with a history of b-lactam
Antimicrobial/Antiviral PK–PD in Neonates and Infants 587

hypersensitivity or previous IgE-mediated reactions should relative to older children and adults. Competition for pro-
not receive ampicillin due to the potential for severe, life- tein binding sites can further increase the fraction of
threatening hypersensitivity reactions. Ampicillin admin- unbound drug, as occurs in neonatal hyperbilirubinemia
istration to neonates and infants with confirmed penicillin where endogenous bilirubin reduces ampicillin protein
allergy should be avoided. Among other infants less than binding [137].
6 months of age, ampicillin-associated drug rashes are rare,
but do occur and include: erythema multiforme, exfoliative 2.2.3 Clinical Implications and Future Research
dermatitis, and urticaria [120]. Approximately 5–10 % of
patients treated with ampicillin experience an exanthema- In the 1970s, Kaplan et al. [127] derived neonatal ampi-
tous cutaneous eruption, although this figure rises to nearly cillin dosing recommendations on the basis of in vitro
100 % among patients with viral infections (e.g., Epstein– experiments demonstrating ampicillin’s activity against
Barr virus) [121]. Management of an exanthematous Gram-positive pathogens and E. coli as well as pharma-
cutaneous reaction includes cessation of ampicillin therapy, cokinetic data obtained after administering doses of 50, 75,
switching to an alternative antimicrobial agent, and the and 100 mg/kg/dose to pre-term and full-term neonates.
addition of antihistamines or corticosteroids [122]. Ampicillin dosing recommendations from the AAP and the
British National Formulary Committee [109, 138] are
2.2.2 Pharmacokinetics similar to those reported by Kaplan et al. [127] and suggest
the use of 50 mg/kg every 12 h administered via IV push
The American Academy of Pediatrics (AAP) recommends for neonates B7 days PNA who weigh\2 kg. For neonates
that the optimal treatment of early-onset neonatal sepsis B7 days PNA who weigh [2 kg or for those who
include therapy with broad-spectrum antimicrobial agents, are [7 days PNA and weigh \2 kg the recommended dose
including ampicillin and an aminoglycoside [123]. The is 50 mg/kg every 8 h. For neonates [7 days PNA who
pharmacokinetic properties of ampicillin have been weigh C2 kg the recommended dose is 50 mg/kg every
extensively studied in pre-term and term-neonates, infants, 6 h.
children, and adults [124–134]. An early study by Boe et al. Ampicillin CSF concentrations range from 6 to 15 % of
[124] examined serum concentrations of ampicillin fol- serum concentrations [127]. Therefore, when ampicillin is
lowing IM administration and reported markedly higher used in combination with an aminoglycoside for the
concentrations and a longer t‘ in pre-term neonates than in treatment of meningitis it is recommended that the dose be
adults exposed to comparable doses. These findings are doubled from 50 to 100 mg/kg/dose [109, 127, 138, 139].
supported by a larger study by Axline et al. [125], who Since the development of ampicillin more than half a
found that the serum t‘ of ampicillin decreases with century ago, much has been learned about its optimal use in
increasing PNA. A nearly identical rate of change was the clinical setting. Current neonatal dosing recommenda-
observed at all PNA, despite differences in weight [125]. tions are predicated upon carefully conducted pharmaco-
Cmax and tmax were not different among pre-term neonates kinetic and pharmacodynamic studies and have been
when compared with older children and adults following associated with good clinical outcomes in multiple retro-
IM administration of ampicillin [125, 135]. However, the spective analyses. Conversely, relatively little is known
bioavailability of orally administered ampicillin is about how genetic variations may affect the response to
increased in neonates and infants due to their high gastric ampicillin therapy. Further investigation is warranted to
pH, which enhances the bioavailability of acid labile drugs identify patient characteristics, including genetic profiles,
[126, 136]. By 3 years of age gastric acid production which may be used to predict the risk of developing an
reaches adult levels; however, until this time, neonates and ampicillin-associated hypersensitivity reaction. Addition-
infants achieve higher serum concentrations of orally ally, further research is needed to define the appropriate
administered ampicillin than older children and adults antihistamine dose needed to effectively and safely treat
[126, 135, 136]. Additionally, neonatal gastric emptying is hypersensitivity reactions in neonates and infants.
prolonged, which results in increased absorption of acid
labile drugs such as ampicillin [131, 135]. In infants 2 2.3 Glycopeptides: Vancomycin
months of age and older, concomitant milk ingestion has
not been shown to alter ampicillin pharmacokinetics [131, 2.3.1 Pharmacodynamics
132]. In contrast, Ehrnebo et al. [137] demonstrated that
ampicillin displays age-dependent ampicillin protein It is well-understood that vancomycin dosing regimens
binding activity, with decreased protein binding observed need to be designed to clear the infection rapidly without
among neonates. This results in a larger fraction of causing toxicity or promoting resistance [140, 141].
unbound ampicillin in the circulation and increases the Vd However, multiple studies of vancomycin use in pediatric
588 J. K. Roberts et al.

patients have found that previously recommended dosing debate is recognizing that the context for the use of van-
regimens often do not achieve designated therapeutic ran- comycin often differs significantly for neonates versus an
ges [142, 143]. This is especially concerning with reports adult population. For instance, in neonates, vancomycin is
of decreased susceptibility of methicillin-resistant S. aur- frequently used empirically for suspected sepsis and for the
eus (MRSA) to vancomycin [140]. Although very limited, treatment of infections caused by coagulase-negative S.
there have been cases of MRSA treatment failure in chil- aureus. Therefore, therapeutic targets based on treating
dren with subtherapeutic vancomycin concentrations MRSA infection in adults may not be ideal for a neonate
(\5 mg/L) and in vulnerable patients, such as pre-term [157].
neonates and immunocompromised children [144, 145].
Yet, reports of clinical failure of vancomycin in the pedi- 2.3.2 Pharmacokinetics
atric population have been rare [146]. In addition to the risk
of inefficacy, subtherapeutic vancomycin concentrations The pharmacokinetic heterogeneity of vancomycin among
select for resistant strains, including the vancomycin neonates and young infants poses a significant challenge
intermediate S. aureus (VISA) [10]. for dose optimization [140] and prevents the use of simple
The adverse drug reactions associated with vancomycin standard dosing regimens without TDM [146, 158]. For
in adults seem to be diminished in both severity and fre- example, neonates have a higher percentage of bodyweight
quency among neonates and infants [143, 146]. This phe- as water and undergo rapid postnatal renal maturation
nomenon is reflected in current therapeutic drug causing changes to elimination and clearance, particularly
monitoring (TDM) practices, where concerns over toxicity within the first week of life. Integral to bacterial response is
have given way to ensuring vancomycin concentrations the concentration of free vancomycin. In adults, protein-
meet therapeutic targets [147]. In light of studies con- bound vancomycin ranges from 24 to 64 % with a mean of
firming therapeutic targets are not consistently being met, 41.5 % [159]. In addition to the variability found in the
consensus statements recommending higher target con- proportion of bound versus free vancomycin, neonates also
centrations, and a lack of evidence for severe vancomycin- have decreased protein–drug binding due to lower albumin
associated toxicity, there has been a trend toward the sue of concentrations comprised, in part, of fetal albumin, which
higher doses of vancomycin across pediatric age groups, has a lower binding affinity for drugs [135]. Furthermore,
including neonates and young infants [148–150]. Conse- patients admitted to intensive care for complex disease
quently, these factors and the collective experience of states such as renal failure, cardiovascular complications,
infrequent treatment failure may be contributing to the or severe lung disease represent pharmacokinetically dis-
wide variation found among institutions in TDM and tinct subsets of infants who may require different dosing
vancomycin use practices in pediatric patients [151, 152]. regimens [160, 161]. Attempts to understand and adapt
The pharmacodynamic target best related to clinical vancomycin use to the pharmacokinetic variability in
outcome is not known for neonates and young infants neonates and young infants are usually based on methods
[142]. Currently, therapeutic targets for MRSA infections seeking to quantify the most relevant covariates contrib-
are derived from adult, animal, and in vitro studies, in uting to vancomycin clearance [162]. Since vancomycin is
addition to therapeutic failures in newborns and infants. In eliminated unchanged almost exclusively by the kidneys,
adults, the best outcomes are associated with an AUC/ covariates associated with the maturational changes in
MIC [400, which correlates with serum trough concen- renal function and development are central to all vanco-
trations of [10 mg/L on a 12 h dosing schedule for an mycin dosing strategies in neonates and young infants
isolate with a MIC \1 mg/L [153]. Recent studies, one of [143, 163].
which included children as young as 3 months, have Vancomycin is distinctive among anti-infectives due to
determined that a significant portion of children (from 75 the intensity with which it has been the focus of pharma-
to [90 %) with serum vancomycin trough concentrations cokinetic modeling and simulation efforts to refine its use.
of approximately 7 to 10 mg/L will achieve an AUC/MIC While there has been much progress, an optimal dosing
of [400, as long as the MIC is B1 mg/L [153, 154]. regimen has yet to be defined [140]. The inter-individual
However, this finding has yet to be confirmed in neonates. variability in vancomycin disposition in neonates and
At present, the drive to increase vancomycin exposure in young infants can, in part, be explained by covariates such
pediatric patients through higher trough targets (i.e., as postmenstrual age (PMA), PNA, weight, serum creati-
increased from a historical [5–10 mg/L at steady state nine, and the concomitant use of certain drugs [164, 165].
to [10 mg/L, and to 15–20 mg/L for invasive infections However, even when the most significant covariates (i.e.,
[155]) based on extrapolation from adult data has raised age, bodyweight, and creatinine clearance) are accounted
concerns [147]. The most appropriate pharmacodynamic for in neonates, the unexplained inter-individual variability
target remains a matter of debate [156]. Central to this for clearance and Vd is still high [166]. Among pre-term
Antimicrobial/Antiviral PK–PD in Neonates and Infants 589

and term neonates (27–47 weeks PMA) the variability, pharmacokinetic perspective, and the situation is further
after accounting for serum creatinine and bodyweight, was complicated by the lack of evidence of the best pharma-
23 % for clearance and 47 % for the Vd as reported by codynamic measure for pediatric clinical outcomes.
Oudin et al. [167]. Likewise, in neonates (\29 days of Moreover, the last decade has seen an evolution in the
age), de Hoog et al. [168] found that clearance had an therapeutic target recommendations for adults, followed
associated 31 % variability (0.057 ± 0.0018 L/h/kg) and by revisions to pediatric targets for vancomycin
the Vd had 25 % (0.43 ± 0.013 L/kg) variability. A sub- concentrations.
sequent study of pre-term neonates (\35 weeks’ PMA and Vancomycin dose optimization requires an understand-
PNA \29 days) determined that the majority of the vari- ing of the pharmacokinetic profile of the patient and the
ability in vancomycin clearance was explained by the pharmacodynamic target best associated with microbio-
child’s weight (49.8 %), PMA (18.2 %), and renal function logical efficacy for that patient’s infection. At present,
(14.1 %) [162]. It is important to recall that the newborn’s there is a necessary complexity to vancomycin use in very
serum creatinine does not reflect endogenous renal function young patients, which has been associated with errors in
until maternal creatinine received across the placenta is use [167]. Therefore, the successful implementation of
cleared, which takes several days after birth. In addition to individualized regimens will also need to address pre-
the covariates that relate to maturational changes, it is also scription difficulties to maximize the benefit to patients
important to consider the influence of the assay method- [167, 174]. Attempts to simplify dosing regimens by
ology (i.e., serum creatinine and vancomycin quantification employing a single parameter, such as weight, may result
analysis) on individual and population estimates [169]. in 50–60 % of patients exposed to sub- or supra-therapeutic
Overall, population pharmacokinetic models still con- concentrations [175]. Furthermore, static formulas and
tain sufficient levels of unexplained variability to warrant nomograms are often regarded as overly simplistic for the
continued TDM for post hoc dose adjustment to achieve a pharmacokinetic variability of neonates and young infants
given pharmacodynamic target concentration [140, 166]. [176].
Nonetheless, a recent external validation analysis across Arguments have been made about simplifying the
multiple population pharmacokinetic models found that manner of administration to reduce confusion and possibly
most models led to ‘acceptable’ vancomycin concentra- improve the safety of vancomycin dosing protocols. Van-
tions in neonates [166]. Although vancomycin pharmaco- comycin has poor oral absorption, requiring IV adminis-
kinetics are best described as multi-compartmental with a tration for systemic infections [151, 172]. The use of
long distribution phase, one-compartment models perform continuous infusion has been proposed to offer advantages
well in estimating pharmacokinetic parameters for the of practicality and safety surrounding the implementation
purpose of individualizing dosage regimens [143, 158]. of complex individualized dosing regimens [167]. Addi-
One-compartment models have the advantage of requiring tionally, bactericidal activity may be improved with con-
fewer serum samples than more complex models to esti- tinuous infusion due to the ability to achieve a more
mate pharmacokinetic parameters, which is particularly consistent AUC than intermittent dosing [141]. Unfortu-
relevant for neonates and infants [158]. nately, rapid infusions of vancomycin that might occur
when a catheter is flushed exposes the patient to risk of the
2.3.3 Clinical Implications and Future Research rapid release of histamine causing what has been called the
‘‘red man syndrome’’ [177]. Currently, both intermittent
The glycopeptide antibacterial vancomycin has been and continuous infusions are used in neonates, with similar
available to treat serious infections for more than half a clinical efficacy between the two methods, but safety and
century, including in newborns and infants [170]. With the pharmacokinetic data are much more limited with respect
relatively recent emergence of MRSA, the use of vanco- to continuous infusion [168, 178].
mycin has increased significantly as it is the recommended The high degree of pharmacokinetic variability in neo-
first-line antibacterial for the prophylaxis and treatment of nates and young infants make dose individualization of
resistant Gram-positive infections [158, 171]. However, vancomycin a challenge and a priority for optimizing
coagulase-negative S. aureus remains the predominant therapy [158]. Among the myriad physiologic, demo-
infectious organism for which vancomycin is used in graphic, and clinical factors that influence vancomycin
neonates and young infants [143, 172]. Within this youn- pharmacokinetics in neonates and young infants, the most
gest patient population the t‘ of vancomycin can range important are weight, age, and renal function. While dosing
from 2 to 12 h, with highly variable rates of clearance and regimens incorporating these covariates have demonstrated
Vd [140, 160, 173]. The physiologic and developmental improved rates of attainment for currently recommended
characteristics inherent in neonates and young infants make therapeutic targets [168], more studies are required to
vancomycin a challenging drug to use from a determine the pharmacodynamic target that best correlates
590 J. K. Roberts et al.

with clinical outcomes. Finally, a key component to ECMO and found that their Vd was higher, but their clearance
improving vancomycin therapy in infants will be imple- and t‘ were similar to non-ECMO infants [201]. The authors
menting dosing regimens and TDM protocols that are less also reported that only one patient had serum concentrations
complicated for clinicians while adapting to the diverse below the MIC for more than half of the dosing interval,
pharmacokinetics of this patient population [179]. suggesting that no change in dosing is required for infants on
ECMO.
2.4 Cephalosporins: Cefotaxime
2.4.3 Clinical Implications and Future Directions
2.4.1 Pharmacodynamics
The third-generation cephalosporin cefotaxime possesses
Cefotaxime and its active metabolite desacetylcefotaxime several features that make it an attractive antibacterial
both display antimicrobial activity and are widely used in agent for the treatment of neonatal sepsis. Principally,
the treatment of neonates and young infants [180, 181]. cefotaxime displays potent activity against group B strep-
Cefotaxime inhibits bacterial wall synthesis by inhibiting tococcus, as well as E. coli, and other Gram-negative
penicillin-binding proteins Ib and III [182–184]. Cefotax- enteric bacilli [202]. However, cefotaxime monotherapy is
ime has activity against most Gram-negative bacteria and not recommended owing to the fact that large microbio-
some Gram-positive bacteria, with some resistance noted in logical surveillance studies have demonstrated that only
Enterobacter spp., Serratia spp., and Citrobacter freundii 79 % of the bacterial organisms identified from neonates
[185]. Cefotaxime penetrates well into the central nervous with late-onset sepsis were susceptible to cefotaxime alone
system (CNS) and is moderately bound by serum proteins [203]. In contrast, 95 % of the isolated organisms were
[186, 187]. Cefotaxime has a relatively large therapeutic susceptible to combination therapies with amoxicillin and
window with a general lack of serious adverse effects cefotaxime or gentamicin and amoxicillin [203]. Never-
associated with treatment in neonates and infants [188]. theless, the empiric use of cefotaxime has been reported to
However, a retrospective study completed by Clark et al. increase the risk of developing invasive candidiasis in pre-
[189] compared cefotaxime combined with ampicillin to term neonates and was associated with an increased risk of
gentamicin combined with ampicillin and found that cef- death in both pre-term and term neonates when compared
otaxime combined with ampicillin for the empiric treat- with gentamicin [189, 204]. For these reasons, the empiric
ment of neonates has an unexplained increase in mortality use of cefotaxime is discouraged [205]. However, in cases
(1.5 odds ratio), which was not related to renal failure or of suspected or proven Gram-negative bacillary meningitis,
meningitis. the use of cefotaxime is encouraged due to its superior CSF
penetration, which has been reported to result in CSF
2.4.2 Pharmacokinetics concentrations ranging from 0 to 20 % of serum concen-
trations, with improved blood–brain barrier penetration
A study completed by Kafetzis et al. [190] compared the when the meninges are inflamed [186, 202]. For neo-
pharmacokinetics of neonates born pre-term and full-term at nates \2 kg with suspected neurologic involvement, the
the ages of \1 and 1–4 weeks. Pre-term neonates \1 week recommended dosing schedule is 75 mg/kg every 12, 8,
had a significantly longer t‘ (5.7 h) than infants and adults and 6 h for neonates B7, 8–28, and [28 days of age,
(1 h). All term neonates and pre-term neonates[1 week old respectively [109]. Similarly, among neonates C2 kg, the
averaged a t‘ around 2–4 h. In addition, Vd was significantly recommended dosing schedule is 50 mg/kg every 6 h for
higher than in adults, but clearance was significantly neonates B7 days of age and 75 mg/kg every 6 h for all
decreased in all neonates. Differences between neonates and neonates and infants [7 days old [109].
adults can be attributed to decreased glomerular filtration
rates. These results are in agreement with other studies
independent of GA, with McCracken et al. [191] suggesting 3 Antifungals
that all infants \1 week receive 50 mg/kg every 12 h and
infants [1 week receive 50 mg/kg every 8 h, which has The incidence of invasive fungal infections is increasing in
subsequently been supported by others [192–197]. However, the neonatal population [213]. Invasive candidiasis is a sig-
a study evaluating VLBW neonates (\1,500 g) found an nificant problem in the neonatal ICU with mortality rates as
even longer t‘ with accumulation of the active metabolite in high as 25 % for sepsis and up to 36 % for meningitis [213].
the serum and suggested increasing the dosing interval to Advances in medical technology, such as parenteral lines,
24 h [198]. Studies among infants \1 year of age reported enteral feeding tubes, central vascular catheters, broad-
similar Vd, clearances, and t‘ values to older children and spectrum antibacterials, and lower GA newborns have
adults [186, 199, 200]. A recent study evaluated infants on increased the susceptibility of pre-term neonates to invasive
Antimicrobial/Antiviral PK–PD in Neonates and Infants 591

fungal infections [214]. Incubators also add challenges with seven infants and their data fit a one-compartment model.
skin care management and incubator humidity. In addition, Serum concentrations in infants were lower than reports of
pre-term infants’ immunodeficiency, immature skin, and adults and children. Infants also cleared the drug more
increased permeability of cutaneous barriers increase their rapidly, possibly protecting this population from the
risk for fungal infections [215, 216]. The most common nephrotoxicity seen in adults, but also suggesting infants
invasive fungal infection that affects neonates and infants is may need a higher dose. Weight was a significant covariate
candidiasis [217]. Invasive candidiasis can lead to death, on Vd and clearance. The youngest pre-term neonate
neurodevelopmental impairment, lung disease, advanced demonstrated an extended t‘ of 693 h, suggesting that pre-
retinopathy of prematurity, and periventricular leukomalacia term neonates may need dosing intervals of [24 h. A
[218–221]. As such, any patient less than 3 months of age similar study evaluated five pre-term neonates and the
should be considered to be at risk for invasive Candida authors concluded that a lower dose of 0.5 mg/kg/day was
infections and should be monitored closely for signs and sufficient to achieve a Cmax of 0.99 ± 0.48 lg/mL [236]. A
symptoms of infection. Polyenes, azoles, and echinocandins study completed by Baley et al. [237] followed up on their
are the most common drug classes used in neonates and previous work, which reported renal failure in VLBW
infants to treat disseminated candidiasis (pharmacodynam- infants when treated with amphotericin B and 5-fluorocy-
ics in Table 3; pharmacokinetics in Table 4). tosine [230]. They sought to determine if lower combined
doses would decrease renal toxicity in infants. The study
3.1 Polyenes: Amphotericin B Deoxycholate and Lipid showed that with combined therapy of amphotericin B and
Formulations 5-fluorocytosine, a lower dose could be used to prevent
renal toxicity. Five infants had CSF fluid obtained and CSF
3.1.1 Pharmacodynamics amphotericin B concentrations were 40–90 % of serum
concentrations, demonstrating good penetration into the
One of the first and most common drugs used to treat CSF. Unfortunately, 5-fluorocytosine is not available in a
invasive fungal infections in neonates is amphotericin B parenteral dosage form and must be given enterally.
deoxycholate [222], which binds to ergosterol in fungal cell Higuchi et al. [238] evaluated continuous infusion of
membranes, resulting in cell death [223]. Resistance to amphotericin B deoxycholate among ELBW neonates with
amphotericin B is rare, though there are reports of Candida and without renal failure. Serum concentrations of
glabrata, C. krusei, C. lusitaniae, and C. parapsilosis amphotericin B ranged from 0.31 to 0.78 lg/mL with daily
resistance [224, 225]. Mechanisms of resistance usually doses ranging from 0.2 to 0.55 mg/kg/day. Linear regres-
stem from a decrease in the ergosterol content in the cell sion using the daily dose and serum creatinine reliably
membrane, leading to a decrease in amphotericin B binding predicted serum concentrations.
affinity [226]. Amphotericin B deoxycholate is poorly Lipid formulations of amphotericin B were developed to
absorbed orally, is highly protein bound, and widely dis- decrease the toxicity of amphotericin B deoxycholate, though
tributed in the liver, spleen, and kidneys [227]. Ampho- neonates appear to be less susceptible to amphotericin B-
tericin B deoxycholate has been associated with high induced nephrotoxicity than older children and adults. Early
toxicity due to binding to the cholesterol elements of studies showed that lipid formulations were safer at higher
mammalian cells [228]. Some studies evaluating ampho- doses [239]. There are three lipid formulations on the market:
tericin B deoxycholate in neonates reported hypokalemia liposomal amphotericin B, amphotericin B lipid complex,
[229–231]. Although nephrotoxicity is common in adults, it and amphotericin B colloidal dispersion. Liposomal
is not as apparent in neonates [232]. However, a study amphotericin B incorporates amphotericin B into unilamellar
completed by Turcu et al. [233] found that infants with liposomes with a diameter \100 nm. Amphotericin B lipid
high sodium intake displayed a lower level of nephrotox- complex incorporates a 1:1 ratio of dimiristoyl phosphati-
icity, confirming a previous study [234], and suggesting dylcholin/dimiristoyl phosphatidylglycerol, with a size
that sodium loading may prevent or decrease nephrotoxi- ranging from 500 to 5,000 nm. The last formulation com-
city in neonates. In addition, some neonates do experience plexes amphotericin B with cholesteryl sulfate with a
infusion-related immediate hypersensitivity reactions diameter of 100 nm [223]. Complexing amphotericin B with
resulting in fevers and chills at a similar rate to adults. lipids results in a longer t‘ and decreased clearance when
Toxicity is rarely reported with lipid formulations [231]. compared with amphotericin B deoxycholate. In addition, the
large size of the complexes prevents glomerular filtration and
3.1.2 Pharmacokinetics renal elimination, protecting the kidneys but also preventing
effective treatment of fungal balls in the bladder [231].
The first pharmacokinetic studies evaluating neonates were One of the first pharmacokinetic papers published with
published in the late 1980s. Starke et al. [235] evaluated neonatal data compared concentrations of liposomal
592

Table 3 Antifungal pharmacodynamics


Class Drug Mechanism of action Mechanism of Resistant microbes Tissue Adverse events References
resistance penetration

Polyenes Amphotericin B Binds to ergosterol Decrease in Candida glabrata, Liver, spleen, Nephrotoxicity [222–227, 229–234, 239,
deoxycholate fungal cell ergosterol C. krusei, C. lusitaniae, kidneys 240, 273, 306–309]
membranes, content in cell C. parapsilosis,
Liposomal resulting in cell membrane C. guilliermondii Lungs, liver,
amphotericin B death spleen
Amphotericin B
lipid complex
Amphotericin B
colloidal
dispersion
Azoles Fluconazole Inhibits ergosterol Alteration in C. glabrata, C. krusei Kidney, CSF, Gastrointestinal, elevation [243–252, 254–257, 265–
biosynthesis, ergosterol liver, joint of liver enzymes 267, 273, 310, 311]
inhibiting cell synthesis spaces, saliva,
membrane repair vitreous humor
Voriconazole Uncharacterized C. glabrata Kidney, CSF, Elevated hepatic
liver transaminases, abnormal
vision, skin reactions due
to photosensitization
Echinocandins Caspofungin Inhibits (1,2)-b-D- Mutations to C. parapsilosis, Lung, liver, None in neonates [279–297, 312–315]
Micafungin glucan synthase FKS1 gene C. albicans, C. glabrata, spleen, kidney Elevated alanine/aspartate
enzyme, disturbing C. krusei, C. tropicalis aminotransferases,
cell wall synthesis temperature elevation,
hypokalemia,
hyperbilirubinemia
CSF cerebrospinal fluid
J. K. Roberts et al.
Table 4 Antifungal pharmacokinetics
Class Drug Recommended dosing Therapeutic range Metabolism Clearance References

Polyenes Amphotericin B deoxycholate All infants 1 mg/kg/day IV NA NA Liver, [222–227, 229–


unchanged 234, 239,
Liposomal amphotericin B All infants 5 mg/kg/day IV Uncharacterized 240, 273, 306–309]
Amphotericin B lipid complex All infants 5 mg/kg/day IV
Amphotericin B colloidal All Infants 5 mg/kg/day IV
Antimicrobial/Antiviral PK–PD in Neonates and Infants

dispersion
Azoles Fluconazole Loading 25 mg/kg IV AUC [400 mgh/L NA Kidney, [243–252, 254–257,
dose unchanged 265–267, 273, 310,
Maintenance 12 mg/kg/day 311]
IV
Voriconazole Loading 7 mg/kg q12 h Plasma 1–5 lg/L Liver—CYP2C19 Liver
dose IV concentrations
Therapeutic monitoring for
maintenance
Echinocandins Caspofungin Loading 70 mg/m2 IV NA Liver—hydrolysis and N- Liver [279–297, 312–315]
dose acetylation
Maintenance 50 mg/m2/day
IV
Micafungin Neonates 10 mg/kg/day
IV
Infants 1–4 mg/kg/day
IV
AUC area under the plasma concentration–time curve, CYP cytochrome P450, IV intravenous, NA not available, q12 h every 12 hours
593
594 J. K. Roberts et al.

amphotericin B in adults, children, and neonates [240]. a-demethylation, inhibiting ergosterol biosynthesis and cell
Neonatal serum concentrations were reported to be sig- membrane repair [243]. Prophylactic use of fluconazole has
nificantly lower than in adults, which the authors attributed contributed to increased resistance of C. glabrata and C.
to a higher Vd. krusei [244–247], and has decreased its use as a primary
The first population pharmacokinetic study of ampho- antifungal agent. Conversely, voriconazole has activity
tericin B lipid complex in neonates enrolled 28 neonates. against a broad spectrum of fungi, including C. glabrata and
Blood serum data were fitted with a one-compartment C. krusei [248–250]. Resistance to triazoles is usually
model with additive error. Clearance and Vd were scaled caused by an alteration in ergosterol synthesis, most com-
allometrically. Mean serum blood concentrations did not monly seen with changes to delta desaturase [251]. High
significantly differ from values reported in the adult liter- concentrations of triazoles have been found in the kidney
ature and a non-compartmental analysis determined a mean and liver [252], with close to 80 % of plasma concentrations
t‘ of 395 h. High concentrations of drug were reported in in the CSF and vitreal humor [253]. The most common
the urine, but very low concentrations in the CSF, demon- adverse effects of fluconazole are gastrointestinal irritation
strating that the lipid formulation did not penetrate the CSF. and an elevation of liver enzymes [254]. Voriconazole
A retrospective data analysis of infants compared studies have also documented abnormal vision in addition
amphotericin B deoxycholate and amphotericin B lipid to elevated hepatic transaminases [255–257].
products, along with fluconazole, an azole antifungal drug
[241]. Infants who were treated with amphotericin B lipid 3.2.2 Pharmacokinetics
products had a higher mortality than those treated with
amphotericin B deoxycholate or fluconazole, which the Fluconazole is a first-generation triazole. The first study to
authors attributed to several factors, including inadequate evaluate fluconazole pharmacokinetics in infants was
penetration of products in the kidneys or inappropriate published by Saxen et al. [258]. Twelve pre-term neonates
dosing in pre-term infants. This finding may suggest that were enrolled. During the first two weeks of life their Vd
alternative antifungal classes with improved safety profiles increased from 1.18 to 2.25 L/kg, their clearance increased
should be considered for neonates with invasive fungal from 0.011 to 0.031 L/kg/h, and their t‘ decreased from
infections. However, such a study suffers from many 88.6 to 55.2 h. These parameters are consistent with
challenges that are common to pediatric data: retrospective physiological changes that occur in the early postnatal
design, no control of severity of illness, and potential for period. The authors suggested that at 6 mg/kg, patients
non-random selection of treatment. should be dosed at least every other day and possibly every
third day.
3.1.3 Clinical Implications and Future Directions Nahata et al. [259] compared enteral to IV dosing of
fluconazole in pre-term neonates. The pharmacokinetic
Historically, given the decreased cost, similar efficacy, parameters were similar between the two dosing methods,
safety, and tolerance compared with liposomal formula- suggesting that enteral and IV dosing were comparable. A
tions, amphotericin B deoxycholate is recommended as the smaller study of three neonates completed by Wenzl et al.
drug of choice for invasive candidal infections in neonates, [260] came to the same conclusion.
dosing at 1 mg/kg/day IV [241]. However, those infants A population pharmacokinetic study by Wade et al.
with existing renal disease or life-threatening infections [261] evaluated fluconazole pharmacokinetics in 55
warranting increased doses may benefit from liposomal infants. A one-compartment model best fit the data, with
formulations [242], which are recommended to be dosed at significant covariates of weight, GA at birth, PNA, and
5 mg/kg/day IV. Unfortunately, limited data exist on the serum creatinine on clearance and weight on Vd. Clearance
pharmacokinetics and pharmacodynamics of lipid-based doubled during the first month of life. This model sug-
formulations of amphotericin B in neonates, complicating gested that dosing of fluconazole will change based on GA
dosing recommendations. Further studies are needed to at birth and PNA due to the role kidney maturity plays in
improve our knowledge of their adverse effect profiles as clearance. This same study population was then used to
the cost of these agents continues to decrease. determine the optimal dosing regimen for neonates [262].
The authors concluded that a dose of 12 mg/kg/day should
3.2 Azoles: Fluconazole and Voriconazole achieve an AUC [400 mgh/L. However, it was noted that
it took 5 days for plasma concentrations to reach the
3.2.1 Pharmacodynamics desired AUC [261–263].
A follow-up study of eight infants evaluated a loading
The mechanism of action for triazoles is to inhibit fun- dose of 25 mg/kg followed by a maintenance dose of
gal cytochrome P450 (CYP)-mediated lanosterol 14 12 mg/kg/day [264]. A median AUC of 479 mgh/L was
Antimicrobial/Antiviral PK–PD in Neonates and Infants 595

achieved after the first 24 h. Five of the eight infants tolerability of fluconazole, as well as the long t‘ and
achieved an AUC [400 mgh/L after the first 24 h, which increased concentration in skin, mucosal, urine, and CSF,
suggests a loading dose achieves the therapeutic target makes fluconazole a strong candidate for a prophylactic
more rapidly than a maintenance dose of 12 mg/kg/day. antifungal [253]. Given high rates of invasive fungal
Voriconazole is a second-generation triazole that infections, as well as the high mortality associated with late
exhibits high pharmacokinetic variability in pediatric treatment, antifungal prophylaxis may be useful in extre-
patients and is frequently reserved for treatment of severe mely premature newborns [276]. Several randomized
fungal infections, such as aspergillosis [265]. It can be controlled trials have evaluated the utility and safety of
administered orally with high bioavailability, although IV fluconazole prophylaxis. A recent Cochrane review showed
administration is preferred in neonates. Voriconazole is prophylactic antifungal therapy reduces the incidence of
extensively metabolized by CYP2C19 and requires dose invasive fungal infections in VLBW infants [277]. How-
adjustments in patients with hepatic failure or with genetic ever, the results must be interpreted cautiously as the
polymorphisms [266, 267]. Neonatal pharmacokinetic data incidence of invasive fungal infection was much higher in
are scarce, potentially owing to reports of visual distur- the control groups than large cohort studies have shown (16
bances in older children and adults, which may adversely vs. \5 %). Also, meta-analysis of these studies did not
affect retinal development [255]. In addition, the require- show any significant effect on mortality [278]. Further
ment to study voriconazole in patients \24 months has study is required to evaluate the effect of antifungal pro-
been waived by the FDA [268]. However, studies and case phylaxis on long-term outcomes, including neurodevelop-
reports have demonstrated that younger children require mental outcomes and patterns of resistance.
higher oral doses due to increased hepatic clearance by Data are sparse regarding voriconazole use in the neo-
CYP2C19 than adults, with children B3 years of age natal and infant populations and are primarily limited to
demonstrating variable pharmacokinetics, further support- anecdotal case reports [271]. However, several case reports
ing the importance of voriconazole TDM [269–272]. have emphasized the need to perform TDM for vorico-
A case report evaluating a single pre-term infant given nazole, owing to its marked variability and the need to
9 mg/kg every 12 h reported a Vd of 1.045 L/h/kg, clear- maintain effective concentrations. Additional studies are
ance of 0.240 L/h/kg, and a t‘ of 3.0 h [271]. The authors needed in neonates and infants to identify dosing algo-
found that an increase in dose was required to achieve rithms that may allow voriconazole to be used more safely
trough concentrations [1 lg/mL due to weight-based and effectively.
dosing not accurately predicting serum concentrations. In
addition, the only clinical study published to date that 3.3 Echinocandins: Caspofungin and Micafungin
solely evaluated neonatal patients was a retrospective study
of 17 patients [257]. Patients were first administered 3.3.1 Pharmacodynamics
amphotericin B, but due to lack of response in treatment,
the infants were switched to voriconazole. The study used a The mechanism of action for echinocandins is to inhibit the
loading dose of 4–6 mg/kg twice daily, followed by a (1,3)-b-D-glucan synthase enzyme, more specifically the
maintenance dose of 2–3 mg/kg twice daily for at least FKS1 gene product, Fsk1p, which disturbs cell wall syn-
21 days. Of those treated, 11 patients were cured. Because thesis [279–281]. This class of drugs has broad-spectrum
of the retrospective data, additional endpoints (Vd, clear- activity against Candida and Aspergillus species [282,
ance, AUC, etc.) were not evaluated in this study. 283]. Clinical resistance is rare for echinocandins, though it
has been shown that mutations in the FKS1 gene confer
3.2.3 Clinical Implications and Future Directions resistance [284–287]. Echinocandins are highly permeable
to all tissues, including the lung, liver, and spleen [288].
Recommendations by the Infectious Diseases Society of Rare adverse effects of micafungin include elevated ala-
America (IDSA) for fluconazole suggest a loading dose of nine/aspartate aminotransferases, temperature elevation,
25 mg/kg IV, followed by 12 mg/kg/day in neonates [273]. hypokalemia, and hyperbilirubinemia [289–292]. No
Fluconazole shows similar efficacy to amphotericin B for adverse effects due to caspofungin exposure have been
the treatment of invasive Candida infections and has also reported in neonates [293–296].
been shown to significantly reduce length of therapy in pre-
term infants [274, 275]. However, its decreased activity 3.3.2 Pharmacokinetics
against C. krusei and C. glabrata has limited its usefulness
as a primary antifungal agent. Caspofungin was approved by the FDA in 2001 for use in
Fluconazole has been studied extensively as a prophy- adults, and in 2008 in children 3 months of age and older.
lactic antifungal agent. The ease of administration and Early studies by Merck and Co. Inc. noted a decrease in
596 J. K. Roberts et al.

trough concentrations when caspofungin was administered 3.3.3 Clinical Implications and Future Directions
concomitantly with dexamethasone [297]. The only study
to evaluate caspofungin pharmacokinetics in only neonates Since the first prospective trials of echinocandins in the
and infants \3 months was published in 2009 [293]. This early 2000s, their use has continued to increase. This is
study enrolled 18 infants and Cmax values were similar to likely related to their comparable efficacy and improved
those in adults. It was suggested that pre-term neonates and safety profile as compared with other classes of systemic
infants may be dosed at 25 mg/m2, which is lower than the antifungals [304]. However, the majority of these studies
currently recommended dosing regimen (Table 4); how- have been performed in older children and adults. Data
ever, there were not enough infants enrolled to issue regarding the efficacy and pharmacokinetic parameters of
definitive dosing recommendations. All other caspofungin these newer treatments in neonates are limited and almost
pharmacokinetic studies in infant populations have also exclusively represented by micafungin, which may be
included older pediatric patients. It is worth mentioning attributed to its high spectrum of activity [305]. However,
that studies including patients 3–24 months [298] or the appropriate dose of micafungin for neonates and infants
3 months to 17 years [299] have achieved pharmacokinetic is still under debate and current clinical trials are trying to
results that were similar to those reported by Saez-Llorens address this issue. Despite these limitations, echinocandins
et al. [293]. appear to have a favorable safety profile in neonates and
Micafungin, a more recently developed echinocandin, young infants. The echinocandins also may have utility in
was approved for adult use by the FDA in 2005 and has treating the youngest patients with resistant fungal organ-
received approval for pediatric patients C4 months of age isms or biofilms that resist penetration by other antifungals.
in 2013. In addition, marketing authorization was obtained Importantly, drug–drug interactions, the appropriate phar-
in Europe in 2008. macodynamic exposure–response relationships, and opti-
A study completed in 2006 by Heresi et al. [291] initially mal dosing of the echinocandins remain to be elucidated in
enrolled 18 pre-term neonates and found that ELBW neo- the neonatal and young infant populations. The higher
nates had a nearly twofold greater clearance than their lar- dosing required for neonates, as compared with adults and
ger counterparts. However, adding seven more infants to the older children, focuses attention on the uniqueness of this
low birthweight group decreased the large discrepancy in population and the need for further study to fully establish
clearance between the two groups. Comparing the clearance clinical utility without compromising safety. Nonetheless,
data from the pre-term neonates with older children and based on the limited safety and efficacy data to date and
adults showed a decrease in clearance of micafungin as the ongoing studies, echinocandins are on a trajectory to
patient population increased in age [300, 301]. This become key agents in the antifungal arsenal for treating
decrease in clearance due to age has been hypothesized to invasive fungal infections in neonates and young infants.
be attributed to changes in protein binding or body com-
position [291, 302]. The authors predicted that a 5–7 mg/kg
dose in infants [1,000 g would be sufficient, though the 4 Antivirals
approved dosing is now 2 mg/kg in infants [4 months.
A pharmacokinetic study completed by Smith et al. Neonatal herpes simplex virus (HSV) infections occur in 1
[289] evaluated high-dose micafungin among 12 pre-term of 3,200 deliveries in the USA [316], of which two-thirds
neonates. This study did not find any differences between develop some form of CNS disease [317]. Eighty-five per-
ELBW patients and those above 1,000 g, as suggested by cent of cases occur as a consequence of peripartum trans-
Heresi et al. [291]. As compared with adults, ELBW mission, whereas 10 % are postnatally acquired, and 5 %
infants required three times the dose to achieve the same are acquired in utero [318]. The risk of neonates contracting
systemic exposure (15 mg/kg/dose compared with 5 mg/ HSV is decreased through cesarean delivery, prior to
kg/dose) [289], demonstrating that clearance for micafun- maternal infection with antibody production, and with
gin is higher in pre-term neonates than in adults. maternal antiviral treatment in the third trimester [317].
The most recent study evaluating neonates included 13 HSV can be divided into three clinical representations: skin,
infants and aimed to determine what doses were appro- eye, and/or mouth disease (SEM); CNS disease; or dis-
priate to achieve serum concentrations that would provide seminated, systematic disease [317]. A guanosine analog
CNS protection [290]. The authors concluded that low that is commonly used to treat HSV is acyclovir (pharma-
birthweight neonates (\1,000 g) dosed at 7 mg/kg/day and codynamics in Table 5; pharmacokinetics in Table 6).
higher birthweight neonates ([1,000 g) dosed at 10 mg/kg/ Cytomegalovirus (CMV) infection is the most common
day provided serum concentrations that are predicted to congenital infection in the developed world, affecting about
yield therapeutic CNS concentrations. This was supported 0.6 % of births in the USA [319]. Of these infants, 7–10 %
by a study by Hope et al. [303]. will have clinically evident symptoms at birth with the
Antimicrobial/Antiviral PK–PD in Neonates and Infants 597

Table 5 Antiviral pharmacodynamics


Class Drug Viral activity Mechanism of Mechanism of Tissue Adverse events References
action resistance penetration

Guanosine Acyclovir Herpes simplex, Inhibits viral Mutations in viral Kidney, Neutropenia, renal [317, 322–
analogs varicella zoster DNA thymidine lung, liver, dysfunction 327]
polymerase kinase or viral heart, skin
activity, chain DNA vesicles,
terminator polymerase CSF
Ganciclovir Cytomegalovirus Inhibits viral Mutations in Aqueous Neutropenia, [317, 342–
[ herpes simplex DNA phosphatase or humor, thrombocytopenia, 356,
polymerase viral DNA subretinal anemia 372]
activity, slows polymerase fluid, CSF,
elongation brain
Valganciclovir Herpes simplex, Inhibits viral Mutations in Aqueous Neutropenia, [121, 317,
cytomegalovirus, DNA phosphatase or humor, anemia, increased 357–
varicella zoster, polymerase viral DNA subretinal ALT, 363]
Epstein–Barr activity, slows polymerase fluid, CSF, hyperbilirubinemia
elongation brain
ALT alanine aminotransferase, CSF cerebrospinal fluid

Table 6 Antiviral pharmacokinetics


Class Drug Recommended Metabolism Clearance Other References
dosing

Guanosine Acyclovir 20 mg/kg/q8 h IV NA Kidney Low oral bioavailability [317, 322–327]


Analogues Ganciclovir 6 mg/kg/q12 h IV NA Kidney Low oral bioavailability [317, 342–356, 372]
Valganciclovir 15 mg/kg/q12 h oral Serum and liver— Kidney Dosed based on age and [121, 317, 357–363]
de-esterification for renal function, pro-drug
activation to ganciclovir
IV intravenous, NA not available, qx h every x hours

majority of these infants having evidence of CNS involve- which is phosphorylated by viral thymidine kinase, and
ment [320]. Approximately half of the symptomatic CMV- subsequently polyphosphorylated by host enzymes [322].
infected infants and nearly 14 % of asymptomatic infants The activated form of acyclovir inhibits viral DNA poly-
will go on to have permanent sequelae, including hearing merase activity and also acts as a chain terminator with 100
loss, mental retardation, learning difficulties, developmental times more affinity for viral than cellular polymerases [322,
delays, chorioretinitis, and optic nerve atrophy [321]. Addi- 323]. The most common modes of resistance to acyclovir are
tionally, the mortality rate from congenital CMV infection is mutations in the viral thymidine kinase gene or mutations in
5–10 %, usually related to non-CNS disease. Most neonatal viral DNA polymerase [324]. A common adverse event seen
cases of CMV are contracted in utero, but transmission can in acyclovir treatment is bone marrow suppression, specif-
also occur during delivery or postnatally through breast milk ically neutropenia, which has been identified with an inci-
[317]. Two guanosine analogs commonly used to treat these dence from 20–40 % of infants [325, 326]. In a recent
infections include ganciclovir and valganciclovir (pharma- pharmacokinetic study, only one adverse event of elevated
codynamics in Table 5; pharmacokinetics in Table 6). serum creatinine was reported in a single infant and no
episodes of neutropenia were reported [327]. Adverse events
4.1 Guanosine Analogs: Acyclovir of bone marrow suppression and elevated serum creatinine
typically resolve upon discontinuation of acyclovir [317].
4.1.1 Pharmacodynamics
4.1.2 Pharmacokinetics
The most common antiviral used to treat HSV and varicella
zoster virus (VZV) infections in neonates and infants is The first IV pharmacokinetic studies evaluating acyclovir
acyclovir (also known as aciclovir), which has been in in infants were published in 1982 [328, 329]. These studies
clinical use for over 30 years [317]. Originally introduced as evaluated mean Cmax and trough concentrations as well as
acycloguanosine, acyclovir is a deoxyguanosine analog, t‘ values for acyclovir in neonatal patients. At doses of 5,
598 J. K. Roberts et al.

10, and 10 mg/kg, the mean Cmax was 30.0, 61.2, and However, more research needs to be completed to confirm
86.1 lg/mL, respectively. The mean t‘ for all doses was these findings.
3.78 h, with an overall mean recovery of 65 % in the urine. The benefits, risks, and cost effectiveness of starting
A study completed 10 years later by Englund et al. [330] acyclovir empirically in other situations is also not clear
further defined the neonatal pharmacokinetics by including [338–340]. Some experts feel that asymptomatic HSV-
pre-term infants and infants with kidney failure. The exposed neonates only require routine prophylactic acy-
authors reported a significant influence of serum creatinine clovir if the maternal HSV infection is a primary infection
levels on clearance in their two-compartment model. with active lesions at the time of delivery. Recurrent HSV
Infants with kidney failure had a t‘ three times longer than infection with active lesions at the time of delivery results
those with normal renal function. Therefore, infants with in a low rate of transmission and some infectious disease
decreased kidney function require dosing adjustments to experts consider prophylactic acyclovir to be unnecessary
avoid acyclovir accumulation. unless additional risk factors for transmission exist (pro-
The most recent IV acyclovir population pharmacoki- longed rupture of membranes, prematurity, scalp electrodes
netic study was in pre-term and term infants with normal or skin abrasions). Similarly, acyclovir is indicated for
kidney function, published by Sampson et al. [331]. Their VZV infection in neonates, although no clinical trials have
final one-compartment model found that PMA and weight evaluated the risks, benefits, and cost of this treatment
were significantly correlated with increased clearance. [341].
Kidney maturation plays a key role in acyclovir accumu-
lation in the neonatal population as demonstrated by a 4.5- 4.2 Guanosine Analogs: Ganciclovir
fold increase in clearance from 25 to 41 weeks. This study and Valganciclovir
also aimed to determine if the recommended 20 mg/kg/
dosage every 8 h was sufficient to achieve a CSF con- 4.2.1 Pharmacodynamics
centration of C1 mg/L, which is required to effectively
exceed the half-maximal inhibitory concentration (IC50) A homolog of acyclovir, ganciclovir is a nucleoside analog
[332]. To study this, two groups were used: one group of of guanosine and was the first antiviral drug to be effective
patients was dosed following the recommended 20 mg/kg against CMV [342, 343]. In addition, ganciclovir is also
every 8 h and another group was dosed according to their effective against herpes viruses and Epstein–Barr virus
GA and PMA. The results of the study showed that all [344–347]. Similar to acyclovir, ganciclovir has to be
infants averaged around 2 mg/L in the CSF, suggesting phosphorylated to ganciclovir triphosphate, which can
they were all above the desired IC50. In addition, they inhibit viral DNA polymerases [348]. The product of the
proposed a new dosing regimen based on PMA: 20 mg/kg CMV UL97 gene, phosphotransferase, phosphorylates
every 12 h for infants \30 weeks, 20 mg/kg every 8 h in ganciclovir for incorporation into DNA and consequently
infants 30 to\36 weeks, and 20 mg/kg every 6 h in infants slows its elongation, disrupting cellular function [343, 348–
36–41 weeks. 351]. The primary mode of resistance to ganciclovir is a
mutation in the UL97 gene preventing phosphorylation of
4.1.3 Clinical Implications and Future Directions ganciclovir [352]. These mutations account for 95 % of
ganciclovir-resistant viral strains [352]. The minority of
Early recognition of HSV infection and prompt initiation of resistance to ganciclovir is caused by mutations in the viral
acyclovir treatment improves mortality and long-term DNA polymerase, encoded by UL54, which prohibits the
sequelae [325, 333–335]. The standard dose of acyclovir binding of ganciclovir [352].
for confirmed HSV infection is 20 mg/kg/dose every 8 h Neutropenia is the most common adverse event seen
for at least 14 days [317, 336]. However, the recent study with ganciclovir [353], with an incidence of 40 % in
by Sampson et al. [331] found that PMA could be used to patients [354] while one in seven patients require dosage
improve pharmacodynamic target achievement. The effect adjustments [355]. In a safety and tolerability study con-
of using a PMA-based dosing regimen on mortality and ducted in neonates, dosing was decreased by 50 % if the
long-term morbidities has not been established or validated absolute neutrophil count fell below 500 cells/lL or
and future work is needed to confirm this suggested dosing platelets below 50,000/lL [353]. In this study, 38 %
regimen. (n = 16) of patients presented with this adverse effect; of
It is also unknown whether long-term suppression with those, 19 % (n = 3) had to discontinue treatment with the
acyclovir in HSV infected neonates prevents CNS recur- drug because of persistent neutropenia. Neutropenia did not
rence. Studies have shown that using oral acyclovir at seem to be dose dependent in neonates, although it does
300 mg/m2 per dose three times per day for 6 months seem to be dose dependent in adults [354]. Similarly, a
appears to improve neurodevelopmental outcomes [337]. study published in 2003 [356] reported that 63 % (n = 19)
Antimicrobial/Antiviral PK–PD in Neonates and Infants 599

of infants who received ganciclovir had neutropenia; 14 elimination, the effect of renal function on clearance, and
required dose reductions and four discontinued treatment. the relationship between increased weight and Vd.
In each case, neutropenia was reversed upon discontinua- Most of the pharmacokinetic studies evaluating val-
tion of the drug. However, this can take a week or more, ganciclovir have included IV dosing with ganciclovir either
increasing the risk for increased infection in these patients. before or after oral valganciclovir administration [362, 367,
Additionally, thrombocytopenia (38 %) and anemia (2 %) 368]. The first pharmacokinetic study conducted by Galli
have been documented in infants while being treated with et al. [367] evaluated eight infants. Patients were first
ganciclovir [353]. administered a week-long course of IV ganciclovir therapy
To resolve the oral bioavailability issues common with and were then transitioned to oral valganciclovir. The first
earlier-generation guanosine analogs, an L-valine ester pro- four infants enrolled received 15 mg/kg/day of valganciclo-
drug of ganciclovir named valganciclovir was developed. vir, but they failed to achieve adequate plasma concentra-
This moiety increased bioavailability to about 60 % [357]. tions. Therefore, the next four infants received 15 mg/kg
Valganciclovir is transported in the intestine by a peptide twice a day. The infants who received twice-daily dosing had
transporter, rapidly converted to ganciclovir in the liver Cmax comparable with IV doses of ganciclovir, demonstrat-
and intestines by esterase activity, and follows the same ing that oral twice-daily dosing of valganciclovir was as
mechanism of action as ganciclovir [121]. Valganciclovir effective as IV ganciclovir. These findings were later cor-
is used to treat CMV, VZV, Epstein–Barr virus, and HSV roborated by Lombardi et al. [363]. A larger study completed
[121]. Adverse events reported for infants administered by Acosta et al. [368] reported results of two trials that
valganciclovir are inconsistent, with some reporting no employed different study designs. The first study had a rigid
adverse events [358–360], and others neutropenia [361, study design that resulted in the consistent refusal of study
362], anemia, and, rarely, hyperbilirubinemia, increased participation. As a result, only five patients were enrolled.
ALT, and thrombocytopenia [362, 363]. The second version of the study enrolled 19 infants that
began oral valganciclovir therapy for one dose, followed by
4.2.2 Pharmacokinetics IV ganciclovir for 2 weeks. At completion of their 2-week
course of ganciclovir, patients began a 4-week course of oral
To date, there have been three pharmacokinetic studies valganciclovir at home. Three dosing regimens were evalu-
published on ganciclovir pharmacokinetics that have ated in the second version of the study (14, 16, and 20 mg/
focused on or enrolled infants and neonates [353, 364, 365]. kg), all of which were well-described by a one-compartment
The first study published by Trang et al. [364] studied the model, and found bodyweight to be the most important
pharmacokinetics of ganciclovir in neonates randomized to covariate. The oral formulation of valganciclovir achieved
two IV dosing groups: 4 or 6 mg/kg every 12 h for 6 weeks. the same pharmacokinetic parameters as IV ganciclovir,
The data were fit to a single-compartment open model including approximately 54 % oral bioavailability. Kimber-
assuming zero-order input and first-order elimination. There lin et al. [362] evaluated the same pharmacokinetic data from
was no significant difference between Vd, clearance, t‘, and these infants and found that bioavailability increased by
Cmax in each dosing group when normalized to bodyweight. 32 % for oral valganciclovir in the first version of the study,
In addition, the values were similar to those reported in resulting in an unchanged AUC from time zero to 12 h
adults [366]. Individual values of Vd and the Vd at steady (AUC12), as compared to IV ganciclovir, which demon-
state (Vss) for each patient varied widely and increased with strated a 41 % decrease in AUC12, corresponding to a 73 %
increasing bodyweight. Ganciclovir was found to be pri- increase in clearance during the first 6 weeks of life. This
marily excreted unchanged in the urine and clearance was finding reveals that oral valganciclovir does not exhibit
reported to increase with age. A similar study by Zhou et al. pharmacokinetic properties similar to other antivirals and
[365] reanalyzed the same data using a population phar- suggests that valganciclovir should be considered for the
macokinetics approach and calculated similar results for Vd prolonged oral treatment of congenital CMV infection.
and clearance, but also evaluated 13 covariates that could
have an impact on ganciclovir pharmacokinetics. The study 4.2.3 Clinical Implications and Future Directions
concluded that serum creatinine affected clearance and
bodyweight influenced Vd, supporting suggestions made by In randomized clinical trials, ganciclovir has not been
Trang et al. [364]. A larger study published by Whitley et al. demonstrated to significantly reduce mortality in neonates
[353] included data from Zhou et al. [365] and Trang et al. with symptomatic CMV disease [356]. However, clinical
[364] as well as additional neonates (totaling 42 patients) trials in symptomatic CMV-positive neonates have dem-
receiving ganciclovir IV and reported results consistent onstrated benefit with ganciclovir in decreasing sensori-
with earlier published reports, including the use of a one- neural hearing loss and lower rates of neurodevelopmental
compartment model with zero-order input and first-order delay [369, 370]. Ongoing clinical trials are currently
600 J. K. Roberts et al.

investigating whether long-term therapy with ganciclovir substantially reduce this barrier to conducting clinically
and/or valganciclovir (6 months) is superior to 6 weeks of important studies in this vulnerable population.
ganciclovir and/or valganciclovir for neonates with symp- The data presented in this review demonstrate that
tomatic CMV disease (NCT00466817). In addition, it is though a considerable amount of research has been con-
currently unknown whether treatment of asymptomatic ducted, the pharmacokinetics and pharmacodynamics of
neonates with CMV infection reduces their risk of devel- several antimicrobial agents are poorly characterized in
oping long-term neurological and auditory sequelae [371]. neonates and infants. Tragically, many of these agents are
Multicenter, longitudinal studies are needed to address this commonly used in routine clinical practice. As such, fur-
pressing clinical question. ther research is needed to decrease the gap between our
understanding of adult, pediatric, infant, and neonate
pharmacokinetics to develop effective and safe dosing
5 Conclusion regimens.

Historically, neonates and infants have not been included in Acknowledgments None.
clinical trials. Continued research has shown that these Funding JKR is supported by the Pharmacotherapy Subspecialty
patients are not miniature adults, nor are they small children. Award from the Primary Children’s Medical Center Foundation.
Their pharmacokinetic and pharmacodynamic parameters RMW is supported by NIH Grants: 1 R01 HD070795-01A1 and 5
differ from those of adults and even older pediatric popu- R01 HD060559-05.
lations. As a consequence of these findings, recent legisla-
Transparency declarations The University of Utah receives
tion including the Best Pharmaceuticals for Children Act, reimbursement for the conduct of a clinical trial involving micafun-
passed as part of the FDA Safety and Innovation Act in 2012, gin. RMW receives no direct payment. All other authors declared no
and the European directive 1901/2006 on medicinal pro- conflicts of interest.
ducts for pediatric use requires new drugs to be studied in
Contributors JKR, CS, JEC, JS, MGS, RMW, and CMTS wrote
newborns, unless the studies are either deferred or waived. the initial draft of the review. All authors also contributed to the
The National Pediatric Research Network Act will encour- reviewing and finalization of the document.
age more research of pediatric populations in general.
Despite the improvements in pediatric drug research, the
study of drugs in neonates is sparse and what little data are
available often does not reflect clinical use [1]. As much of References
the low-hanging fruit has already been harvested, to advance
the field in the twenty-first century multicenter collabora- 1. Laughon MM, Avant D, Tripathi N, Hornik CP, Cohen-Wol-
tions will be essential. To this end, central institutional kowiez M, Clark RH, et al. Drug labeling and exposure in
review boards (IRBs) and standing trial protocols may be neonates. JAMA Pediatr. 2014;168:130–6.
2. Ward RM, Kern SE. Clinical trials in neonates: a therapeutic
useful in further defining the pharmacokinetics and phar- imperative. Clin Pharmacol Ther. 2009;86(6):585–7.
macodynamics of antibacterials, antifungals, and antivirals 3. Food and Drug Administration Safety and Innovation Act.
that are used in neonatal medicine. Individual institutions Public Law No. 112–144, Session No. 112. 2012;933.
can also play an important role by leveraging their retro- 4. Bizzarro MJ, Raskind C, Baltimore RS, Gallagher PG. Seventy-
five years of neonatal sepsis at Yale: 1928–2003. Pediatrics.
spective electronic medical record data to identify clinical 2005;116(3):595–602.
characteristics, patient co-morbidities, and adverse events 5. Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz
that impact the safety and effectiveness of many commonly SR, Vohr B, et al. Neurodevelopmental and growth impairment
prescribed drugs. As many antimicrobials are no longer on- among extremely low-birth-weight infants with neonatal infec-
tion. JAMA. 2004;292(19):2357–65.
patent, there is little financial incentive for drug makers to 6. Camacho-Gonzalez A, Spearman PW, Stoll BJ. Neonatal
sponsor new pediatric and neonatal clinical trials. Conse- infectious diseases: evaluation of neonatal sepsis. Pediatr Clin
quently, the responsibility for improving use of these drugs North Am. 2013;60(2):367–89.
rests on the shoulders of clinicians and biomedical 7. Ward RM, Lugo RA. Drug therapy in the newborn. In: Mac-
Donald MG, Seshia MM, Mullett MD, editors. Avery’s neona-
researchers. To successfully meet this challenge, innovative tology: pathophysiology and management of the newborn. 6th
approaches may be needed, including analytical techniques ed. Philadelphia: Lippincott Williams and Wilkins; 2005.
that make use of dried blood spots and/or scavenged blood p. 1507–56.
samples. An inherent challenge in conducting neonatal 8. Gardner P. Antimicrobial drug therapy in pediatric practice.
Pediatr Clin North Am. 1974;21(3):617–48.
pharmacokinetic studies is the limited volume of blood that 9. Patterson JE, Zervos MJ. High-level gentamicin resistance in
may be sampled to establish drug concentrations; however, Enterococcus: microbiology, genetic basis, and epidemiology.
we are convinced that further research in this field can Rev Infect Dis. 1990;12(4):644–52.
Antimicrobial/Antiviral PK–PD in Neonates and Infants 601

10. Aurangzeb B, Hameed A. Neonatal sepsis in hospital-born course of treatment with aminoglycoside. Ther Drug Monit.
babies: bacterial isolates and antibiotic susceptibility patterns. 1991;13(3):277–80.
J Coll Physicians Surg Pak. 2003;13(11):629–32. 32. Dodge WF, Jelliffe RW, Richardson CJ, McCleery RA, Ho-
11. Bryan CS, John JF Jr, Pai MS, Austin TL. Gentamicin vs cef- kanson JA, Snodgrass WR. Gentamicin population pharmaco-
otaxime for therapy of neonatal sepsis. Relationship to drug kinetic models for low birth weight infants using a new
resistance. Am J Dis Child. 1985;139(11):1086–9. nonparametric method. Clin Pharmacol Ther. 1991;50(1):25–31.
12. Cordero L, Sananes M, Ayers LW. Bloodstream infections in a 33. Faura CC, Feret MA, Horga JF. Monitoring serum levels of
neonatal intensive-care unit: 12 years’ experience with an anti- gentamicin to develop a new regimen for gentamicin dosage in
biotic control program. Infect Control Hosp Epidemiol. newborns. Ther Drug Monit. 1991;13(3):268–76.
1999;20(4):242–6. 34. Brion LP, Fleischman AR, Schwartz GJ. Gentamicin interval in
13. D’Angio CT, McGowan KL, Baumgart S, St Geme J, Harris newborn infants as determined by renal function and postcon-
MC. Surface colonization with coagulase-negative staphylo- ceptional age. Pediatr Nephrol. 1991;5(6):675–9.
cocci in premature neonates. J Pediatr. 1989;114(6):1029–34. 35. Skopnik H, Wallraf R, Nies B, Troster K, Heimann G. Phar-
14. Friedman S, Shah V, Ohlsson A, Matlow AG. Neonatal esche- macokinetics and antibacterial activity of daily gentamicin.
richia coli infections: concerns regarding resistance to current Arch Dis Child. 1992;67(1 Spec No):57–61.
therapy. Acta Paediatr. 2000;89(6):686–9. 36. Sirinavin S, McCracken GH Jr, Nelson JD. Determining gen-
15. Toltzis P, Dul MJ, Hoyen C, Salvator A, Walsh M, Zetts L, et al. tamicin dosage in infants and children with renal failure. J Pe-
The effect of antibiotic rotation on colonization with antibiotic- diatr. 1980;96(2):331–4.
resistant bacilli in a neonatal intensive care unit. Pediatrics. 37. Reimche LD, Rooney ME, Hindmarsh KW, Gorecki DK, Re-
2002;110(4):707–11. millard AJ, Sankaran K. An evaluation of gentamicin dosing
16. Singh N, Patel KM, Leger MM, Short B, Sprague BM, Kalu N, according to renal function in neonates with suspected sepsis.
et al. Risk of resistant infections with Enterobacteriaceae in Am J Perinatol. 1987;4(3):262–5.
hospitalized neonates. Pediatr Infect Dis J. 2002;21(11): 38. Kanthawatana S, Uruwannakul K, Chotinarumon S. Gentamicin
1029–33. pharmacokinetics in Thai neonates: recommendation for a dos-
17. Phillips AM, Milner RD. Tissue concentrations of netilmicin ing guideline. J Med Assoc Thai. 1998;81(2):136–40.
and gentamicin in neonates. J Infect Dis. 1984;149(3):474. 39. Rocha MJ, Almeida AM, Afonso E, Martins V, Santos J, Leitao
18. Fisk KL. A review of gentamicin use in neonates. Neonatal F, et al. The kinetic profile of gentamicin in premature neonates.
Netw. 1993;12(7):19–23 (quiz 4-8). J Pharm Pharmacol. 2000;52(9):1091–7.
19. de Cos MA, Gomez-Ullate J, Gomez F, Armijo JA. Time course 40. Tugay S, Bircan Z, Caglayan C, Arisoy AE, Gokalp AS. Acute
of trough serum gentamicin concentrations in preterm and term effects of gentamicin on glomerular and tubular functions in
neonates. Clin Pharmacokinet. 1992;23(5):391–401. preterm neonates. Pediatr Nephrol. 2006;21(10):1389–92.
20. Craig WA. Pharmacokinetic/pharmacodynamic parameters: 41. Edgren B, Karna P, Sciamanna D, Dolanski E. Gentamicin
rationale for antibacterial dosing of mice and men. Clin Infect dosing in the newborn. Use of a one-compartment open phar-
Dis. 1998;26(1):1–10 quiz 1-2. macokinetic model to individualize dosing. Dev Pharmacol
21. McCracken GH Jr. Clinical pharmacology of gentamicin in Ther. 1984;7(4):263–72.
infants 2 to 24 months of age. Am J Dis Child. 1972;124(6): 42. Jensen PD, Edgren BE, Brundage RC. Population pharmacoki-
884–7. netics of gentamicin in neonates using a nonlinear, mixed-
22. McCracken GH Jr, Jones LG. Gentamicin in the neonatal period. effects model. Pharmacotherapy. 1992;12(3):178–82.
Am J Dis Child. 1970;120(6):524–33. 43. Weber W, Kewitz G, Rost KL, Looby M, Nitz M, Harnisch L.
23. McCracken GH, West NR, Horton LJ. Urinary excretion of Population kinetics of gentamicin in neonates. Eur J Clin
gentamicin in the neonatal period. J Infect Dis. 1971;123(3): Pharmacol. 1993;44(Suppl 1):S23–5.
257–62. 44. Garcia B, Barcia E, Perez F, Molina IT. Population pharmaco-
24. Koren G, James A, Perlman M. A simple method for the esti- kinetics of gentamicin in premature newborns. J Antimicrob
mation of glomerular filtration rate by gentamicin pharmacoki- Chemother. 2006;58(2):372–9.
netics during routine drug monitoring in the newborn. Clin 45. Stolk LM, Degraeuwe PL, Nieman FH, de Wolf MC, de Boer A.
Pharmacol Ther. 1985;38(6):680–5. Population pharmacokinetics and relationship between demo-
25. Assael BM, Gianni V, Marini A, Peneff P, Sereni F. Gentamicin graphic and clinical variables and pharmacokinetics of genta-
dosage in preterm and term neonates. Arch Dis Child. micin in neonates. Ther Drug Monit. 2002;24(4):527–31.
1977;52(11):883–6. 46. Kasik JW, Jenkins S, Leuschen MP, Nelson RM Jr. Postcon-
26. Rameis H, Popow C, Graninger W. Gentamicin monitoring in ceptional age and gentamicin elimination half-life. J Pediatr.
low-birth-weight newborns. Biol Res Pregnancy Perinatol. 1985;106(3):502–5.
1983;4(3):123–6. 47. Thomson AH, Way S, Bryson SM, McGovern EM, Kelman
27. Koren G, Leeder S, Harding E, Jacques D, MacLeod SM. AW, Whiting B. Population pharmacokinetics of gentamicin in
Optimization of gentamicin therapy in very low birth weight neonates. Dev Pharmacol Ther. 1988;11(3):173–9.
infants. Pediatr Pharmacol (New York). 1985;5(1):79–87. 48. Botha JH, du Preez MJ, Adhikari M. Population pharmacoki-
28. Pons G, d’Athis P, Rey E, de Lauture D, Richard MO, Badoual netics of gentamicin in South African newborns. Eur J Clin
J, et al. Gentamicin monitoring in neonates. Ther Drug Monit. Pharmacol. 2003;59(10):755–9.
1988;10(4):421–7. 49. Szefler SJ, Wynn RJ, Clarke DF, Buckwald S, Shen D, Schentag
29. Nakae S, Yamada M, Ito T, Chiba Y, Sasaki E, Sakamoto M, JJ. Relationship of gentamicin serum concentrations to gesta-
et al. Gentamicin dosing and pharmacokinetics in low birth tional age in preterm and term neonates. J Pediatr. 1980;97(2):
weight infants. Tohoku J Exp Med. 1988;155(3):213–23. 312–5.
30. Shahidullah M, Talukder MQ, Chowdhury AK, Ali S, Rashid A. 50. Hindmarsh KW, Nation RL, Williams GL, John E, French JN.
Serum levels of gentamicin at peak and trough in neonates and Pharmacokinetics of gentamicin in very low birth weight pre-
infants. Indian J Pediatr. 1991;58(2):217–21. term infants. Eur J Clin Pharmacol. 1983;24(5):649–53.
31. Faura CC, Garcia MR, Horga JF. Changes in gentamicin serum 51. Kildoo C, Modanlou HD, Komatsu G, Harralson A, Hodding J.
levels and pharmacokinetic parameters in the newborn in the Developmental pattern of gentamicin kinetics in very low birth
602 J. K. Roberts et al.

weight (VLBW) sick infants. Dev Pharmacol Ther. 1984;7(6): 71. DiCenzo R, Forrest A, Slish JC, Cole C, Guillet R. A gentamicin
345–56. pharmacokinetic population model and once-daily dosing algo-
52. Hayani KC, Hatzopoulos FK, Frank AL, Thummala MR, Han- rithm for neonates. Pharmacotherapy. 2003;23(5):585–91.
tsch MJ, Schatz BM, et al. Pharmacokinetics of once-daily 72. Lopez SA, Mulla H, Durward A, Tibby SM. Extended-interval
dosing of gentamicin in neonates. J Pediatr. 1997;131(1 Pt gentamicin: population pharmacokinetics in pediatric critical
1):76–80. illness. Pediatr Crit Care Med. 2010;11(2):267–74.
53. Krishnan L, George SA. Gentamicin therapy in preterms: a 73. Tiwari S, Rehan HS, Chandra J, Mathur NN, Singh V. Efficacy
comparison of two dosage regimens. Indian Pediatr. and safety of a single daily dose of gentamicin in hospitalized
1997;34(12):1075–80. Indian children: a quasi-randomized trial. J Antimicrob Che-
54. Davies MW, Cartwright DW. Gentamicin dosage intervals in mother. 2009;64(5):1096–101.
neonates: longer dosage interval–less toxicity. J Paediatr Child 74. Thingvoll ES, Guillet R, Caserta M, Dicenzo R. Observational
Health. 1998;34(6):577–80. trial of a 48-hour gentamicin dosing regimen derived from
55. Stickland MD, Kirkpatrick CM, Begg EJ, Duffull SB, Oddie SJ, Monte Carlo simulations in infants born at less than 28 weeks’
Darlow BA. An extended interval dosing method for gentamicin gestation. J Pediatr. 2008;153(4):530–4.
in neonates. J Antimicrob Chemother. 2001;48(6):887–93. 75. Knight JA, Davis EM, Manouilov K, Hoie EB. The effect of
56. Chotigeat U, Narongsanti A, Ayudhya DP. Gentamicin in neo- postnatal age on gentamicin pharmacokinetics in neonates.
natal infection: once versus twice daily dosage. J Med Assoc Pharmacotherapy. 2003;23(8):992–6.
Thai. 2001;84(8):1109–15. 76. Thomson AH, Kokwaro GO, Muchohi SN, English M,
57. Rastogi A, Agarwal G, Pyati S, Pildes RS. Comparison of two Mohammed S, Edwards G. Population pharmacokinetics of
gentamicin dosing schedules in very low birth weight infants. intramuscular gentamicin administered to young infants with
Pediatr Infect Dis J. 2002;21(3):234–40. suspected severe sepsis in Kenya. Br J Clin Pharmacol.
58. Agarwal G, Rastogi A, Pyati S, Wilks A, Pildes RS. Comparison 2003;56(1):25–31.
of once-daily versus twice-daily gentamicin dosing regimens in 77. Vervelde ML, Rademaker CM, Krediet TG, Fleer A, van Asten
infants [ or = 2500 g. J Perinatol. 2002;22(4):268–74. P, van Dijk A. Population pharmacokinetics of gentamicin in
59. Alsaedi SA. Once daily gentamicin dosing in full term neonates. preterm neonates: evaluation of a once-daily dosage regimen.
Saudi Med J. 2003;24(9):978–81. Ther Drug Monit. 1999;21(5):514–9.
60. Mercado MC, Brodsky NL, McGuire MK, Hurt H. Extended 78. Izquierdo M, Lanao JM, Cervero L, Jimenez NV, Dominguez-
interval dosing of gentamicin in preterm infants. Am J Perinatol. Gil A. Population pharmacokinetics of gentamicin in premature
2004;21(2):73–7. infants. Ther Drug Monit. 1992;14(3):177–83.
61. Lanao JM, Calvo MV, Mesa JA, Martin-Suarez A, Carbajosa 79. Paisley JW, Smith AL, Smith DH. Gentamicin in newborn
MT, Miguelez F, et al. Pharmacokinetic basis for the use of infants. Comparison of intramuscular and intravenous adminis-
extended interval dosage regimens of gentamicin in neonates. tration. Am J Dis Child. 1973;126(4):473–7.
J Antimicrob Chemother. 2004;54(1):193–8. 80. Mulhall A. Antibiotic treatment of neonates—does route of
62. Kosalaraksa P, Janthep P, Jirapradittha J, Taksaphan S, Kia- administration matter? Dev Pharmacol Ther. 1985;8(1):1–8.
tchoosakun P. Once versus twice daily dose of gentamicin 81. McCracken GH Jr, Threlkeld N, Thomas ML. Intravenous
therapy in Thai neonates. J Med Assoc Thai. 2004;87(4):372–6. administration of kanamycin and gentamicin in newborn infants.
63. Rao SC, Ahmed M, Hagan R. One dose per day compared to Pediatrics. 1977;60(4):463–6.
multiple doses per day of gentamicin for treatment of suspected 82. Watterberg KL, Kelly HW, Angelus P, Backstrom C. The need
or proven sepsis in neonates. Cochrane Database Syst Rev. for a loading dose of gentamicin in neonates. Ther Drug Monit.
2006;(1):CD005091. 1989;11(1):16–20.
64. Darmstadt GL, Hossain MM, Jana AK, Saha SK, Choi Y, Sri- 83. Semchuk W, Borgmann J, Bowman L. Determination of a
dhar S, et al. Determination of extended-interval gentamicin gentamicin loading dose in neonates and infants. Ther Drug
dosing for neonatal patients in developing countries. Pediatr Monit. 1993;15(1):47–51.
Infect Dis J. 2007;26(6):501–7. 84. Gal P, Ransom JL, Weaver RL. Gentamicin in neonates: the
65. Miranda JC, Schimmel MM, James LS, Spinelli W, Rosen TS. need for loading doses. Am J Perinatol. 1990;7(3):254–7.
Gentamicin kinetics in the neonate. Pediatr Pharmacol (New 85. Nielsen EI, Sandstrom M, Honore PH, Ewald U, Friberg LE.
York). 1985;5(1):57–61. Developmental pharmacokinetics of gentamicin in preterm and
66. Zarowitz BJ, Wynn RJ, Buckwald S, Szefler SJ. High genta- term neonates: population modelling of a prospective study. Clin
micin trough concentrations in neonates of less than 28 weeks Pharmacokinet. 2009;48(4):253–63.
gestational age. Dev Pharmacol Ther. 1982;5(1–2):68–75. 86. Isemann BT, Kotagal UR, Mashni SM, Luckhaupt EJ, Johnson
67. Alshaikh B, Dersch-Mills D, Taylor R, Akierman AR, Yusuf K. CJ. Optimal gentamicin therapy in preterm neonates includes
Extended interval dosing of gentamicin in premature neo- loading doses and early monitoring. Ther Drug Monit.
nates \/= 28-week gestation. Acta Paediatr. 2012;101(11): 1996;18(5):549–55.
1134–9. 87. Giacoia GP, Schentag JJ. Pharmacokinetics and nephrotoxicity
68. Mohamed AF, Nielsen EI, Cars O, Friberg LE. Pharmacoki- of continuous intravenous infusion of gentamicin in low birth
netic-pharmacodynamic model for gentamicin and its adaptive weight infants. J Pediatr. 1986;109(4):715–9.
resistance with predictions of dosing schedules in newborn 88. Sherwin CM, McCaffrey F, Broadbent RS, Reith DM, Medlicott
infants. Antimicrob Agents Chemother. 2012;56(1):179–88. NJ. Discrepancies between predicted and observed rates of
69. Hossain MM, Chowdhury NA, Shirin M, Saha SK, Miller-Bell intravenous gentamicin delivery for neonates. J Pharm Phar-
M, Edwards D, et al. Simplified dosing of gentamicin for macol. 2009;61(4):465–71.
treatment of sepsis in Bangladeshi neonates. J Health Popul 89. Haughey DB, Hilligoss DM, Grassi A, Schentag JJ. Two-com-
Nutr. 2009;27(5):640–5. partment gentamicin pharmacokinetics in premature neonates: a
70. Hoff DS, Wilcox RA, Tollefson LM, Lipnik PG, Commers AR, comparison to adults with decreased glomerular filtration rates.
Liu M. Pharmacokinetic outcomes of a simplified, weight-based, J Pediatr. 1980;96(2):325–30.
extended-interval gentamicin dosing protocol in critically ill 90. De Cock RF, Allegaert K, Schreuder MF, Sherwin CM, de Hoog
neonates. Pharmacotherapy. 2009;29(11):1297–305. M, van den Anker JN, et al. Maturation of the glomerular
Antimicrobial/Antiviral PK–PD in Neonates and Infants 603

filtration rate in neonates, as reflected by amikacin clearance. 109. Red Book. 2012 Report of the committee on infectious diseases.
Clin Pharmacokinet. 2012;51(2):105–17. Elk Grove: American Academy of Pediatrics; 2012.
91. Landers S, Berry PL, Kearns GL, Kaplan SL, Rudolph AJ. 110. Rao SC, Srinivasjois R, Hagan R, Ahmed M. One dose per day
Gentamicin disposition and effect on development of renal compared to multiple doses per day of gentamicin for treatment
function in the very low birth weight infant. Dev Pharmacol of suspected or proven sepsis in neonates. Cochrane Database
Ther. 1984;7(5):285–302. Syst Rev. 2011;(11):CD005091.
92. Friedman CA, Parks BR, Rawson JE. Gentamicin disposition in 111. Mohamed AF, Nielsen EI, Cars O, Friberg LE. Pharmacoki-
asphyxiated newborns: relationship to mean arterial blood netic-pharmacodynamic model for gentamicin and its adaptive
pressure and urine output. Pediatr Pharmacol (New York). resistance with predictions of dosing schedules in newborn
1982;2(3):189–97. infants. Antimicrob Agents Chemother. 2012;56(1):179–88.
93. Bravo ME, Arancibia A, Jarpa S, Carpentier PM, Jahn AN. 112. Schauf V, Deveikis A, Riff L, Serota A. Antibiotic-killing
Pharmacokinetics of gentamicin in malnourished infants. Eur J kinetics of group B streptococci. J Pediatr. 1976;89(2):194–8.
Clin Pharmacol. 1982;21(6):499–504. 113. Danelon C, Nestorovich EM, Winterhalter M, Ceccarelli M,
94. Watterberg KL, Kelly HW, Johnson JD, Aldrich M, Angelus P. Bezrukov SM. Interaction of zwitterionic penicillins with the
Effect of patent ductus arteriosus on gentamicin pharmacoki- OmpF channel facilitates their translocation. Biophys J.
netics in very low birth weight (less than 1,500 g) babies. Dev 2006;90(5):1617–27.
Pharmacol Ther. 1987;10(2):107–17. 114. Izaki K, Matsuhashi M, Strominger JL. Glycopeptide transpepti-
95. Williams BS, Ransom JL, Gal P, Carlos RQ, Smith M, Schall dase and D-alanine carboxypeptidase: penicillin-sensitive enzy-
SA. Gentamicin pharmacokinetics in neonates with patent duc- matic reactions. Proc Natl Acad Sci U S A. 1966;55(3):656–63.
tus arteriosus. Crit Care Med. 1997;25(2):273–5. 115. de Hoog M, Mouton JW, van den Anker JN. New dosing
96. Touw DJ, Proost JH, Stevens R, Lafeber HN, van Weissenbruch strategies for antibacterial agents in the neonate. Semin Fetal
MM. Gentamicin pharmacokinetics in preterm infants with a Neonatal Med. 2005;10(2):185–94.
patent and a closed ductus arteriosus. Pharm World Sci. 116. Tessin I, Trollfors B, Thiringer K, Larsson P. Ampicillin-ami-
2001;23(5):200–4. noglycoside combinations as initial treatment for neonatal sep-
97. Southgate WM, DiPiro JT, Robertson AF. Pharmacokinetics of ticaemia or meningitis. A retrospective evaluation of 12 years’
gentamicin in neonates on extracorporeal membrane oxygena- experience. Acta Paediatrica Scandinavica. 1991;80(10):911–6.
tion. Antimicrob Agents Chemother. 1989;33(6):817–9. 117. Cars O. The hidden impact of antibacterial resistance in respi-
98. Cohen P, Collart L, Prober CG, Fischer AF, Blaschke TF. ratory tract infection. Steering an appropriate course: principles
Gentamicin pharmacokinetics in neonates undergoing extracor- to guide antibiotic choice. Respir Med. 2001;95(Suppl A):S20–5
poral membrane oxygenation. Pediatr Infect Dis J. 1990;9(8): discussion S6-7.
562–6. 118. Regoes RR, Wiuff C, Zappala RM, Garner KN, Baquero F,
99. Dodge WF, Jelliffe RW, Zwischenberger JB, Bellanger RA, Levin BR. Pharmacodynamic functions: a multiparameter
Hokanson JA, Snodgrass WR. Population pharmacokinetic approach to the design of antibiotic treatment regimens. Anti-
models: effect of explicit versus assumed constant serum con- microb Agents Chemother. 2004;48(10):3670–6.
centration assay error patterns upon parameter values of genta- 119. Craig WA. The hidden impact of antibacterial resistance in
micin in infants on and off extracorporeal membrane respiratory tract infection. Re-evaluating current antibiotic ther-
oxygenation. Ther Drug Monit. 1994;16(6):552–9. apy. Respir Med. 2001;95(Suppl A):S12–9 (discussion S26-7).
100. Munzenberger PJ, Massoud N. Pharmacokinetics of gentamicin 120. Carder KR. Hypersensitivity reactions in neonates and infants.
in neonatal patients supported with extracorporeal membrane Dermatol Ther. 2005;18(2):160–75.
oxygenation. ASAIO Trans. 1991;37(1):16–8. 121. Marshall BC, Koch WC. Antivirals for cytomegalovirus infec-
101. Bhatt-Mehta V, Johnson CE, Schumacher RE. Gentamicin tion in neonates and infants: focus on pharmacokinetics, for-
pharmacokinetics in term neonates receiving extracorporeal mulations, dosing, and adverse events. Paediatr Drugs.
membrane oxygenation. Pharmacotherapy. 1992;12(1):28–32. 2009;11(5):309–21.
102. Lingvall M, Reith D, Broadbent R. The effect of sepsis upon 122. Shin HT, Chang MW. Drug eruptions in children. Curr Probl
gentamicin pharmacokinetics in neonates. Br J Clin Pharmacol. Pediatr. 2001;31(7):207–34.
2005;59(1):54–61. 123. Polin RA. Management of neonates with suspected or proven
103. Sherwin CM, Kostan E, Broadbent RS, Medlicott NJ, Reith DM. early-onset bacterial sepsis. Pediatrics. 2012;129(5):1006–15.
Evaluation of the effect of intravenous volume expanders upon 124. Boe RW, Williams CP, Bennett JV, Oliver TK Jr. Serum levels
the volume of distribution of gentamicin in septic neonates. of methicillin and ampicillin in newborn and premature infants
Biopharm Drug Dispos. 2009;30(5):276–80. in relation to postnatal age. Pediatrics. 1967;39(2):194–201.
104. Papadatos J, Kafetzis D, Papadatos C. Influence of phototherapy 125. Axline SG, Yaffe SJ, Simon HJ. Clinical pharmacology of
on the pharmacokinetics of gentamicin. J Infect Dis. antimicrobials in premature infants. II. Ampicillin, methicillin,
1982;146(4):567. oxacillin, neomycin, and colistin. Pediatrics. 1967;39(1):97–107.
105. Zenk KE, Miwa L, Cohen JL, Waffarn F, Huxtable RF. Effect of 126. Silverio J, Poole JW. Serum concentrations of ampicillin in
body weight on gentamicin pharmacokinetics in neonates. Clin newborn infants after oral administration. Pediatrics. 1973;
Pharm. 1984;3(2):170–3. 51(3):578–80.
106. Pacifici G. Clinical pharmacokinetics of penicillins, cephalo- 127. Kaplan JM, McCracken GH Jr, Horton LJ, Thomas ML, Davis
sporins, and aminoglycosides in the neonate: a review. Phar- N. Pharmacologic studies in neonates given large dosages of
maceuticals. 2010;3:2568–91. ampicillin. J Pediatr. 1974;84(4):571–7.
107. Skopnik H, Wallraf R, Nies B, Troster K, Heimann G. Phar- 128. Yoshioka H, Takimoto M, Riley HD Jr. Pharmacokinetics of
macokinetics and antibacterial activity of daily gentamicin. ampicillin in the newborn infant. J Infect Dis. 1974;129(4):
Arch Dis Child. 1992;67(1 Spec No):57–61. 461–4.
108. Dahlgren JG, Anderson ET, Hewitt WL. Gentamicin blood 129. Colburn WA, Gilbaldi M, Yoshioka H, Takimoto M, Riley HD
levels: a guide to nephrotoxicity. Antimicrob Agents Chemo- Jr. Pharmacokinetic model for serum concentrations of ampi-
ther. 1975;8(1):58–62. cillin in the newborn infant. J Infect Dis. 1976;134(1):67–9.
604 J. K. Roberts et al.

130. Driessen OM, Sorgedrager N, Michel MF, Kerrebijn KF, Her- 149. Frymoyer A, Guglielmo BJ, Wilson SD, Scarpace SB, Benet
mans J. Pharmacokinetic aspects of therapy with ampicillin and LZ, Hersh AL. Impact of a hospitalwide increase in empiric
kanamycin in new-born infants. Eur J Clin Pharmacol. pediatric vancomycin dosing on initial trough concentrations.
1978;13:449–57. Pharmacotherapy. 2011;31(9):871–6.
131. McCracken GH Jr, Ginsburg CM, Clahsen JC, Thomas ML. 150. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz
Pharmacologic evaluation of orally administered antibiotics in RJ, et al. Clinical practice guidelines by the infectious diseases
infants and children: effect of feeding on bioavailability. Pedi- society of america for the treatment of methicillin-resistant
atrics. 1978;62(5):738–43. Staphylococcus aureus infections in adults and children. Clin
132. Ginsburg CM, McCracken GH Jr, Thomas ML, Clahsen J. Infect Dis. 2011;52(3):e18–55.
Comparative pharmacokinetics of amoxicillin and ampicillin in 151. Lasky T, Greenspan J, Ernst FR, Gonzalez L. Pediatric vanco-
infants and children. Pediatrics. 1979;64(5):627–31. mycin use in 421 hospitals in the United States, 2008. PLoS
133. Sutton AM, Turner TL, Cockburn F, McAllister TA. Pharma- One. 2012;7(8):e43258.
cokinetic study of sulbactam and ampicillin administered con- 152. Kadambari S, Heath PT, Sharland M, Lewis S, Nichols A,
comitantly by intraarterial or intravenous infusion in the Turner MA. Variation in gentamicin and vancomycin dosage
newborn. Rev Infect Dis. 1986;8(Suppl 5):S518–22. and monitoring in UK neonatal units. J Antimicrob Chemother.
134. Dahl LB, Melby K, Gutteberg TJ, Storvold G. Serum levels of 2011;66(11):2647–50.
ampicillin and gentamycin in neonates of varying gestational 153. Frymoyer A, Guglielmo BJ, Hersh AL. Desired vancomycin
age. Eur J Pediatr. 1986;145(3):218–21. trough serum concentration for treating invasive methicillin-
135. Koren G. Therapeutic drug monitoring principles in the neonate. resistant Staphylococcal infections. Pediatr Infect Dis J.
National Academy of Clinical Biochemistry. Clin Chem. 2013;32(10):1077–9.
1997;43(1):222–7. 154. Le J, Bradley JS, Murray W, Romanowski GL, Tran TT, Nguyen
136. Huang NN, High RH. Comparison of serum levels following the N, et al. Improved vancomycin dosing in children using area under
administration of oral and parenteral preparations of penicillin to the curve exposure. Pediatr Infect Dis J. 2013;32(4):e155–63.
infants and children of various age groups. J Pediatr. 155. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz
1953;42(6):657–8. RJ, et al. Clinical practice guidelines by the Infectious Diseases
137. Ehrnebo M, Agurell S, Jalling B, Boreus LO. Age differences in Society of America for the treatment of methicillin-resistant
drug binding by plasma proteins: studies on human foetuses, Staphylococcus aureus infections in adults and children. Clin
neonates and adults. Eur J Clin Pharmacol. 1971;3(4):189–93. Infect Dis. 2011;52(3):e18–55.
138. Paediatric Formulary Committee. BNF for children 2011–2012. 156. Frymoyer A, Hersh AL, Coralic Z, Benet LZ, Joseph Guglielmo
London: BMJ Group, Pharmaceutical Press and RCPCH Publi- B. Prediction of vancomycin pharmacodynamics in children with
cations; 2011. invasive methicillin-resistant Staphylococcus aureus infections:
139. Noya FJD. Antibiotic usage in neonates. Semin Pediatr Infect a Monte Carlo simulation. Clin Ther. 2010;32(3):534–42.
Dis. 1998;9(4):322–9. 157. Ward RM, Allegaert K, de Groot R, van den Anker JN. Com-
140. Jacqz-Aigrain E, Zhao W, Sharland M, van den Anker JN. Use mentary: continuous infusion of vancomycin in neonates: to use
of antibacterial agents in the neonate: 50 years of experience or not to use remains the question. Pediatr Infect Dis J. Epub
with vancomycin administration. Semin Fetal Neonatal Med. 2013 Dec 30.
2013;18(1):28–34. 158. van den Anker JN. Getting the dose of vancomycin right in the
141. Pawlotsky F, Thomas A, Kergueris MF, Debillon T, Roze JC. neonate. Int J Clin Pharmacol Ther. 2011;49(4):247–9.
Constant rate infusion of vancomycin in premature neonates: a 159. Butterfield JM, Patel N, Pai MP, Rosano TG, Drusano GL,
new dosage schedule. Br J Clin Pharmacol. 1998;46(2):163–7. Lodise TP. Refining vancomycin protein binding estimates:
142. Camaione L, Elliott K, Mitchell-Van Steele A, Lomaestro B, Pai identification of clinical factors that influence protein binding.
MP. Vancomycin dosing in children and young adults: back to Antimicrob Agents Chemother. 2011;55(9):4277–82.
the drawing board. Pharmacotherapy. 2013;33(12):1278–87. 160. Rodvold KA, Everett JA, Pryka RD, Kraus DM. Pharmacoki-
143. de Hoog M, Mouton JW, van den Anker JN. Vancomycin: netics and administration regimens of vancomycin in neonates,
pharmacokinetics and administration regimens in neonates. Clin infants and children. Clin Pharmacokinet. 1997;33(1):32–51.
Pharmacokinet. 2004;43(7):417–40. 161. Stockmann C, Sherwin CM, Zobell JT, Lubsch L, Young DC,
144. Frymoyer A, Hersh AL, Benet LZ, Guglielmo BJ. Current rec- Olson J, et al. Population pharmacokinetics of intermittent
ommended dosing of vancomycin for children with invasive vancomycin in children with cystic fibrosis. Pharmacotherapy.
methicillin-resistant Staphylococcus aureus infections is inade- 2013;33(12):1288–96.
quate. Pediatr Infect Dis J. 2009;28(5):398–402. 162. Anderson BJ, Allegaert K, Van den Anker JN, Cossey V, Hol-
145. Welsh KJ, Abbott AN, Lewis EM, Gardiner JM, Kruzel MC, ford NH. Vancomycin pharmacokinetics in preterm neonates
Lewis CT, et al. Clinical characteristics, outcomes, and micro- and the prediction of adult clearance. Br J Clin Pharmacol.
biologic features associated with methicillin-resistant Staphy- 2007;63(1):75–84.
lococcus aureus bacteremia in pediatric patients treated with 163. De Cock RF, Allegaert K, Schreuder MF, Sherwin CM, de Hoog
vancomycin. J Clin Microbiol. 2010;48(3):894–9. M, van den Anker JN, et al. Maturation of the glomerular fil-
146. Marques-Minana MR, Saadeddin A, Peris JE. Population phar- tration rate in neonates, as reflected by amikacin clearance. Clin
macokinetic analysis of vancomycin in neonates. A new pro- Pharmacokinet. 2012;51(2):105–17.
posal of initial dosage guideline. Br J Clin Pharmacol. 164. Smits A, Kulo A, de Hoon JN, Allegaert K. Pharmacokinetics of
2010;70(5):713–20. drugs in neonates: pattern recognition beyond compound spe-
147. Gordon CL, Thompson C, Carapetis JR, Turnidge J, Kilburn C, cific observations. Curr Pharm Des. 2012;18(21):3119–46.
Currie BJ. Trough concentrations of vancomycin: adult thera- 165. James A, Koren G, Milliken J, Soldin S, Prober C. Vancomycin
peutic targets are not appropriate for children. Pediatr Infect Dis pharmacokinetics and dose recommendations for preterm
J. 2012;31(12):1269–71. infants. Antimicrob Agents Chemother. 1987;31(1):52–4.
148. Eiland LS, English TM, Eiland EH 3rd. Assessment of vanco- 166. Marsot A, Boulamery A, Bruguerolle B, Simon N. Vancomycin:
mycin dosing and subsequent serum concentrations in pediatric a review of population pharmacokinetic analyses. Clin Phar-
patients. Ann Pharmacother. 2011;45(5):582–9. macokinet. 2012;51(1):1–13.
Antimicrobial/Antiviral PK–PD in Neonates and Infants 605

167. Oudin C, Vialet R, Boulamery A, Martin C, Simon N. Vanco- 188. Kearns GL, Young RA. Pharmacokinetics of cefotaxime and
mycin prescription in neonates and young infants: toward a desacetylcefotaxime in the young. Diagn Microbiol Infect Dis.
simplified dosage. Arch Dis Child Fetal Neonatal Ed. 1995;22(1–2):97–104.
2011;96(5):F365–70. 189. Clark RH, Bloom BT, Spitzer AR, Gerstmann DR. Empiric use
168. de Hoog M, Schoemaker RC, Mouton JW, van den Anker JN. of ampicillin and cefotaxime, compared with ampicillin and
Vancomycin population pharmacokinetics in neonates. Clin gentamicin, for neonates at risk for sepsis is associated with an
Pharmacol Ther. 2000;67(4):360–7. increased risk of neonatal death. Pediatrics. 2006;117(1):67–74.
169. Zhao W, Kaguelidou F, Biran V, Zhang D, Allegaert K, Cap- 190. Kafetzis DA, Brater DC, Kapiki AN, Papas CV, Dellagramm-
parelli EV, et al. External evaluation of population pharmaco- aticas H, Papadatos CJ. Treatment of severe neonatal infections
kinetic models of vancomycin in neonates: the transferability of with cefotaxime. Efficacy and pharmacokinetics. J Pediatr.
published models to different clinical settings. Br J Clin Phar- 1982;100(3):483–9.
macol. Epub 2012 Aug 1. doi:10.1111/j.1365-2125.2012.04406.x. 191. Schaad UB, McCracken GH Jr, Loock CA, Thomas ML.
170. Spears RL, Koch R. The use of vancomycin in pediatrics. Pharmacokinetics and bacteriologic efficacy of moxalactam,
Antibiot Ann. 1959;7:798–803. cefotaxime, cefoperazone, and rocephin in experimental bacte-
171. Rana D, Abughali N, Kumar D, Super DM, Jacobs MR, Kumar rial meningitis. J Infect Dis. 1981;143(2):156–63.
ML. Staphylococcus aureus, including community-acquired 192. Baird-Lambert J, Doyle PE, Thomas D, Cvejic M, Buchanan N.
methicillin-resistant S. aureus, in a level III NICU: 2001 to Pharmacokinetics of cefotaxime in neonates. J Antimicrob
2008. Am J Perinatol. 2012;29(6):401–8. Chemother. 1984;13(5):471–7.
172. Venkatesh MP, Placencia F, Weisman LE. Coagulase-negative 193. Aujard Y, Brion F, Jacqz-Aigrain E, Kasse MC, Chretien P,
staphylococcal infections in the neonate and child: an update. Criqui C, et al. Pharmacokinetics of cefotaxime and desa-
Semin Pediatr Infect Dis. 2006;17(3):120–7. cetylcefotaxime in the newborn. Diagn Microbiol Infect Dis.
173. Broome L, So TY. An evaluation of initial vancomycin dosing 1989;12(1):87–91.
in infants, children, and adolescents. Int J Pediatr. 2011;2011: 194. de Louvois J, Mulhall A, Hurley R. The safety and pharmaco-
470364. kinetics of cefotaxime in the treatment of neonates. Pediatr
174. Sammons HM, Starkey E. Vancomycin use in neonates and Pharmacol (New York). 1982;2(4):275–84.
children: evidence-based practice is needed. Arch Dis Child. 195. von Hattingberg HM, Marget W, Belohradsky BH, Roos R.
2013;98(6):447–8. Pharmacokinetics of cefotaxime in neonates and children: clin-
175. Goutelle S, Neely M, Bleyzac N. Comment: assessment of ical aspects. J Antimicrob Chemother. 1980;6(Suppl A):113–8.
vancomycin dosing and subsequent serum concentrations in 196. Crooks J, White LO, Burville LJ, Speidel BD, Reeves DS.
pediatric patients. Ann Pharmacother. 2011;45(9):1171–2. Pharmacokinetics of cefotaxime and desacetyl-cefotaxime in
176. Crumby T, Rinehart E, Carby MC, Kuhl D, Talati AJ. Phar- neonates. J Antimicrob Chemother. 1984;14(Suppl B):97–101.
macokinetic comparison of nomogram-based and individualized 197. Gouyon JB, Pechinot A, Safran C, Chretien P, Sandre D, Ka-
vancomycin regimens in neonates. Am J Health Syst Pharm. zmierczak A. Pharmacokinetics of cefotaxime in preterm
2009;66(2):149–53. infants. Dev Pharmacol Ther. 1990;14(1):29–34.
177. Myers AL, Gaedigk A, Dai H, James LP, Jones BL, Neville KA. 198. Kearns GL, Jacobs RF, Thomas BR, Darville TL, Trang JM.
Defining risk factors for red man syndrome in children and Cefotaxime and desacetylcefotaxime pharmacokinetics in very
adults. Pediatr Infect Dis J. 2012;31(5):464–8. low birth weight neonates. J Pediatr. 1989;114(3):461–7.
178. Samiee-Zafarghandy S, van den Anker JN. Do we really need 199. Kafetzis DA, Brater DC, Kanarios J, Sinaniotis CA, Papadatos
continuous vancomycin infusion in neonates? Arch Dis Child. CJ. Clinical pharmacology of cefotaxime in pediatric patients.
2013;98(12):1023–4. Antimicrob Agents Chemother. 1981;20(4):487–90.
179. Patel AD, Anand D, Lucas C, Thomson AH. Continuous infusion 200. Begue P, Safran C, Quiniou F, Lasfargues G, Quinet B. Com-
of vancomycin in neonates. Arch Dis Child. 2013;98(6):478–9. parative pharmacokinetics of four new cephalosporins: moxa-
180. Chamberlain J, Coombes JD, Dell D, Fromson JM, Ings RJ, lactam, cefotaxime, cefoperazone and ceftazidime in neonates.
Macdonald CM, et al. Metabolism of cefotaxime in animals and Dev Pharmacol Ther. 1984;7(Suppl 1):105–8.
man. J Antimicrob Chemother. 1980;6(Suppl A):69–78. 201. Ahsman MJ, Wildschut ED, Tibboel D, Mathot RA. Pharma-
181. Ings RM, Reeves DS, White LO, Bax RP, Bywater MJ, Holt cokinetics of cefotaxime and desacetylcefotaxime in infants
HA. The human pharmacokinetics of cefotaxime and its during extracorporeal membrane oxygenation. Antimicrob
metabolites and the role of renal tubular secretion on their Agents Chemother. 2010;54(5):1734–41.
elimination. J Pharmacokinet Biopharm. 1985;13(2):121–42. 202. Odio CM. Cefotaxime for treatment of neonatal sepsis and
182. Schrinner E, Limbert M, Penasse L, Lutz A. Antibacterial activity meningitis. Diagn Microbiol Infect Dis. 1995;22(1–2):111–7.
of cefotaxime and other newer cephalosporins (in vitro and 203. Muller-Pebody B, Johnson AP, Heath PT, Gilbert RE, Hender-
in vivo). J Antimicrob Chemother. 1980;6(Suppl A):25–30. son KL, Sharland M. Empirical treatment of neonatal sepsis: are
183. Mitsuhashi S, Inoue M, Masuyoshi S. Antibacterial activity of the current guidelines adequate? Arch Dis Child Fetal Neonatal
cefotaxime. J Antimicrob Chemother. 1980;6(Suppl A):37–46. Ed. 2011;96(1):F4–8.
184. Labia R, Kazmierczak A, Guionie M, Masson JM. Some bac- 204. Cotten CM, McDonald S, Stoll B, Goldberg RN, Poole K,
terial proteins with affinity for cefotaxime. J Antimicrob Che- Benjamin DK Jr. The association of third-generation cephalo-
mother. 1980;6(Suppl A):19–23. sporin use and invasive candidiasis in extremely low birth-
185. Plosker GL, Foster RH, Benfield P. Cefotaxime. A pharmaco- weight infants. Pediatrics. 2006;118(2):717–22.
economic review of its use in the treatment of infections. 205. Stockmann C, Spigarelli MG, Campbell SC, Constance JE,
Pharmacoeconomics. 1998;13(1 Pt 1):91–106. Courter JD, Thorell EA, et al. Considerations in the pharma-
186. Trang JM, Jacobs RF, Kearns GL, Brown AL, Wells TG, cologic treatment and prevention of neonatal sepsis. Paediatr
Underwood FL, et al. Cefotaxime and desacetylcefotaxime Drugs. 2014;16(1):67–81.
pharmacokinetics in infants and children with meningitis. An- 206. APP Pharmaceuticals Ltd. Gentamicin Injection, USP.
timicrob Agents Chemother. 1985;28(6):791–5. 2008. http://www.baxter.ca/en/downloads/product_information/
187. Bergan T. Pharmacokinetic properties of the cephalosporins. GENTAMICIN(E)_PM_AUG282012_EN.pdf. Accessed 1 Nov
Drugs. 1987;34(Suppl 2):89–104. 2013.
606 J. K. Roberts et al.

207. Kaplan JM, McCracken GH Jr, Horton LJ, Thomas ML, Davis 227. Janknegt R, de Marie S, Bakker-Woudenberg IA, Crommelin
N. Pharmacologic studies in neonates given large dosages of DJ. Liposomal and lipid formulations of amphotericin B. Clin-
ampicillin. J Pediatr. 1974;84(4):571–7. ical pharmacokinetics. Clin Pharmacokinet. 1992;23(4):279–91.
208. Izaki K, Matsuhashi M, Strominger JL. Glycopeptide trans- 228. Gallis HA, Drew RH, Pickard WW. Amphotericin B: 30 years
peptidase and D-alanine carboxypeptidase: penicillin-sensitive of clinical experience. Rev Infect Dis. 1990;12(2):308–29.
enzymatic reactions. Proc Natl Acad Sci USA. 1966;55(3): 229. Linder N, Klinger G, Shalit I, Levy I, Ashkenazi S, Haski G,
656–63. et al. Treatment of candidaemia in premature infants: compari-
209. Marques-Minana MR, Saadeddin A, Peris JE. Population phar- son of three amphotericin B preparations. J Antimicrob Che-
macokinetic analysis of vancomycin in neonates. A new pro- mother. 2003;52(4):663–7.
posal of initial dosage guideline. Br J Clin Pharmacol. 230. Baley JE, Meyers C, Kliegman RM, Jacobs MR, Blumer JL.
2010;70(5):713–20. Pharmacokinetics, outcome of treatment, and toxic effects of
210. Croes S, Stolk LM. Vancomycin therapeutic guidelines: closer amphotericin B and 5-fluorocytosine in neonates. J Pediatr.
examination of neonatal pharmacokinetics. Clin Infect Dis. 1990;116(5):791–7.
2011;53(9):966–7. 231. Turkova A, Roilides E, Sharland M. Amphotericin B in neo-
211. Boger DL. Vancomycin, teicoplanin, and ramoplanin: synthetic nates: deoxycholate or lipid formulation as first-line therapy—is
and mechanistic studies. Med Res Rev. 2001;21(5):356–81. there a ‘right’ choice? Curr Opin Infect Dis. 2011;24(2):163–71.
212. West-Ward Pharmaceuticals. Cefotaxime for injection, 232. Le J, Adler-Shohet FC, Nguyen C, Lieberman JM. Nephrotox-
USP. November 2012. http://www.west-ward.com/images/files/ icity associated with amphotericin B deoxycholate in neonates.
package/Cefotaxime%20for%20Inj.%20-%20PI%20-500mg% Pediatr Infect Dis J. 2009;28(12):1061–3.
20-%201g%20-%202g%20-%20Approved.pdf. Accessed 11 233. Turcu R, Patterson MJ, Omar S. Influence of sodium intake on
Nov 2013 Amphotericin B-induced nephrotoxicity among extremely pre-
213. Stoll BJ, Hansen N, Fanaroff AA, Wright LL, Carlo WA, Eh- mature infants. Pediatr Nephrol. 2009;24(3):497–505.
renkranz RA, et al. Late-onset sepsis in very low birth weight 234. Holler B, Omar SA, Farid MD, Patterson MJ. Effects of fluid
neonates: the experience of the NICHD Neonatal Research and electrolyte management on amphotericin B-induced neph-
Network. Pediatrics. 2002;110(2 Pt 1):285–91. rotoxicity among extremely low birth weight infants. Pediatrics.
214. Saiman L, Ludington E, Pfaller M, Rangel-Frausto S, Wiblin 2004;113(6):e608–16.
RT, Dawson J, et al. Risk factors for candidemia in Neonatal 235. Starke JR, Mason EO Jr, Kramer WG, Kaplan SL. Pharmaco-
Intensive Care Unit patients. The National Epidemiology of kinetics of amphotericin B in infants and children. J Infect Dis.
Mycosis Survey study group. Pediatr Infect Dis J. 2000;19(4): 1987;155(4):766–74.
319–24. 236. Hall JE, Cox F, Karlson K, Robertson A. Amphotericin B
215. Saxen H, Virtanen M, Carlson P, Hoppu K, Pohjavuori M, Vaara dosage for disseminated candidiasis in premature infants. J Per-
M, et al. Neonatal Candida parapsilosis outbreak with a high inatol. 1987;7(3):194–8.
case fatality rate. Pediatr Infect Dis J. 1995;14(9):776–81. 237. Baley JE, Kliegman RM, Fanaroff AA. Disseminated fungal
216. Rowen JL, Atkins JT, Levy ML, Baer SC, Baker CJ. Invasive infections in very low-birth-weight infants: therapeutic toxicity.
fungal dermatitis in the \ or = 1000-gram neonate. Pediatrics. Pediatrics. 1984;73(2):153–7.
1995;95(5):682–7. 238. Higuchi R, Kusumoto S, Ban H, Iwahashi S, Kobayashi M,
217. Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: Sumiyama K, et al. Increased level/dose ratio of amphotericin-B
a persistent public health problem. Clin Microbiol Rev. in premature infants with renal failure. Acta Paediatr Jpn.
2007;20(1):133–63. 1993;35(3):238–42.
218. Benjamin DK Jr, Stoll BJ, Fanaroff AA, McDonald SA, Oh W, 239. Steinbach WJ, Perfect J. Antifungal agents. In: Yaffe SJ, Aranda
Higgins RD, et al. Neonatal candidiasis among extremely low JV, editors. Neonatal and pediatric pharmacology: therapeutic
birth weight infants: risk factors, mortality rates, and neurode- principles in practice. 3rd ed. Philadelphia: Lippincot Williams
velopmental outcomes at 18 to 22 months. Pediatrics. & Wilkins; 2005. p. 459–62.
2006;117(1):84–92. 240. Kotwani RN, Gokhale PC, Bodhe PV, Kirodian BG, Kshirsagar
219. Friedman S, Richardson SE, Jacobs SE, O’Brien K. Systemic NA, Pandya SK. A comparative study of plasma concentrations
Candida infection in extremely low birth weight infants: short of liposomal amphotericin B (L-AMP-LRC-1) in adults, chil-
term morbidity and long term neurodevelopmental outcome. dren and neonates. Int J Pharm. 2002;238(1–2):11–5.
Pediatr Infect Dis J. 2000;19(6):499–504. 241. Ascher SB, Smith PB, Watt K, Benjamin DK, Cohen-Wolko-
220. Mittal M, Dhanireddy R, Higgins RD. Candida sepsis and wiez M, Clark RH, et al. Antifungal therapy and outcomes in
association with retinopathy of prematurity. Pediatrics. infants with invasive Candida infections. Pediatr Infect Dis J.
1998;101(4 Pt 1):654–7. 2012;31(5):439–43.
221. Kremer I, Naor N, Davidson S, Arbizo M, Nissenkorn I. Sys- 242. Williams KM, Kearns GL. Lipid amphotericin preparations.
temic candidiasis in babies with retinopathy of prematurity. Pediatr Infect Dis J. 2000;19(6):567–9.
Graefes Arch Clin Exp Ophthalmol. 1992;230(6):592–4. 243. Klepser ME, Wolfe EJ, Jones RN, Nightingale CH, Pfaller MA.
222. Rowen JL. Mucocutaneous candidiasis. Semin Perinatol. Antifungal pharmacodynamic characteristics of fluconazole and
2003;27(5):406–13. amphotericin B tested against Candida albicans. Antimicrob
223. Coukell AJ, Brogden RN. Liposomal amphotericin B. Thera- Agents Chemother. 1997;41(6):1392–5.
peutic use in the management of fungal infections and visceral 244. Pfaller MA, Messer SA, Hollis RJ, Jones RN, Doern GV, Brandt
leishmaniasis. Drugs. 1998;55(4):585–612. ME, et al. Trends in species distribution and susceptibility to
224. Chapman RL. Prevention and treatment of Candida infections in fluconazole among blood stream isolates of Candida species in
neonates. Semin Perinatol. 2007;31(1):39–46. the United States. Diagn Microbiol Infect Dis. 1999;33(4):
225. Bliss JM, Wellington M, Gigliotti F. Antifungal pharmacother- 217–22.
apy for neonatal candidiasis. Semin Perinatol. 2003;27(5): 245. Rowen JL, Tate JM, Nordoff N, Passarell L, McGinnis MR.
365–74. Candida isolates from neonates: frequency of misidentification
226. Hamilton-Miller JM. Chemistry and biology of the polyene and reduced fluconazole susceptibility. J Clin Microbiol.
macrolide antibiotics. Bacteriol Rev. 1973;37(2):166–96. 1999;37(11):3735–7.
Antimicrobial/Antiviral PK–PD in Neonates and Infants 607

246. Mann PA, McNicholas PM, Chau AS, Patel R, Mendrick C, 264. Piper L, Smith PB, Hornik CP, Cheifetz IM, Barrett JS, Moorthy
Ullmann AJ, et al. Impact of antifungal prophylaxis on coloni- G, et al. Fluconazole loading dose pharmacokinetics and safety
zation and azole susceptibility of Candida species. Antimicrob in infants. Pediatr Infect Dis J. 2011;30(5):375–8.
Agents Chemother. 2009;53(12):5026–34. 265. Spriet I, Cosaert K, Renard M, Uyttebroeck A, Meyts I, Proes-
247. Dotis J, Prasad PA, Zaoutis T, Roilides E. Epidemiology, risk mans M, et al. Voriconazole plasma levels in children are highly
factors and outcome of Candida parapsilosis bloodstream variable. Eur J Clin Microbiol Infect Dis. 2011;30(2):283–7.
infection in children. Pediatr Infect Dis J. 2012;31(6):557–60. 266. Almirante B, Rodriguez D. Antifungal agents in neonates: issues
248. Mandras N, Tullio V, Allizond V, Scalas D, Banche G, Roana J, and recommendations. Paediatr Drugs. 2007;9(5):311–21.
et al. In vitro activities of fluconazole and voriconazole against 267. Scholz I, Oberwittler H, Riedel KD, Burhenne J, Weiss J,
clinical isolates of Candida spp. determined by disk diffusion Haefeli WE, et al. Pharmacokinetics, metabolism and bioavail-
testing in Turin, Italy. Antimicrob Agents Chemother. ability of the triazole antifungal agent voriconazole in relation to
2009;53(4):1657–9. CYP2C19 genotype. Br J Clin Pharmacol. 2009;68(6):906–15.
249. Pfaller MA, Messer SA, Hollis RJ, Jones RN, Diekema DJ. 268. Watt KM, Cohen-Wolkowiez M, Ward RM, Benjamin DK Jr.
In vitro activities of ravuconazole and voriconazole compared Commentary: pediatric antifungal drug development: lessons
with those of four approved systemic antifungal agents against learned and recommendations for the future. Pediatr Infect Dis J.
6,970 clinical isolates of Candida spp. Antimicrob Agents 2012;31(6):635–7.
Chemother. 2002;46(6):1723–7. 269. Karlsson MO, Lutsar I, Milligan PA. Population pharmacoki-
250. Diekema DJ, Messer SA, Hollis RJ, Boyken L, Tendolkar S, netic analysis of voriconazole plasma concentration data from
Kroeger J, et al. A global evaluation of voriconazole activity pediatric studies. Antimicrob Agents Chemother. 2009;53(3):
tested against recent clinical isolates of Candida spp. Diagn 935–44.
Microbiol Infect Dis. 2009;63(2):233–6. 270. Shima H, Miharu M, Osumi T, Takahashi T, Shimada H. Dif-
251. Kelly SL, Lamb DC, Corran AJ, Baldwin BC, Kelly DE. Mode ferences in voriconazole trough plasma concentrations per oral
of action and resistance to azole antifungals associated with the dosages between children younger and older than 3 years of age.
formation of 14 alpha-methylergosta-8,24(28)-dien-3 beta,6 Pediatr Blood Cancer. 2010;54(7):1050–2.
alpha-diol. Biochem Biophys Res Commun. 1995;207(3):910–5. 271. Doby EH, Benjamin DK Jr, Blaschke AJ, Ward RM, Pavia AT,
252. Tripathi N, Watt K, Benjamin DK Jr. Treatment and prophylaxis Martin PL, et al. Therapeutic monitoring of voriconazole in
of invasive candidiasis. Semin Perinatol. 2012;36(6):416–23. children less than three years of age: a case report and summary
253. Wildfeuer A, Laufen H, Schmalreck AF, Yeates RA, Zimmer- of voriconazole concentrations for ten children. Pediatr Infect
mann T. Fluconazole: comparison of pharmacokinetics, therapy Dis J. 2012;31(6):632–5.
and in vitro susceptibility. Mycoses. 1997;40(7–8):259–65. 272. Gerin M, Mahlaoui N, Elie C, Lanternier F, Bougnoux ME,
254. Fasano C, O’Keeffe J, Gibbs D. Fluconazole treatment of neo- Blanche S, et al. Therapeutic drug monitoring of voriconazole
nates and infants with severe fungal infections not treatable with after intravenous administration in infants and children with
conventional agents. Eur J Clin Microbiol Infect Dis. primary immunodeficiency. Ther Drug Monit. 2011;33(4):
1994;13(4):351–4. 464–6.
255. Steinbach WJ, Benjamin DK. New antifungal agents under 273. Pappas PG, Kauffman CA, Andes D, Benjamin DK Jr, Calandra
development in children and neonates. Curr Opin Infect Dis. TF, Edwards JE Jr, et al. Clinical practice guidelines for the
2005;18(6):484–9. management of candidiasis: 2009 update by the Infectious
256. Walsh TJ, Lutsar I, Driscoll T, Dupont B, Roden M, Ghahra- Diseases Society of America. Clin Infect Dis. 2009;48(5):
mani P, et al. Voriconazole in the treatment of aspergillosis, 503–35.
scedosporiosis and other invasive fungal infections in children. 274. Driessen M, Ellis JB, Cooper PA, Wainer S, Muwazi F, Hahn D,
Pediatr Infect Dis J. 2002;21(3):240–8. et al. Fluconazole vs. amphotericin B for the treatment of neo-
257. Celik IH, Demirel G, Oguz SS, Uras N, Erdeve O, Dilmen U. natal fungal septicemia: a prospective randomized trial. Pediatr
Compassionate use of voriconazole in newborn infants diag- Infect Dis J. 1996;15(12):1107–12.
nosed with severe invasive fungal sepsis. Eur Rev Med Phar- 275. Wainer S, Cooper PA, Gouws H, Akierman A. Prospective
macol Sci. 2013;17(6):729–34. study of fluconazole therapy in systemic neonatal fungal infec-
258. Saxen H, Hoppu K, Pohjavuori M. Pharmacokinetics of fluco- tion. Pediatr Infect Dis J. 1997;16(8):763–7.
nazole in very low birth weight infants during the first two 276. Healy CM, Baker CJ, Zaccaria E, Campbell JR. Impact of
weeks of life. Clin Pharmacol Ther. 1993;54(3):269–77. fluconazole prophylaxis on incidence and outcome of invasive
259. Nahata MC, Tallian KB, Force RW. Pharmacokinetics of candidiasis in a neonatal intensive care unit. J Pediatr.
fluconazole in young infants. Eur J Drug Metab Pharmacokinet. 2005;147(2):166–71.
1999;24(2):155–7. 277. Austin N, McGuire W. Prophylactic systemic antifungal agents
260. Wenzl TG, Schefels J, Hornchen H, Skopnik H. Pharmacoki- to prevent mortality and morbidity in very low birth weight
netics of oral fluconazole in premature infants. Eur J Pediatr. infants. Cochrane Database Syst Rev. 2013;4:CD003850.
1998;157(8):661–2. 278. Austin N, McGuire W. Prophylactic systemic antifungal agents
261. Wade KC, Wu D, Kaufman DA, Ward RM, Benjamin DK Jr, to prevent mortality and morbidity in very low birth weight
Sullivan JE, et al. Population pharmacokinetics of fluconazole in infants. Cochrane Database Syst Rev 2013;4:CD003850.
young infants. Antimicrob Agents Chemother. 2008;52(11): 279. Garcia-Effron G, Park S, Perlin DS. Correlating echinocandin
4043–9. MIC and kinetic inhibition of fks1 mutant glucan synthases for
262. Wade KC, Benjamin DK Jr, Kaufman DA, Ward RM, Smith PB, Candida albicans: implications for interpretive breakpoints.
Jayaraman B, et al. Fluconazole dosing for the prevention or Antimicrob Agents Chemother. 2009;53(1):112–22.
treatment of invasive candidiasis in young infants. Pediatr Infect 280. Douglas CM. Fungal beta(1,3)-D-glucan synthesis. Med Mycol.
Dis J. 2009;28(8):717–23. 2001;39(Suppl 1):55–66.
263. Stockmann C, Constance J, Roberts JK, Olsen J, Doby EH, 281. Kurtz MB, Douglas CM. Lipopeptide inhibitors of fungal glucan
Ampofo K, et al. Pharmacokinetics and pharmacodynamics of synthase. J Med Vet Mycol. 1997;35(2):79–86.
antifungals in children and their clinical implications. Clin 282. Denning DW. Echinocandin antifungal drugs. Lancet.
Pharmacokinet. 2014 May;53(5):429-54. 2003;362(9390):1142–51.
608 J. K. Roberts et al.

283. Morrison VA. Echinocandin antifungals: review and update. volunteers, volunteers with moderate liver disease, and volun-
Expert Rev Anti Infect Ther. 2006;4(2):325–42. teers with renal dysfunction. J Clin Pharmacol. 2005;45(10):
284. Niimi K, Monk BC, Hirai A, Hatakenaka K, Umeyama T, 1145–52.
Lamping E, et al. Clinically significant micafungin resistance in 302. Yanni SB, Smith PB, Benjamin DK Jr, Augustijns PF, Thakker
Candida albicans involves modification of a glucan synthase DR, Annaert PP. Higher clearance of micafungin in neonates
catalytic subunit GSC1 (FKS1) allele followed by loss of het- compared with adults: role of age-dependent micafungin serum
erozygosity. J Antimicrob Chemother. 2010;65(5):842–52. binding. Biopharm Drug Dispos. 2011;32(4):222–32.
285. Balashov SV, Park S, Perlin DS. Assessing resistance to the 303. Hope WW, Smith PB, Arrieta A, Buell DN, Roy M, Kaibara A,
echinocandin antifungal drug caspofungin in Candida albicans et al. Population pharmacokinetics of micafungin in neonates
by profiling mutations in FKS1. Antimicrob Agents Chemother. and young infants. Antimicrob Agents Chemother. 2010;54(6):
2006;50(6):2058–63. 2633–7.
286. Pfeiffer CD, Garcia-Effron G, Zaas AK, Perfect JR, Perlin DS, 304. Prasad PA, Coffin SE, Leckerman KH, Walsh TJ, Zaoutis TE.
Alexander BD. Breakthrough invasive candidiasis in patients on Pediatric antifungal utilization: new drugs, new trends. Pediatr
micafungin. J Clin Microbiol. 2010;48(7):2373–80. Infect Dis J. 2008;27(12):1083–8.
287. Park S, Kelly R, Kahn JN, Robles J, Hsu MJ, Register E, et al. 305. Caudle KE, Inger AG, Butler DR, Rogers PD. Echinocandin use
Specific substitutions in the echinocandin target Fks1p account in the neonatal intensive care unit. Ann Pharmacother.
for reduced susceptibility of rare laboratory and clinical Can- 2012;46(1):108–16.
dida sp. isolates. Antimicrob Agents Chemother. 2005;49(8): 306. Testoni D, Smith PB, Benjamin DK Jr. The use of antifungal
3264–73. therapy in neonatal intensive care. Clin Perinatol. 2012;39(1):
288. Chen SC, Slavin MA, Sorrell TC. Echinocandin antifungal 83–98.
drugs in fungal infections: a comparison. Drugs. 2011;71(1): 307. Wurthwein G, Groll AH, Hempel G, Adler-Shohet FC, Lieber-
11–41. man JM, Walsh TJ. Population pharmacokinetics of amphoter-
289. Smith PB, Walsh TJ, Hope W, Arrieta A, Takada A, Kovanda icin B lipid complex in neonates. Antimicrob Agents
LL, et al. Pharmacokinetics of an elevated dosage of micafungin Chemother. 2005;49(12):5092–8.
in premature neonates. Pediatr Infect Dis J. 2009;28(5):412–5. 308. Evdoridou J, Roilides E, Bibashi E, Kremenopoulos G. Multi-
290. Benjamin DK Jr, Smith PB, Arrieta A, Castro L, Sanchez PJ, focal osteoarthritis due to Candida albicans in a neonate: serum
Kaufman D, et al. Safety and pharmacokinetics of repeat-dose level monitoring of liposomal amphotericin B and literature
micafungin in young infants. Clin Pharmacol Ther. 2010;87(1): review. Infection. 1997;25(2):112–6.
93–9. 309. Bekersky I, Fielding RM, Dressler DE, Lee JW, Buell DN,
291. Heresi GP, Gerstmann DR, Reed MD, van den Anker JN, Walsh TJ. Pharmacokinetics, excretion, and mass balance of
Blumer JL, Kovanda L, et al. The pharmacokinetics and safety liposomal amphotericin B (AmBisome) and amphotericin B
of micafungin, a novel echinocandin, in premature infants. Pe- deoxycholate in humans. Antimicrob Agents Chemother.
diatr Infect Dis J. 2006;25(12):1110–5. 2002;46(3):828–33.
292. Natarajan G, Lulic-Botica M, Aranda JV. Refractory neonatal 310. Driessen M, Ellis JB, Cooper PA, Wainer S, Muwazi F, Hahn D,
candidemia and high-dose micafungin pharmacotherapy. J Per- et al. Fluconazole vs. amphotericin B for the treatment of neo-
inatol. 2009;29(11):738–43. natal fungal septicemia: a prospective randomized trial. Pediatr
293. Saez-Llorens X, Macias M, Maiya P, Pineros J, Jafri HS, Infect Dis J. 1996;15(12):1107–12.
Chatterjee A, et al. Pharmacokinetics and safety of caspofungin 311. Pfaller MA, Diekema DJ, Rex JH, Espinel-Ingroff A, Johnson
in neonates and infants less than 3 months of age. Antimicrob EM, Andes D, et al. Correlation of MIC with outcome for
Agents Chemother. 2009;53(3):869–75. Candida species tested against voriconazole: analysis and pro-
294. Odio CM, Araya R, Pinto LE, Castro CE, Vasquez S, Alfaro B, posal for interpretive breakpoints. J Clin Microbiol. 2006;44(3):
et al. Caspofungin therapy of neonates with invasive candidiasis. 819–26.
Pediatr Infect Dis J. 2004;23(12):1093–7. 312. Groll AH, Walsh TJ. Caspofungin: pharmacology, safety and
295. Filippi L, Poggi C, Gozzini E, Meleleo R, Mirabile L, Fiorini P. therapeutic potential in superficial and invasive fungal infec-
Neonatal liver abscesses due to Candida infection effectively tions. Expert Opin Investig Drugs. 2001;10(8):1545–58.
treated with caspofungin. Acta Paediatr. 2009;98(5):906–9. 313. Watt K, Benjamin DK Jr, Cohen-Wolkowiez M. Pharmacoki-
296. Manzar S, Kamat M, Pyati S. Caspofungin for refractory can- netics of antifungal agents in children. Early Hum Dev.
didemia in neonates. Pediatr Infect Dis J. 2006;25(3):282–3. 2011;87(Suppl 1):S61–5.
297. Merk and Compnay Inc. Cancidas (caspofungin acetate) for 314. Eschenauer G, Depestel DD, Carver PL. Comparison of
injection: US prescribing information. http://www.merck.com/ echinocandin antifungals. Ther Clin Risk Manag.
product/usa/pi_circulars/c/cancidas/cancidas_pi.pdf. Accessed 2007;3(1):71–97.
18 Oct 2013. 315. Stone JA, Xu X, Winchell GA, Deutsch PJ, Pearson PG, Migoya
298. Neely M, Jafri HS, Seibel N, Knapp K, Adamson PC, Bradshaw EM, et al. Disposition of caspofungin: role of distribution in
SK, et al. Pharmacokinetics and safety of caspofungin in determining pharmacokinetics in plasma. Antimicrob Agents
older infants and toddlers. Antimicrob Agents Chemother. Chemother. 2004;48(3):815–23.
2009;53(4):1450–6. 316. Brown ZA, Wald A, Morrow RA, Selke S, Zeh J, Corey L.
299. Li CC, Sun P, Dong Y, Bi S, Desai R, Dockendorf MF, et al. Effect of serologic status and cesarean delivery on transmission
Population pharmacokinetics and pharmacodynamics of caspo- rates of herpes simplex virus from mother to infant. JAMA.
fungin in pediatric patients. Antimicrob Agents Chemother. 2003;289(2):203–9.
2011;55(5):2098–105. 317. Whitley RJ. The use of antiviral drugs during the neonatal
300. Seibel NL, Schwartz C, Arrieta A, Flynn P, Shad A, Albano E, period. Clin Perinatol. 2012;39(1):69–81.
et al. Safety, tolerability, and pharmacokinetics of Micafungin 318. Kimberlin DW. Management of HSV encephalitis in adults and
(FK463) in febrile neutropenic pediatric patients. Antimicrob neonates: diagnosis, prognosis and treatment. Herpes.
Agents Chemother. 2005;49(8):3317–24. 2007;14(1):11–6.
301. Hebert MF, Smith HE, Marbury TC, Swan SK, Smith WB, 319. Demmler GJ. Infectious Diseases Society of America and
Townsend RW, et al. Pharmacokinetics of micafungin in healthy Centers for Disease Control. Summary of a workshop on
Antimicrobial/Antiviral PK–PD in Neonates and Infants 609

surveillance for congenital cytomegalovirus disease. Rev Infect 338. Long SS. In defense of empiric acyclovir therapy in certain
Dis. 1991;13(2):315–29. neonates. J Pediatr. 2008;153(2):157–8.
320. Istas AS, Demmler GJ, Dobbins JG, Stewart JA. Surveillance 339. Caviness AC, Demmler GJ, Swint JM, Cantor SB. Cost-effec-
for congenital cytomegalovirus disease: a report from the tiveness analysis of herpes simplex virus testing and treatment
National Congenital Cytomegalovirus Disease Registry. Clin strategies in febrile neonates. Arch Pediatr Adolesc Med.
Infect Dis. 1995;20(3):665–70. 2008;162(7):665–74.
321. Griffiths P. Cytomegalovirus infection of the central nervous 340. Kimberlin DW. When should you initiate acyclovir therapy in a
system. Herpes. 2004;11(Suppl 2):95a–104a. neonate? J Pediatr. 2008;153(2):155–6.
322. Elion GB, Furman PA, Fyfe JA, de Miranda P, Beauchamp L, 341. Whitley RJ. The use of antiviral drugs during the neonatal
Schaeffer HJ. Selectivity of action of an antiherpetic agent, 9-(2- period. Clin Perinatol. 2012;39(1):69–81.
hydroxyethoxymethyl) guanine. Proc Natl Acad Sci U S A. 342. Ashton WT, Karkas JD, Field AK, Tolman RL. Activation by
1977;74(12):5716–20. thymidine kinase and potent antiherpetic activity of 20 -nor-20 -
323. Schaeffer HJ, Beauchamp L, de Miranda P, Elion GB, Bauer DJ, deoxyguanosine (20 NDG). Biochem Biophys Res Commun.
Collins P. 9-(2-hydroxyethoxymethyl) guanine activity against 1982;108(4):1716–21.
viruses of the herpes group. Nature. 1978;272(5654):583–5. 343. Martin JC, Dvorak CA, Smee DF, Matthews TR, Verheyden JP.
324. Englund JA, Zimmerman ME, Swierkosz EM, Goodman JL, 9-[(1,3-Dihydroxy-2-propoxy)methyl]guanine: a new potent and
Scholl DR, Balfour HH Jr. Herpes simplex virus resistant to selective antiherpes agent. J Med Chem. 1983;26(5):759–61.
acyclovir. A study in a tertiary care center. Ann Intern Med. 344. Cheng YC, Huang ES, Lin JC, Mar EC, Pagano JS, Dutschman
1990;112(6):416–22. GE, et al. Unique spectrum of activity of 9-[(1,3-dihydroxy-2-
325. Kimberlin DW, Lin CY, Jacobs RF, Powell DA, Corey L, propoxy)methyl]-guanine against herpesviruses in vitro and its
Gruber WC, et al. Safety and efficacy of high-dose intravenous mode of action against herpes simplex virus type 1. Proc Natl
acyclovir in the management of neonatal herpes simplex virus Acad Sci U S A. 1983;80(9):2767–70.
infections. Pediatrics. 2001;108(2):230–8. 345. Field AK, Davies ME, DeWitt C, Perry HC, Liou R, Germers-
326. Kimberlin D, Powell D, Gruber W, Diaz P, Arvin A, Kumar M, hausen J, et al. 9-([2-hydroxy-1-(hydroxymethyl)ethoxy]methyl)
et al. Administration of oral acyclovir suppressive therapy after guanine: a selective inhibitor of herpes group virus replication.
neonatal herpes simplex virus disease limited to the skin, eyes Proc Natl Acad Sci U S A. 1983;80(13):4139–43.
and mouth: results of a phase I/II trial. Pediatr Infect Dis J. 346. Smith KO, Galloway KS, Kennell WL, Ogilvie KK, Radatus
1996;15(3):247–54. BK. A new nucleoside analog, 9-[[2-hydroxy-1-(hydroxy-
327. Sampson MR, Bloom BT, Lenfestey RW, Harper B, Kashuba methyl)ethoxyl]methyl]guanine, highly active in vitro against
AD, Anand R, et al. Population pharmacokinetics of intravenous herpes simplex virus types 1 and 2. Antimicrob Agents Che-
acyclovir in preterm and term infants. Pediatr Infect Dis J. mother. 1982;22(1):55–61.
2014;33:42–9. 347. Lin JC, Smith MC, Pagano JS. Prolonged inhibitory effect of
328. Blum MR, Liao SH, de Miranda P. Overview of acyclovir 9-(1,3-dihydroxy-2-propoxymethyl)guanine against replication
pharmacokinetic disposition in adults and children. Am J Med. of Epstein-Barr virus. J Virol. 1984;50(1):50–5.
1982;73(1A):186–92. 348. Cheng YC, Grill SP, Dutschman GE, Nakayama K, Bastow KF.
329. Hintz M, Connor JD, Spector SA, Blum MR, Keeney RE, Metabolism of 9-(1,3-dihydroxy-2-propoxymethyl)guanine, a
Yeager AS. Neonatal acyclovir pharmacokinetics in patients new anti-herpes virus compound, in herpes simplex virus-
with herpes virus infections. Am J Med. 1982;73(1A):210–4. infected cells. J Biol Chem. 1983;258(20):12460–4.
330. Englund JA, Fletcher CV, Balfour HH Jr. Acyclovir therapy in 349. Sullivan V, Talarico CL, Stanat SC, Davis M, Coen DM, Biron
neonates. J Pediatr. 1991;119(1 Pt 1):129–35. KK. A protein kinase homologue controls phosphorylation of
331. Sampson MR, Bloom BT, Lenfestey RW, Harper B, Kashuba ganciclovir in human cytomegalovirus-infected cells. Nature.
AD, Anand R, et al. Population pharmacokinetics of intravenous 1992;359(6390):85.
acyclovir in preterm and term infants. Pediatr Infect Dis J. 350. Littler E, Stuart AD, Chee MS. Human cytomegalovirus UL97
2014;33(1):42–9. open reading frame encodes a protein that phosphorylates
332. Rabalais GP, Nusinoff-Lehrman S, Arvin AM, Levin MJ. the antiviral nucleoside analogue ganciclovir. Nature.
Antiviral susceptibilities of herpes simplex virus isolates from 1992;358(6382):160–2.
infants with recurrent mucocutaneous lesions after neonatal 351. Chee MS, Lawrence GL, Barrell BG. Alpha-, beta- and gam-
infection. Pediatr Infect Dis J. 1989;8(4):221–3. maherpesviruses encode a putative phosphotransferase. J Gen
333. Pinninti SG, Kimberlin DW. Neonatal herpes simplex virus Virol. 1989;70(Pt 5):1151–60.
infections. Pediatr Clin N Am. 2013;60(2):351–65. 352. Erice A. Resistance of human cytomegalovirus to antiviral
334. Jones CA, Walker KS, Badawi N. Antiviral agents for treatment drugs. Clin Microbiol Rev. 1999;12(2):286–97.
of herpes simplex virus infection in neonates. Cochrane Data- 353. Whitley RJ, Cloud G, Gruber W, Storch GA, Demmler GJ,
base Syst Rev. 2009;(3):CD004206. Jacobs RF, et al. Ganciclovir treatment of symptomatic con-
335. Shah SS, Aronson PL, Mohamad Z, Lorch SA. Delayed acy- genital cytomegalovirus infection: results of a phase II study.
clovir therapy and death among neonates with herpes simplex National Institute of Allergy and Infectious Diseases Collabo-
virus infection. Pediatrics. 2011;128(6):1153–60. rative Antiviral Study Group. J Infect Dis. 1997;175(5):1080–6.
336. Whitley R, Arvin A, Prober C, Burchett S, Corey L, Powell D, 354. Kimberlin DW. Antiviral therapy for cytomegalovirus infections
et al. A controlled trial comparing vidarabine with acyclovir in in pediatric patients. Semin Pediatr Infect Dis. 2002;13(1):22–30.
neonatal herpes simplex virus infection. Infectious Diseases 355. Jacobson MA, Gambertoglio JG, Aweeka FT, Causey DM,
Collaborative Antiviral Study Group. N Engl J Med. Portale AA. Foscarnet-induced hypocalcemia and effects of
1991;324(7):444–9. foscarnet on calcium metabolism. J Clin Endocrinol Metab.
337. Kimberlin DW, Whitley RJ, Wan W, Powell DA, Storch G, 1991;72(5):1130–5.
Ahmed A, et al. Oral acyclovir suppression and neurodevelop- 356. Kimberlin DW, Lin CY, Sanchez PJ, Demmler GJ, Dankner W,
ment after neonatal herpes. N Engl J Med. 2011;365(14): Shelton M, et al. Effect of ganciclovir therapy on hearing in
1284–92. symptomatic congenital cytomegalovirus disease involving the
610 J. K. Roberts et al.

central nervous system: a randomized, controlled trial. J Pediatr. 365. Zhou XJ, Gruber W, Demmler G, Jacobs R, Reuman P, Adler S,
2003;143(1):16–25. et al. Population pharmacokinetics of ganciclovir in newborns
357. Cocohoba JM, McNicholl IR. Valganciclovir: an advance in with congenital cytomegalovirus infections. NIAID Collabora-
cytomegalovirus therapeutics. Ann Pharmacother. 2002;36(6): tive Antiviral Study Group. Antimicrob Agents Chemother.
1075–9. 1996;40(9):2202–5.
358. Meine Jansen CF, Toet MC, Rademaker CM, Ververs TF, 366. Markham A, Faulds D. Ganciclovir. An update of its therapeutic
Gerards LJ, van Loon AM. Treatment of symptomatic congen- use in cytomegalovirus infection. Drugs. 1994;48(3):455–84.
ital cytomegalovirus infection with valganciclovir. J Perinat 367. Galli L, Novelli A, Chiappini E, Gervaso P, Cassetta MI, Fallani
Med. 2005;33(4):364–6. S, et al. Valganciclovir for congenital CMV infection: a pilot
359. Muller A, Eis-Hubinger AM, Brandhorst G, Heep A, Bartmann study on plasma concentration in newborns and infants. Pediatr
P, Franz AR. Oral valganciclovir for symptomatic congenital Infect Dis J. 2007;26(5):451–3.
cytomegalovirus infection in an extremely low birth weight 368. Acosta EP, Brundage RC, King JR, Sanchez PJ, Sood S,
infant. J Perinatol. 2008;28(1):74–6. Agrawal V, et al. Ganciclovir population pharmacokinetics in
360. Schulzke S, Buhrer C. Valganciclovir for treatment of congen- neonates following intravenous administration of ganciclovir
ital cytomegalovirus infection. Eur J Pediatr. 2006;165(8): and oral administration of a liquid valganciclovir formulation.
575–6. Clin Pharmacol Ther. 2007;81(6):867–72.
361. Buonuomo PS, Maurizi P, Valentini P, Mastrangelo S, Lazz- 369. Kimberlin DW, Lin CY, Sanchez PJ, Demmler GJ, Dankner W,
areschi I, Ridola V, et al. Successful treatment with oral val- Shelton M, et al. Effect of ganciclovir therapy on hearing in
ganciclovir in immunocompetent infant with gastrointestinal symptomatic congenital cytomegalovirus disease involving the
manifestations of cytomegalovirus infection. J Perinatol. central nervous system: a randomized, controlled trial. J Pediatr.
2006;26(10):648–9. 2003;143(1):16–25.
362. Kimberlin DW, Acosta EP, Sanchez PJ, Sood S, Agrawal V, 370. Oliver SE, Cloud GA, Sánchez PJ, Demmler GJ, Dankner W,
Homans J, et al. Pharmacokinetic and pharmacodynamic Shelton M, et al. Neurodevelopmental outcomes following
assessment of oral valganciclovir in the treatment of symp- ganciclovir therapy in symptomatic congenital cytomegalovirus
tomatic congenital cytomegalovirus disease. J Infect Dis. infections involving the central nervous system. J Clin Virol.
2008;197(6):836–45. 2009;46(Suppl 4):S22–6.
363. Lombardi G, Garofoli F, Villani P, Tizzoni M, Angelini M, 371. Nassetta L, Kimberlin D, Whitley R. Treatment of congenital
Cusato M, et al. Oral valganciclovir treatment in newborns with cytomegalovirus infection: implications for future therapeutic
symptomatic congenital cytomegalovirus infection. Eur J Clin strategies. J Antimicrob Chemother. 2009;63(5):862–7.
Microbiol Infect Dis. 2009;28(12):1465–70. 372. Frenkel LM, Capparelli EV, Dankner WM, Xu J, Smith IL,
364. Trang JM, Kidd L, Gruber W, Storch G, Demmler G, Jacobs R, Ballow A, et al. Oral ganciclovir in children: pharmacokinetics,
et al. Linear single-dose pharmacokinetics of ganciclovir in safety, tolerance, and antiviral effects. The Pediatric AIDS
newborns with congenital cytomegalovirus infections. NIAID Clinical Trials Group. J Infect Dis. 2000;182(6):1616–24.
Collaborative Antiviral Study Group. Clin Pharmacol Ther.
1993;53(1):15–21.
Copyright of Clinical Pharmacokinetics is the property of Springer Science & Business Media
B.V. and its content may not be copied or emailed to multiple sites or posted to a listserv
without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like