Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 1.
Published in final edited form as: Clin Perinatol. 2012 Jan 11;39(1):83–98. doi: 10.1016/j.clp.2011.12.008

The Use of Antifungal Therapy in Neonatal Intensive Care

Daniela Testoni a, P Brian Smith a,b, Daniel K Benjamin Jr a,b,*
PMCID: PMC3285432  NIHMSID: NIHMS345662  PMID: 22341539

Invasive candidiasis in extremely premature infants is the second most common cause of infectious disease-related death.1 Birth weight is strongly related to the incidence of invasive candidiasis (1% of infants born weighing 1000–1500 g versus up to 12% of infants born weighing 401–750 g).2 The morbidity and mortality of premature infants with invasive candidiasis are high.3,4 In a cohort of 320 extremely-low-birth-weight (ELBW, <1000 g birth weight) infants with invasive candidiasis, 73% died or were neurodevelopmentally impaired at 18–22 months corrected age.3

A unique characteristic of invasive candidiasis in infants is the frequent involvement of the central nervous system (CNS). The incidence of Candida meningitis among candidemic infants varies from 5–25%.3,5,6 Meningitis is not the only manifestation of CNS disease; parenchymal abscesses and vasculitis are also frequent in infants with invasive candidiasis.7 Therefore, CNS involvement in invasive candidiasis among infants can best be termed meningo-encephalitis. In meningo-encephalitis due to Candida, cerebrospinal fluid (CSF) cultures are often negative, CSF parameters (e.g., white blood cell count) are often normal,5 and imaging is unreliable.

Given the high incidence of meningo-encephalitis in the setting of candidemia and the lack of reliability of testing, the presence of meningo-encephalitis should be assumed in the candidemic neonate. This assumption influences length of therapy, dosing, and other key components of antifungal drug development and selection.

Although antifungals have long been used in infants, their efficacy in this population is based on extrapolation from trials performed in adults.8 Randomized trials to evaluate prophylactic systemic antifungal agents in very-low-birth-weight (VLBW, <1500 g birth weight) and ELBW infants exist, but no well-powered trials exist to guide treatment for invasive fungal infection in preterm infants.913 However, a number of pharmacokinetic (PK) antifungal studies have been completed (Table 1). In this article, we summarize those findings.

Table 1.

Pediatric Antifungal Dosing1425

Drug Formulation Infants
(31 days–2 years)
Neonates
(0–30 days)
FDA label
Polyenes
Amphotericin B
deoxycholate
IV 1 mg/kg/day 1 mg/kg/day children and adults
ABLC IV unknown unknown >16 months
ABCD IV unknown unknown children and adults
L-Amphotericin B IV 5 mg/kg/day 5 mg/kg/day ≥1 month
Nucleoside analogs
5-Flucytosine PO 50–150 mg/kg/day
q 6 hr
50–150 mg/kg/day
q 6 hr
adults
Triazoles
Fluconazole IV, PO 12 mg/kg/day
(25 mg/kg/loading dose)
12 mg/kg/day
(25 mg/kg load)
≥6 months
Voriconazole IV, PO unknown unknown ≥12 years
Posaconazole PO unknown unknown ≥13 years
Echinocandins
Caspofungin IV 50 mg/m2/day 25 mg/m2/day >3 months
Micafungin IV 10 mg/kg/day 10 mg/kg/day adults
Anidulafungin IV 1.5 mg/kg/day
(3 mg/kg/loading dose)
1.5 mg/kg/day
(3 mg/kg/load)
adults

IV: intravenous; PO: oral

POLYENES

Amphotericin B deoxycholate

Amphotericin B deoxycholate was approved for use in adults in 1958 and is now approved for use in children and adults. It acts by binding to a cytoplasmic membrane ergosterol of the fungus, thereby creating pores in cell membranes.26 Amphotericin B deoxycholate is poorly absorbed after oral administration and is highly protein-bound (95%).27 It is widely distributed in the body and can be detected in the liver, spleen, and kidneys.27

Amphotericin B deoxycholate has a longer half-life in infants (15 hours) than in adults and greater potential for drug accumulation.14 The half-life, volume of distribution, and clearance are highly variable in infants.14 CSF penetration in infants is higher than in adults, with concentrations 40–90% of the serum concentrations14; importantly, the amphotericin products tend to have substantial brain tissue penetration.

Nephrotoxicity is an important side effect observed with use of amphotericin B deoxycholate.14,28,29 In a retrospective study evaluating 92 infants with a median gestational age of 26 (range 23–41) weeks, 16% experienced nephrotoxicity and 17% had hypokalemia.30 However, in another study designed to compare the effectiveness and tolerability of 3 antifungal preparations—amphotericin B deoxycholate, liposomal amphotericin B (L-amB), and amphotericin B colloidal dispersion (ABCD)—no infant (n=56) experienced renal function deterioration during treatment.15

Amphotericin lipid formulations

Three lipid formulations of amphotericin are available: L-amB (or AmBisome), amphotericin B lipid complex (ABLC or Abelcet), and ABCD (or Amphotec). The Food and Drug Administration (FDA) has approved L-amB for use in children ≥1 month of age, ABLC for children ≥16 months of age, and ABCD for children and adults.

L-amB at a dose 1 mg/kg daily demonstrated cumulative dose effect, and the peak plasma concentrations were higher in adults compared with children and infants.31 Accumulation of amphotericin B in rabbits’ kidneys following 5 mg/kg/day of L-amB was only 0.87 μg/g compared with 12.7 μg/g following 1 mg/kg/day of amphotericin B deoxycholate.32 The clearance of ABLC in infants was similar to that observed in older patients.33 The recommended dose for ABLC is 2.5–5 mg/kg/day.33

Infants in a prospective single-center study were given 1 mg/kg/day of amphotericin B deoxycholate (n=34) if their serum creatinine was <1.2; otherwise, they were given either 5 mg/kg/day of L-amB (n=6) or 3 mg/kg/day of ABCD (n=16). No statistical difference in mortality was noted among the 3 groups.15 In a study of 46 VLBW infants who received L-amB at a dose of 1–3 mg/kg/day (26 infants) or amphotericin B deoxycholate at a dose of 0.5–1 mg/kg/day (20 infants), the fungal eradication rate was similar between groups: 84% of the L-amB group and 89% of the amphotericin B deoxycholate group.26 Effectiveness of L-amB was 73% (n=44) in a prospective cohort of infants with invasive candidiasis and 63% (n=21) among VLBW infants.34 L-amB was effective in 95% of infants (n=41) with invasive candidiasis (28 were ELBW) treated with a high dose (5–7 mg/kg/day). The infection cleared faster if the target dose was reached earlier.16

In a prospective cohort of 21 VLBW infants receiving L-amB, hypokalemia was the only side effect observed and was supplementation responsive.34 The number of studies of ABLC and ABCD in pediatric populations is small, but both agents were well-tolerated.33,35,36 In a prospective study comparing the 3 formulations, no significant renal or hepatic toxicities were noted with any of the preparations.15 Because renal penetration is limited with lipid formulations, the clinician should document negative urine cultures in infants for whom these preparations are used as monotherapy.32

NUCLEOSIDE ANALOGS

Flucytosine

Flucytosine (5-FC), through its antimetabolite 5-fluoracil, alters RNA and DNA synthesis of the mycotic cell. 5-FC is converted to 5-fluoracil by cytosine deaminase, a fungi enzyme absent from human cells.37 Flucytosine is active against Candida sp., Aspergillus sp., and Cryptococcus sp.37 The occurrence of resistance with the use of 5-FC as monotherapy precludes its use as a single agent.38 5-FC is highly bioavailable and has excellent penetration of body fluids, with CSF concentrations at 74% of plasma levels.37 5-FC is FDA-approved only for use in adults.39 The recommended dose is 25–100 mg/kg/day.14 In 13 infants (24–40 weeks’ gestational age), the median half-life was twice that of adults, with considerable inter-individual variability.14 5-FC can be toxic and can result in hepatic injury, bone marrow suppression, and gastrointestinal intolerance.40 The risk for developing toxic events increases when 5-FC levels exceed 100 mg/L; therefore, therapeutic drug monitoring is necessary.40,41

The PK data in infants are limited, and there has been no clinical trial to evaluate 5-FC efficacy in this population. A cohort study of 27 ELBW infants with meningo-encephalitis showed that time to clear infection was longer in infants given combination flucytosine and amphotericin B deoxycholate than in those treated with amphotericin B deoxycholate alone.3 5-FC use is limited by its toxicities and the need for oral dosing, and we typically discourage its use except in rare circumstances.

TRIAZOLES

Fluconazole

Fluconazole is a water-soluble triazole whose mode of action is inhibition of the demethylase enzyme that is involved in the synthesis of ergosterol.42 Fluconazole is available in both oral and intravenous formulations, with oral bioavailability greater than 90%. Fluconazole is labeled by the FDA for use in children ≥6 months of age. The volume of distribution in adults is 0.7 L/kg with a low plasma protein-binding (12%). Plasma half-life is ~30 hours in adults.43

The PK properties of fluconazole are well-described in children. The volume of distribution varies with age; it is greatest during the neonatal period and decreases by young adulthood. In children, fluconazole clearance is more rapid than in adults, with a mean half-life of 20 hours. Fluconazole is eliminated renally; therefore, dosing adjustments are necessary in patients with substantial renal impairment.44,45 In the premature infant, we typically reserve dose adjustment for the infant with substantially impaired urine output and elevated creatinine.17

Fluconazole is commonly used to treat candidiasis in infants and is active against the most frequently isolated species of Candida.46 Infants with invasive candidiasis should receive 12 mg/kg/day of fluconazole to achieve an area under the curve of >400 mg*h/L.47 A loading dose of 25 mg/kg on the first day is likely to provide optimal exposure; otherwise, infants may not reach target exposures for several days.18 Fluconazole is also well-absorbed in infants48 and is found in the CSF at 80% of the levels observed in plasma.49

Fluconazole appears to be safe in infants. Laboratory abnormalities in patients receiving fluconazole are uncommon; a transient increase in liver enzymes was seen in <5% of children (n=564; from premature neonatal age to 17 years of age).50 Fluconazole was given to 493 infants in clinical trials to evaluate fluconazole prophylaxis, and no serious adverse events were found.913,51 One trial showed differences in liver enzyme levels for fluconazole patients compared with controls (alanine transaminase 18 IU/ml and 15 IU/ml for treated vs. controls, respectively), but levels returned to baseline levels after the end of therapy.11

Several randomized trials and a number of prospective cohort studies found that antifungal prophylaxis with fluconazole decreased the incidence of invasive candidiasis (Table 2).913,5258 Although fluconazole prophylaxis decreases invasive candidiasis in high-risk populations, it is unknown if prophylaxis decreases overall mortality, decreases candidiasis in low-incidence settings, or what the effects of prophylaxis may be on long-term neurodevelopment. There are also concerns that antifungal prophylaxis may increase the incidence of fluconazole-resistant Candida.61

Table 2.

Fluconazole Prophylaxis Studies

Author/Year Study Design Population N Drug Regimen Outcomes
Aydemir9
2010
RCT VLBW 93 FP
94 NP
91 Placebo
fluconazole: 3 mg/kg q 3 days
nystatin: 1 ml every 8 hours
IC rate 3% FP, 4% NP vs. 17%
placebo (p<0.001)
Similar mortality: 9% FP, 9% NP, 12%
placebo
Rueda57
2010
retrospective cohort <1250 g 252 FP
272 Control
3 mg/kg every q 48 hours IC rate decreased 8% control – 1% FP
(p=0.007)
IC-related mortality decreased 6% to
1% (p=0.003)
Aziz52
2010
retrospective cohort ELBW 163 FP
99 Control
3 mg/kg q 48–72 hours IC rate decreased 7% control – 2% FP
(p=0.045)
Weitkamp58
2008
retrospective cohort <750 g 42 FP
44 Control
3 mg/kg twice weekly IC rate decreased 20% control – 0%
FP (p=0.004)
Similar mortality: 20% control, 26% FP
Healy54
2008
retrospective cohort ELBW 362 FP
206 Control
3 mg/kg q 24–72 hours IC rate decreased 7% control – 2% FP
(p=0.003)
IC-related mortality 2% control, 0% FP
(p=0.1)
Parikh13
2007
RCT VLBW 60 FP
60 Placebo
3 mg/kg q 24–72 hours IC rate 25% placebo – 26% FP
(p=0.835)
Same mortality: 28% in both groups
Manzoni12
2007
RCT VLBW 112 HD
104 LD
106 Placebo
6 mg/kg (HD) or 3 mg/kg (LD)
q 48–72 hours
IC rate decreased 13% (placebo) – 3%
HD (p=0.05) and 13.2% (placebo) –
3.8% LD (p=0.002)
Similar mortality: 8% HD, 10% LD, and
9% placebo
McCrossan56
2007
retrospective cohort VLBW 31 FP
33 Control
6 mg/kg q 24–72 hours IC rate decreased 6% control – 0% FP
(p=0.03)
IC-related mortality: 12% control, 0%
FP (p=0.11)
Manzoni55
2006
retrospective cohort VLBW 225 FP
240 Control
6 mg/kg q 48–72 hours IC rate decreased 17% control – 5%
FP (p<0.0001)
Similar mortality: 11% control, 11% FP
Kaufman59
2005
RCT ELBW 41 FP previous
40 FP twice/week
3 mg/kg twice weekly or q 24–
72 hours
IC rate 5% previous – 3% twice a
week (p=0.68)
Similar mortality: 13% control, 8% FP
(p=8.1%)
Bertini53
2005
retrospective cohort VLBW 136 FP
119 Control
6 mg/kg q 24–72 hours IC rate decreased from 8% control –
0% FP (p=0.003)
Healy60
2005
retrospective cohort ELBW 240 FP
206 Control
3 mg/kg q 24–72 hours IC rate decreased 7% control – 2% FP
(p=0.01)
IC-related mortality: 12% control, 0%
FP (p=0.4)
Kicklighter11
2001
RCT VLBW 53 FP
50 Placebo
6 mg/kg q 24–72 hours Fungal colonization decreased 46%
placebo – 15% FP (p=0.0005)
IC rate or mortality not related
Kaufman10
2001
RCT ELBW 50 FP
50 Placebo
3 mg/kg q 24–72 hours IC rate decreased 20% placebo – 0%
FP (p=0.008)
Similar mortality: 20% placebo, 8% FP
(p=0.22)

FP: fluconazole prophylaxis group; NP: nystatin prophylaxis group; IC: invasive candidiasis; HD: high-dosage group; LD: low-dosage group; RCT: randomized controlled trial.

Voriconazole

Voriconazole, a second-generation triazole, exhibits broad-spectrum antifungal activity against fungal pathogens such as Candida spp., Aspergillus spp., and Cryptococcus neoformans, and less common mold pathogens including several species of Fusarium and Penicillium marneffei.62 Voriconazole has high oral bioavailability (90%) and a mean half-life of 6 hours.62 It is FDA-approved for children ≥12 years of age. CSF penetration is high, and the drug is metabolized by the liver cytochrome P4502C19; genetic polymorphisms of this enzyme play a role in its PK.62,63

Walsh et al. demonstrated a non-linear elimination for the dose range of 4 mg/kg every 12 hours and 8 mg/kg every 12 hours.64 The clearance in children is higher than in adults, and the bioavailability is lower (65%); a dose of 7 mg/kg may provide plasma concentrations comparable to exposures in adults given 4 mg/kg.64,65 Plasma levels of voriconazole in children are highly variable; this is especially true in young infants. Plasma levels should therefore be monitored in a neonate who receives this product.6668 In a study of 10 children with a median age 17 months (range 2 weeks–35 months), voriconazole trough concentrations were highly variable and did not correlate with the dose administered.69

Voriconazole is safe in adults. Children and adults experience the same adverse effects: transient visual disturbances and photosensitivity.64,67,7072 Mild transient elevation of hepatic enzyme levels has also been observed.64,67 Voriconazole clinical trials have not been performed in infants.73 This product should be reserved as a second- (or third-) line agent, primarily in the context of resistance or in the rare case of invasive aspergillosis in the nursery.

Posaconazole

Posaconazole is available in an oral formulation and has extended activity against Candida spp., Aspergillus spp., C. neoformans, and zygomycetes.74,75 Posaconazole is approved for prophylaxis of invasive Aspergillus and Candida infections and for treatment of oropharyngeal candidiasis in patients ≥13 years of age.

Posaconazole has a half-life of 25 hours76 and is excreted unchanged in the feces; renal elimination is minor, and the drug is not a substrate for the cytochrome P-450 enzymatic system.77

Experience with posaconazole in children is limited. In a retrospective study including 15 patients aged 3–17 years, posaconazole was used as salvage therapy for invasive fungal infections in immuno-compromised children and adolescents.78 In a PK study that included 12 patients <18 years of age, posaconazole concentrations in plasma were similar for juvenile and adult patients, following a dose of 400 mg/kg twice daily.79 There are no published studies in infants, but higher doses are likely needed for younger patients.

ECHINOCANDINS

Echinocandins are non-competitive inhibitors of 1,3-β-D-glucan synthase, an enzyme that is necessary for the synthesis of 1,3-β-D glucan. Without this component, the fungal wall cell is compromised.80 The echinocandins have a large molecular weight and poor penetration into the CSF81; however, micafungin and anidulafungin can be given in dosages in which the products successfully achieve maximum killing concentrations inside the CNS. These studies have been conducted in a series of animal model experiments followed by neonatal PK trials.82,83

Micafungin

Micafungin exhibits broad-spectrum activity against clinically important pathogens including azole-resistant Candida albicans.84 It is fungicidal against Candida spp. and fungistatic in vitro against Aspergillus spp.84,85 Micafungin is an intravenous antifungal approved in the United States for adult patients and has been approved for use in children (including infants) in Europe for treatment of invasive candidiasis.86 Trials in patients >16 years of age (and 1 including children from 0 week to 16 years of age) found that micafungin has the same efficacy against invasive candidiasis as amphotericin B,8789 fluconazole,90 or caspofungin91 with fewer adverse events than amphotericin B deoxycholate.87,88

Micafungin has a half-life of approximately 12 hours in adults; the highest drug concentrations are detected in the lungs, liver, spleen, and kidneys.92,93 Micafungin is highly plasma protein-bound. Metabolism occurs mainly in the liver but, because the echinocandins are poor substrates for the cytochrome P450 enzymes, few drug interactions are described. Fecal excretion is the major route of elimination.81

Of the echinocandins, the PK of micafungin is the best described across all pediatric age groups. In children with fever and neutropenia, micafungin (0.5–4 mg/kg/day) demonstrated linear PK, and clearance was inversely related to age.94 To achieve micafungin exposures equivalent to adults, children require dosages >3 mg/kg.19 However, the clearance of micafungin in premature infants is faster than in older children and adults; age-dependent serum protein-binding of micafungin might be responsible for its higher clearance.95

Elevated dosing in the premature infant is thought to be critical for getting micafungin into the central nervous system.82 After administration of >2 mg/kg of micafungin to rabbits, micafungin was detected in most brain compartments (meninges, spinal cord, choroid, cerebrum, cerebellum, and aqueous humor).82 For lower doses (0.5–1.0 mg/kg), micafungin was not detected.82 PK data obtained in 12 premature infants (mean gestational age of 27 weeks) suggest that a micafungin dose of 15 mg/kg/day achieves similar exposures to those observed in adults receiving 5 mg/kg/day.96 A dose of 10 mg/kg/day provided target systemic exposure corresponding to levels adequate to provide CNS penetration and is thought to be the optimal dose for premature and term infants.20,97

The most common adverse events related to micafungin include: gastrointestinal tract manifestations (nausea, diarrhea), hypersensitivity reactions, elevation of liver enzymes, and hypokalemia.89,94 King et al. presented a case of hepatitis related to micafungin in a 44–day-old infant.98 Micafungin doses from 0.75–15 mg/kg/day have been studied in children.21,96,99

Anidulafungin

Anidulafungin has activity against Candida and Aspergillus sp. in adults. It has been used in the treatment of esophageal and systemic candidiasis100102 and is FDA-approved for use in adults. Anidulafungin demonstrates linear PK with a long half-life: approximately 26 hours for adults and 20 hours for children. 22,103 Clearance of anidulafungin appears to be primarily due to slow chemical degradation, and it is eliminated in the feces predominantly as a degradation product. Fecal elimination likely occurs via biliary excretion. Once it is degraded in the blood, it does not require dosage adjustment in subjects with hepatic or renal impairment.104 The tissue concentrations after multiple dosing are highest in the lungs, liver, spleen, and kidneys, with measurable concentrations in the brain tissue.105

Children ages 2–17 years with neutropenia were given anidulafungin (1.5–3 mg/kg loading dose, 0.75–1.5 mg/kg/day maintenance dose) and had exposures similar to adult patients receiving the same weight-adjusted dose; they achieved steady-state plasma concentrations after administration of a loading dose.22 Anidulafungin was administered to 15 patients <18 months of age intravenously as a loading dose of 3 mg/kg on day 1, with daily maintenance dosages of 1.5 mg/kg. Among the patients, there were 8 newborns with a median gestational age of 27 weeks (range 26–39), median birth weight of 1120 g (770–3730), and median postnatal age of 28 days (range 2–451).23 Infants receiving 1.5 mg/kg/day had similar anidulafungin exposures compared with children receiving similar weight-based dosing and adult patients receiving 100 mg/day. 22,23,100 Two patients supported by extracorporeal membrane oxygenation had lower drug exposure.23,106 High-dose anidulafungin (10 mg/kg/day) reduced fungal load in murine brains, but a lower dose (5 mg/kg/day) in the same animal model was not effective.83 The adequate dose for infants to achieve CNS penetration with anidulafungin is unknown.

Anidulafungin is well-tolerated in the pediatric population.22,23 Adverse events occurred in few patients; the most commonly reported adverse event was transient liver function test elevations, and 2 infants showed worsening of baseline bilirubin levels.22,23

Caspofungin

Caspofungin has activity against both Candida sp. and Aspergillus sp.107 Caspofungin is FDA-approved for adults and children >3 months of age. The typical adult dose is 50 mg daily after a 70 mg loading dose.108,109 The metabolism of caspofungin is hepatic, and its half-life is 9–10 hours.110 Patients with renal insufficiency do not need dose adjustments111; however, decrease of daily dosage is necessary in patients with hepatic insufficiency.112

The PK of caspofungin was first evaluated in children in a study of 39 patients aged 2–17 years (29 patients <1 years of age); a dose of 50 mg/m2/day provided similar exposures to an adult dose of 50 mg/kg.24 Caspofungin clearance is increased in children, as demonstrated by lower exposures and shorter half-life (8 hours) compared with adults.24 At the same time, the maximum concentration is higher after a loading dose.25 In children, the main use of caspofungin has been in refractory cases of invasive candidiasis as a salvage or combination therapy.113,114 Most recently, some studies have made use of the medication as the primary treatment for invasive candidiasis115 or for empiric antifungal therapy.116

In infants and toddlers with fever and neutropenia, caspofungin 50 mg/m2/day produced similar steady-state exposures as those observed in older children with the same dose.117 A dose of 25 mg/m2/day in 18 infants <3 months of age resulted in peak concentrations lower than those in older children given 50 mg/m2/day and similar concentrations to those seen in adults administered 50 mg/day.25

The efficacy of caspofungin against CNS candidiasis has not yet been demonstrated.

The most common adverse events are fever, headache, and rash. Increase of hepatic transaminases and hypokalemia was found in few patients.24,116 In a cohort of 18 infants (gestational age 24–41 weeks), 94% of the patients presented 1 or more clinical adverse events; none were considered to be related to caspofungin.25 In a retrospective study with infants (≤3 months of age), caspofungin was not associated with any serious adverse events.118

_ENREF_89_ENREF_94_ENREF_95_ENREF_93_ENREF_96_ENREF_75_ENREF_98

Summary

Invasive fungal infections remain a significant cause of infection-related mortality and morbidity in preterm infants. CNS involvement is the hallmark of neonatal candidiasis, differentiating the disease’s impact on young infants as compared with all other patient populations. As such, CNS involvement substantially influences candidiasis treatment in infants. Over the past decade, the number of antifungal agents in development has grown exponentially, but most are not labeled for use in newborns. More clinical trials and PK studies are required for the new antifungals to be used in infants.

Synopsis.

Invasive fungal infections remain a significant cause of infection-related mortality and morbidity in preterm infants. Central nervous system involvement is the hallmark of neonatal candidiasis, differentiating the disease’s impact on young infants from that among all other patient populations. Over the past decade, the number of antifungal agents in development has grown, but most are not labeled for use in newborns. We summarize the findings of a number of antifungal studies that have been completed to date emphasizing those including infant populations. We conclude that more studies are required for antifungals to be used safely and effectively in infants.

Acknowledgments

Dr. Benjamin receives support from the United States government for his work in pediatric and neonatal clinical pharmacology (1R01HD057956-02, 1R01FD003519-01, 1U10-HD45962-06, 1K24HD058735-01, and Government Contract HHSN275201000003I), the non-profit organization Thrasher Research Foundation for his work in neonatal candidiasis (http://www.thrasherresearch.org), and from industry for neonatal and pediatric drug development (http://www.dcri.duke.edu/research/coi.jsp).

Dr. Smith receives support from NICHD 1K23HD060040-01 and DHHS-1R18AE000028-01 and from industry for neonatal and pediatric drug development (http://www.dcri.duke.edu/research/coi.jsp).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures Dr. Testoni has no disclosures to report.

References

  • 1.Benjamin DK, Jr, Stoll BJ, Gantz MG, et al. Neonatal candidiasis: epidemiology, risk factors, and clinical judgment. Pediatrics. 2010;126(4):e865–73. doi: 10.1542/peds.2009-3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Smith PB, Steinbach WJ, Benjamin DK., Jr Neonatal candidiasis. Infect Dis Clin North Am. 2005;19(3):603–15. doi: 10.1016/j.idc.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 3.Benjamin DK, Jr, Stoll BJ, Fanaroff AA, et al. Neonatal candidiasis among extremely low birth weight infants: risk factors, mortality rates, and neurodevelopmental outcomes at 18 to 22 months. Pediatrics. 2006;117(1):84–92. doi: 10.1542/peds.2004-2292. [DOI] [PubMed] [Google Scholar]
  • 4.Smith PB, Morgan J, Benjamin JD, et al. Excess costs of hospital care associated with neonatal candidemia. Pediatr Infect Dis J. 2007;26(3):197–200. doi: 10.1097/01.inf.0000253973.89097.c0. [DOI] [PubMed] [Google Scholar]
  • 5.Cohen-Wolkowiez M, Smith PB, Mangum B, et al. Neonatal Candida meningitis: significance of cerebrospinal fluid parameters and blood cultures. J Perinatol. 2007;27(2):97–100. doi: 10.1038/sj.jp.7211628. [DOI] [PubMed] [Google Scholar]
  • 6.Fernandez M, Moylett EH, Noyola DE, et al. Candidal meningitis in neonates: a 10-year review. Clin Infect Dis. 2000;31(2):458–63. doi: 10.1086/313973. [DOI] [PubMed] [Google Scholar]
  • 7.Benjamin DK, Jr, Poole C, Steinbach WJ, et al. Neonatal candidemia and end-organ damage: a critical appraisal of the literature using meta-analytic techniques. Pediatrics. 2003;112(3 Pt 1):634–40. doi: 10.1542/peds.112.3.634. [DOI] [PubMed] [Google Scholar]
  • 8.Watt K, Benjamin DK, Jr, Cohen-Wolkowiez M. Pharmacokinetics of antifungal agents in children. Early Hum Dev. 2011;87(Suppl 1):S61–5. doi: 10.1016/j.earlhumdev.2011.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aydemir C, Oguz SS, Dizdar EA, et al. Randomised controlled trial of prophylactic fluconazole versus nystatin for the prevention of fungal colonisation and invasive fungal infection in very low birth weight infants. Arch Dis Child Fetal Neonatal Ed. 2011;96(3):F164–8. doi: 10.1136/adc.2009.178996. [DOI] [PubMed] [Google Scholar]
  • 10.Kaufman D, Boyle R, Hazen KC, et al. Fluconazole prophylaxis against fungal colonization and infection in preterm infants. N Engl J Med. 2001;345(23):1660–6. doi: 10.1056/NEJMoa010494. [DOI] [PubMed] [Google Scholar]
  • 11.Kicklighter SD, Springer SC, Cox T, et al. Fluconazole for prophylaxis against candidal rectal colonization in the very low birth weight infant. Pediatrics. 2001;107(2):293–8. doi: 10.1542/peds.107.2.293. [DOI] [PubMed] [Google Scholar]
  • 12.Manzoni P, Stolfi I, Pugni L, et al. A multicenter, randomized trial of prophylactic fluconazole in preterm neonates. N Engl J Med. 2007;356(24):2483–95. doi: 10.1056/NEJMoa065733. [DOI] [PubMed] [Google Scholar]
  • 13.Parikh TB, Nanavati RN, Patankar CV, et al. Fluconazole prophylaxis against fungal colonization and invasive fungal infection in very low birth weight infants. Indian Pediatr. 2007;44(11):830–7. [PubMed] [Google Scholar]
  • 14.Baley JE, Meyers C, Kliegman RM, et al. Pharmacokinetics, outcome of treatment, and toxic effects of amphotericin B and 5-fluorocytosine in neonates. J Pediatr. 1990;116(5):791–7. doi: 10.1016/s0022-3476(05)82674-5. [DOI] [PubMed] [Google Scholar]
  • 15.Linder N, Klinger G, Shalit I, et al. Treatment of candidaemia in premature infants: comparison of three amphotericin B preparations. J Antimicrob Chemother. 2003;52(4):663–7. doi: 10.1093/jac/dkg419. [DOI] [PubMed] [Google Scholar]
  • 16.Juster-Reicher A, Flidel-Rimon O, Amitay M, et al. High-dose liposomal amphotericin B in the therapy of systemic candidiasis in neonates. Eur J Clin Microbiol Infect Dis. 2003;22(10):603–7. doi: 10.1007/s10096-003-0993-4. [DOI] [PubMed] [Google Scholar]
  • 17.Wade KC, Wu D, Kaufman DA, et al. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother. 2008;52(11):4043–9. doi: 10.1128/AAC.00569-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Piper L, Smith PB, Hornik CP, et al. Fluconazole loading dose pharmacokinetics and safety in infants. Pediatr Infect Dis J. 2011;30(5):375–8. doi: 10.1097/INF.0b013e318202cbb3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hope WW, Seibel NL, Schwartz CL, et al. Population pharmacokinetics of micafungin in pediatric patients and implications for antifungal dosing. Antimicrob Agents Chemother. 2007;51(10):3714–9. doi: 10.1128/AAC.00398-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Benjamin DK, Jr, Smith PB, Arrieta A, et al. Safety and pharmacokinetics of repeat-dose micafungin in young infants. Clin Pharmacol Ther. 2010;87(1):93–9. doi: 10.1038/clpt.2009.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Heresi GP, Gerstmann DR, Reed MD, et al. The pharmacokinetics and safety of micafungin, a novel echinocandin, in premature infants. Pediatr Infect Dis J. 2006;25(12):1110–5. doi: 10.1097/01.inf.0000245103.07614.e1. [DOI] [PubMed] [Google Scholar]
  • 22.Benjamin DK, Jr, Driscoll T, Seibel NL, et al. Safety and pharmacokinetics of intravenous anidulafungin in children with neutropenia at high risk for invasive fungal infections. Antimicrob Agents Chemother. 2006;50(2):632–8. doi: 10.1128/AAC.50.2.632-638.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cohen-Wolkowiez M, Benjamin DK, Jr, Piper L, et al. Safety and pharmacokinetics of multiple-dose anidulafungin in infants and neonates. Clin Pharmacol Ther. 2011;89(5):702–7. doi: 10.1038/clpt.2011.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Walsh TJ, Adamson PC, Seibel NL, et al. Pharmacokinetics, safety, and tolerability of caspofungin in children and adolescents. Antimicrob Agents Chemother. 2005;49(11):4536–45. doi: 10.1128/AAC.49.11.4536-4545.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Saez-Llorens X, Macias M, Maiya P, et al. Pharmacokinetics and safety of caspofungin in neonates and infants less than 3 months of age. Antimicrob Agents Chemother. 2009;53(3):869–75. doi: 10.1128/AAC.00868-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jeon GW, Koo SH, Lee JH, et al. A comparison of AmBisome to amphotericin B for treatment of systemic candidiasis in very low birth weight infants. Yonsei Med J. 2007;48(4):619–26. doi: 10.3349/ymj.2007.48.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Janknegt R, de Marie S, Bakker-Woudenberg IA, et al. Liposomal and lipid formulations of amphotericin B. Clinical pharmacokinetics. Clin Pharmacokinet. 1992;23(4):279–91. doi: 10.2165/00003088-199223040-00004. [DOI] [PubMed] [Google Scholar]
  • 28.Holler B, Omar SA, Farid MD, et al. Effects of fluid and electrolyte management on amphotericin B-induced nephrotoxicity among extremely low birth weight infants. Pediatrics. 2004;113(6):e608–16. doi: 10.1542/peds.113.6.e608. [DOI] [PubMed] [Google Scholar]
  • 29.Makhoul IR, Kassis I, Smolkin T, et al. Review of 49 neonates with acquired fungal sepsis: further characterization. Pediatrics. 2001;107(1):61–6. doi: 10.1542/peds.107.1.61. [DOI] [PubMed] [Google Scholar]
  • 30.Le J, Adler-Shohet FC, Nguyen C, et al. Nephrotoxicity associated with amphotericin B deoxycholate in neonates. Pediatr Infect Dis J. 2009;28(12):1061–3. doi: 10.1097/INF.0b013e3181af6201. [DOI] [PubMed] [Google Scholar]
  • 31.Kotwani RN, Gokhale PC, Bodhe PV, et al. A comparative study of plasma concentrations of liposomal amphotericin B (L-AMP-LRC-1) in adults, children and neonates. Int J Pharm. 2002;238(1-2):11–5. doi: 10.1016/s0378-5173(02)00066-2. [DOI] [PubMed] [Google Scholar]
  • 32.Lee JW, Amantea MA, Francis PA, et al. Pharmacokinetics and safety of a unilamellar liposomal formulation of amphotericin B (AmBisome) in rabbits. Antimicrob Agents Chemother. 1994;38(4):713–8. doi: 10.1128/aac.38.4.713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wurthwein G, Groll AH, Hempel G, et al. Population pharmacokinetics of amphotericin B lipid complex in neonates. Antimicrob Agents Chemother. 2005;49(12):5092–8. doi: 10.1128/AAC.49.12.5092-5098.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Scarcella A, Pasquariello MB, Giugliano B, et al. Liposomal amphotericin B treatment for neonatal fungal infections. Pediatr Infect Dis J. 1998;17(2):146–8. doi: 10.1097/00006454-199802000-00013. [DOI] [PubMed] [Google Scholar]
  • 35.Anaissie EJ, Mattiuzzi GN, Miller CB, et al. Treatment of invasive fungal infections in renally impaired patients with amphotericin B colloidal dispersion. Antimicrob Agents Chemother. 1998;42(3):606–11. doi: 10.1128/aac.42.3.606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.White MH, Bowden RA, Sandler ES, et al. Randomized, double-blind clinical trial of amphotericin B colloidal dispersion vs. amphotericin B in the empirical treatment of fever and neutropenia. Clin Infect Dis. 1998;27(2):296–302. doi: 10.1086/514672. [DOI] [PubMed] [Google Scholar]
  • 37.Bennet JE. Flucytosine. Ann Intern Med. 1977;86(3):319–21. doi: 10.7326/0003-4819-86-3-319. [DOI] [PubMed] [Google Scholar]
  • 38.Polak A, Scholer HJ. Mode of action of 5-fluorocytosine and mechanisms of resistance. Chemotherapy. 1975;21(3-4):113–30. doi: 10.1159/000221854. [DOI] [PubMed] [Google Scholar]
  • 39.Smego RA, Jr, Perfect JR, Durack DT. Combined therapy with amphotericin B and 5-fluorocytosine for Candida meningitis. Rev Infect Dis. 1984;6(6):791–801. doi: 10.1093/clinids/6.6.791. [DOI] [PubMed] [Google Scholar]
  • 40.Vermes A, van Der Sijs H, Guchelaar HJ. Flucytosine: correlation between toxicity and pharmacokinetic parameters. Chemotherapy. 2000;46(2):86–94. doi: 10.1159/000007260. [DOI] [PubMed] [Google Scholar]
  • 41.Soltani M, Tobin CM, Bowker KE, et al. Evidence of excessive concentrations of 5-flucytosine in children aged below 12 years: a 12-year review of serum concentrations from a UK clinical assay reference laboratory. Int J Antimicrob Agents. 2006;28(6):574–7. doi: 10.1016/j.ijantimicag.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • 42.Galgiani JN. Fluconazole, a new antifungal agent. Ann Intern Med. 1990;113(3):177–9. doi: 10.7326/0003-4819-113-3-177. [DOI] [PubMed] [Google Scholar]
  • 43.Brammer KW, Farrow PR, Faulkner JK. Pharmacokinetics and tissue penetration of fluconazole in humans. Rev Infect Dis. 1990;12(Suppl 3):S318–26. doi: 10.1093/clinids/12.supplement_3.s318. [DOI] [PubMed] [Google Scholar]
  • 44.Berl T, Wilner KD, Gardner M, et al. Pharmacokinetics of fluconazole in renal failure. J Am Soc Nephrol. 1995;6(2):242–7. doi: 10.1681/ASN.V62242. [DOI] [PubMed] [Google Scholar]
  • 45.Brammer KW, Coates PE. Pharmacokinetics of fluconazole in pediatric patients. Eur J Clin Microbiol Infect Dis. 1994;13(4):325–9. doi: 10.1007/BF01974613. [DOI] [PubMed] [Google Scholar]
  • 46.Hajjeh RA, Sofair AN, Harrison LH, et al. Incidence of bloodstream infections due to Candida species and in vitro susceptibilities of isolates collected from 1998 to 2000 in a population-based active surveillance program. J Clin Microbiol. 2004;42(4):1519–27. doi: 10.1128/JCM.42.4.1519-1527.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wade KC, Benjamin DK, Jr, Kaufman DA, et al. Fluconazole dosing for the prevention or treatment of invasive candidiasis in young infants. Pediatr Infect Dis J. 2009;28(8):717–23. doi: 10.1097/INF.0b013e31819f1f50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nahata MC, Tallian KB, Force RW. Pharmacokinetics of fluconazole in young infants. Eur J Drug Metab Pharmacokinet. 1999;24(2):155–7. doi: 10.1007/BF03190361. [DOI] [PubMed] [Google Scholar]
  • 49.Wildfeuer A, Laufen H, Schmalreck AF, et al. Fluconazole: comparison of pharmacokinetics, therapy and in vitro susceptibility. Mycoses. 1997;40(7-8):259–65. doi: 10.1111/j.1439-0507.1997.tb00230.x. [DOI] [PubMed] [Google Scholar]
  • 50.Novelli V, Holzel H. Safety and tolerability of fluconazole in children. Antimicrob Agents Chemother. 1999;43(8):1955–60. doi: 10.1128/aac.43.8.1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kaufman D, Boyle R, Hazen KC, et al. Twice weekly fluconazole prophylaxis for prevention of invasive Candida infection in high-risk infants of <1000 grams birth weight. J Pediatr. 2005;147(2):172–9. doi: 10.1016/j.jpeds.2005.03.036. [DOI] [PubMed] [Google Scholar]
  • 52.Aziz M, Patel AL, Losavio J, et al. Efficacy of fluconazole prophylaxis for prevention of invasive fungal infection in extremely low birth weight infants. Pediatr Infect Dis J. 2010;29(4):352–6. doi: 10.1097/INF.0b013e3181bf8eb1. [DOI] [PubMed] [Google Scholar]
  • 53.Bertini G, Perugi S, Dani C, et al. Fluconazole prophylaxis prevents invasive fungal infection in high-risk, very low birth weight infants. J Pediatr. 2005;147(2):162–5. doi: 10.1016/j.jpeds.2005.02.020. [DOI] [PubMed] [Google Scholar]
  • 54.Healy CM, Campbell JR, Zaccaria E, et al. Fluconazole prophylaxis in extremely low birth weight neonates reduces invasive candidiasis mortality rates without emergence of fluconazole-resistant Candida species. Pediatrics. 2008;121(4):703–10. doi: 10.1542/peds.2007-1130. [DOI] [PubMed] [Google Scholar]
  • 55.Manzoni P, Arisio R, Mostert M, et al. Prophylactic fluconazole is effective in preventing fungal colonization and fungal systemic infections in preterm neonates: a single-center, 6-year, retrospective cohort study. Pediatrics. 2006;117(1):e22–32. doi: 10.1542/peds.2004-2227. [DOI] [PubMed] [Google Scholar]
  • 56.McCrossan BA, McHenry E, O’Neill F, et al. Selective fluconazole prophylaxis in high-risk babies to reduce invasive fungal infection. Arch Dis Child Fetal Neonatal Ed. 2007;92(6):F454–8. doi: 10.1136/adc.2006.094359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Rueda K, Moreno MT, Espinosa M, et al. Impact of routine fluconazole prophylaxis for premature infants with birth weights of less than 1250 grams in a developing country. Pediatr Infect Dis J. 2010;29(11):1050–2. doi: 10.1097/INF.0b013e3181e9dea2. [DOI] [PubMed] [Google Scholar]
  • 58.Weitkamp JH, Ozdas A, LaFleur B, et al. Fluconazole prophylaxis for prevention of invasive fungal infections in targeted highest risk preterm infants limits drug exposure. J Perinatol. 2008;28(6):405–11. doi: 10.1038/sj.jp.7211914. [DOI] [PubMed] [Google Scholar]
  • 59.Kaufman D, Boyle R, Hazen KC, et al. Twice weekly fluconazole prophylaxis for prevention of invasive Candida infection in high-risk infants of <1000 grams birth weight. J Pediatr. 2005;147(2):172–9. doi: 10.1016/j.jpeds.2005.03.036. [DOI] [PubMed] [Google Scholar]
  • 60.Healy CM, Baker CJ, Zaccaria E, et al. Impact of fluconazole prophylaxis on incidence and outcome of invasive candidiasis in a neonatal intensive care unit. J Pediatr. 2005;147(2):166–71. doi: 10.1016/j.jpeds.2005.04.016. [DOI] [PubMed] [Google Scholar]
  • 61.Manzoni P, Leonessa M, Galletto P, et al. Routine use of fluconazole prophylaxis in a neonatal intensive care unit does not select natively fluconazole-resistant Candida subspecies. Pediatr Infect Dis J. 2008;27(8):731–7. doi: 10.1097/INF.0b013e318170bb0c. [DOI] [PubMed] [Google Scholar]
  • 62.Sheehan DJ, Hitchcock CA, Sibley CM. Current and emerging azole antifungal agents. Clin Microbiol Rev. 1999;12(1):40–79. doi: 10.1128/cmr.12.1.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Walsh TJ, Karlsson MO, Driscoll T, et al. Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration. Antimicrob Agents Chemother. 2004;48(6):2166–72. doi: 10.1128/AAC.48.6.2166-2172.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Walsh TJ, Driscoll T, Milligan PA, et al. Pharmacokinetics, safety, and tolerability of voriconazole in immunocompromised children. Antimicrob Agents Chemother. 2010;54(10):4116–23. doi: 10.1128/AAC.00896-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Michael C, Bierbach U, Frenzel K, et al. Voriconazole pharmacokinetics and safety in immunocompromised children compared to adult patients. Antimicrob Agents Chemother. 2010;54(8):3225–32. doi: 10.1128/AAC.01731-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Spriet I, Cosaert K, Renard M, et al. Voriconazole plasma levels in children are highly variable. Eur J Clin Microbiol Infect Dis. 2011;30(2):283–7. doi: 10.1007/s10096-010-1079-8. [DOI] [PubMed] [Google Scholar]
  • 67.Shima H, Miharu M, Osumi T, et al. Differences in voriconazole trough plasma concentrations per oral dosages between children younger and older than 3 years of age. Pediatr Blood Cancer. 2010;54(7):1050–2. doi: 10.1002/pbc.22451. [DOI] [PubMed] [Google Scholar]
  • 68.Bruggemann RJ, van der Linden JW, Verweij PE, et al. Impact of therapeutic drug monitoring of voriconazole in a pediatric population. Pediatr Infect Dis J. 2011;30(6):533–4. doi: 10.1097/INF.0b013e318204d227. [DOI] [PubMed] [Google Scholar]
  • 69.Doby EH, Benjamin DK, Jr, Blaschke AJ, et al. Therapeutic drug monitoring of voriconazole in children less than 3 years of age: a case report and summary of pharmacokinetic data for 10 children. Pediatr Infect Dis J. doi: 10.1097/INF.0b013e31824acc33. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cowen EW, Nguyen JC, Miller DD, et al. Chronic phototoxicity and aggressive squamous cell carcinoma of the skin in children and adults during treatment with voriconazole. J Am Acad Dermatol. 2010;62(1):31–7. doi: 10.1016/j.jaad.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rubenstein M, Levy ML, Metry D. Voriconazole-induced retinoid-like photosensitivity in children. Pediatr Dermatol. 2004;21(6):675–8. doi: 10.1111/j.0736-8046.2004.21614.x. [DOI] [PubMed] [Google Scholar]
  • 72.Purkins L, Wood N, Greenhalgh K, et al. The pharmacokinetics and safety of intravenous voriconazole—a novel wide-spectrum antifungal agent. Br J Clin Pharmacol. 2003;56(Suppl 1):2–9. doi: 10.1046/j.1365-2125.2003.01992.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kohli V, Taneja V, Sachdev P, et al. Voriconazole in newborns. Indian Pediatr. 2008;45(3):236–8. [PubMed] [Google Scholar]
  • 74.Uchida K, Yokota N, Yamaguchi H. In vitro antifungal activity of posaconazole against various pathogenic fungi. Int J Antimicrob Agents. 2001;18(2):167–72. doi: 10.1016/s0924-8579(01)00363-6. [DOI] [PubMed] [Google Scholar]
  • 75.Sun QN, Fothergill AW, McCarthy DI, et al. In vitro activities of posaconazole, itraconazole, voriconazole, amphotericin B, and fluconazole against 37 clinical isolates of zygomycetes. Antimicrob Agents Chemother. 2002;46(5):1581–2. doi: 10.1128/AAC.46.5.1581-1582.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Courtney R, Pai S, Laughlin M, et al. Pharmacokinetics, safety, and tolerability of oral posaconazole administered in single and multiple doses in healthy adults. Antimicrob Agents Chemother. 2003;47(9):2788–95. doi: 10.1128/AAC.47.9.2788-2795.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Krieter P, Flannery B, Musick T, et al. Disposition of posaconazole following single-dose oral administration in healthy subjects. Antimicrob Agents Chemother. 2004;48(9):3543–51. doi: 10.1128/AAC.48.9.3543-3551.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lehrnbecher T, Attarbaschi A, Duerken M, et al. Posaconazole salvage treatment in paediatric patients: a multicentre survey. Eur J Clin Microbiol Infect Dis. 2010;29(8):1043–5. doi: 10.1007/s10096-010-0957-4. [DOI] [PubMed] [Google Scholar]
  • 79.Krishna G, AbuTarif M, Xuan F, et al. Pharmacokinetics of oral posaconazole in neutropenic patients receiving chemotherapy for acute myelogenous leukemia or myelodysplastic syndrome. Pharmacotherapy. 2008;28(10):1223–32. doi: 10.1592/phco.28.10.1223. [DOI] [PubMed] [Google Scholar]
  • 80.Sucher AJ, Chahine EB, Balcer HE. Echinocandins: the newest class of antifungals. Ann Pharmacother. 2009;43(10):1647–57. doi: 10.1345/aph.1M237. [DOI] [PubMed] [Google Scholar]
  • 81.Denning DW. Echinocandin antifungal drugs. Lancet. 2003;362(9390):1142–51. doi: 10.1016/S0140-6736(03)14472-8. [DOI] [PubMed] [Google Scholar]
  • 82.Hope WW, Mickiene D, Petraitis V, et al. The pharmacokinetics and pharmacodynamics of micafungin in experimental hematogenous Candida meningoencephalitis: implications for echinocandin therapy in neonates. J Infect Dis. 2008;197(1):163–71. doi: 10.1086/524063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kang CI, Rouse MS, Mandrekar JN, et al. Anidulafungin treatment of candidal central nervous system infection in a murine model. Antimicrob Agents Chemother. 2009;53(8):3576–8. doi: 10.1128/AAC.00646-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tawara S, Ikeda F, Maki K, et al. In vitro activities of a new lipopeptide antifungal agent, FK463, against a variety of clinically important fungi. Antimicrob Agents Chemother. 2000;44(1):57–62. doi: 10.1128/aac.44.1.57-62.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hatano K, Morishita Y, Nakai T, et al. Antifungal mechanism of FK463 against Candida albicans and Aspergillus fumigatus. J Antibiot (Tokyo) 2002;55(2):219–22. doi: 10.7164/antibiotics.55.219. [DOI] [PubMed] [Google Scholar]
  • 86.Infante-Lopez ME, Rojo-Conejo P. Micafungin for the treatment of neonatal invasive candidiasis. Rev Iberoam Micol. 2009;26(1):56–61. doi: 10.1016/S1130-1406(09)70009-9. [DOI] [PubMed] [Google Scholar]
  • 87.Dupont BF, Lortholary O, Ostrosky-Zeichner L, et al. Treatment of candidemia and invasive candidiasis in the intensive care unit: post hoc analysis of a randomized, controlled trial comparing micafungin and liposomal amphotericin B. Crit Care. 2009;13(5):R159. doi: 10.1186/cc8117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kuse ER, Chetchotisakd P, da Cunha CA, et al. Micafungin versus liposomal amphotericin B for candidaemia and invasive candidosis: a phase III randomised double-blind trial. Lancet. 2007;369(9572):1519–27. doi: 10.1016/S0140-6736(07)60605-9. [DOI] [PubMed] [Google Scholar]
  • 89.Queiroz-Telles F, Berezin E, Leverger G, et al. Micafungin versus liposomal amphotericin B for pediatric patients with invasive candidiasis: substudy of a randomized double-blind trial. Pediatr Infect Dis J. 2008;27(9):820–6. doi: 10.1097/INF.0b013e31817275e6. [DOI] [PubMed] [Google Scholar]
  • 90.van Burik JA, Ratanatharathorn V, Stepan DE, et al. Micafungin versus fluconazole for prophylaxis against invasive fungal infections during neutropenia in patients undergoing hematopoietic stem cell transplantation. Clin Infect Dis. 2004;39(10):1407–16. doi: 10.1086/422312. [DOI] [PubMed] [Google Scholar]
  • 91.Pappas PG, Rotstein CM, Betts RF, et al. Micafungin versus caspofungin for treatment of candidemia and other forms of invasive candidiasis. Clin Infect Dis. 2007;45(7):883–93. doi: 10.1086/520980. [DOI] [PubMed] [Google Scholar]
  • 92.Groll AH, Mickiene D, Petraitis V, et al. Compartmental pharmacokinetics and tissue distribution of the antifungal echinocandin lipopeptide micafungin (FK463) in rabbits. Antimicrob Agents Chemother. 2001;45(12):3322–7. doi: 10.1128/AAC.45.12.3322-3327.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yamada N, Kumada K, Kishino S, et al. Distribution of micafungin in the tissue fluids of patients with invasive fungal infections. J Infect Chemother. 2011;17(5):731–4. doi: 10.1007/s10156-011-0240-3. [DOI] [PubMed] [Google Scholar]
  • 94.Seibel NL, Schwartz C, Arrieta A, et al. Safety, tolerability, and pharmacokinetics of micafungin (FK463) in febrile neutropenic pediatric patients. Antimicrob Agents Chemother. 2005;49(8):3317–24. doi: 10.1128/AAC.49.8.3317-3324.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yanni SB, Smith PB, Benjamin DK, Jr, et al. Higher clearance of micafungin in neonates compared with adults: role of age-dependent micafungin serum binding. Biopharm Drug Dispos. 2011;32(4):222–32. doi: 10.1002/bdd.752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Smith PB, Walsh TJ, Hope W, et al. Pharmacokinetics of an elevated dosage of micafungin in premature neonates. Pediatr Infect Dis J. 2009;28(5):412–5. doi: 10.1097/INF.0b013e3181910e2d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hope WW, Smith PB, Arrieta A, et al. Population pharmacokinetics of micafungin in neonates and young infants. Antimicrob Agents Chemother. 2010;54(6):2633–7. doi: 10.1128/AAC.01679-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.King KY, Edwards MS, Word BM. Hepatitis associated with micafungin use in a preterm infant. J Perinatol. 2009;29(4):320–2. doi: 10.1038/jp.2008.197. [DOI] [PubMed] [Google Scholar]
  • 99.Natarajan G, Lulic-Botica M, Aranda JV. Refractory neonatal candidemia and high-dose micafungin pharmacotherapy. J Perinatol. 2009;29(11):738–43. doi: 10.1038/jp.2009.75. [DOI] [PubMed] [Google Scholar]
  • 100.Pfaller MA, Diekema DJ, Boyken L, et al. Effectiveness of anidulafungin in eradicating Candida species in invasive candidiasis. Antimicrob Agents Chemother. 2005;49(11):4795–7. doi: 10.1128/AAC.49.11.4795-4797.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Vazquez JA, Schranz JA, Clark K, et al. A phase 2, open-label study of the safety and efficacy of intravenous anidulafungin as a treatment for azole-refractory mucosal candidiasis. J Acquir Immune Defic Syndr. 2008;48(3):304–9. doi: 10.1097/QAI.0b013e31817af47a. [DOI] [PubMed] [Google Scholar]
  • 102.Reboli AC, Rotstein C, Pappas PG, et al. Anidulafungin versus fluconazole for invasive candidiasis. N Engl J Med. 2007;356(24):2472–82. doi: 10.1056/NEJMoa066906. [DOI] [PubMed] [Google Scholar]
  • 103.Dowell JA, Knebel W, Ludden T, et al. Population pharmacokinetic analysis of anidulafungin, an echinocandin antifungal. J Clin Pharmacol. 2004;44(6):590–8. doi: 10.1177/0091270004265644. [DOI] [PubMed] [Google Scholar]
  • 104.Dowell JA, Stogniew M, Krause D, et al. Anidulafungin does not require dosage adjustment in subjects with varying degrees of hepatic or renal impairment. J Clin Pharmacol. 2007;47(4):461–70. doi: 10.1177/0091270006297227. [DOI] [PubMed] [Google Scholar]
  • 105.Damle B, Stogniew M, Dowell J. Pharmacokinetics and tissue distribution of anidulafungin in rats. Antimicrob Agents Chemother. 2008;52(7):2673–6. doi: 10.1128/AAC.01596-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Leitner JM, Meyer B, Fuhrmann V, et al. Multiple-dose pharmacokinetics of anidulafungin during continuous venovenous haemofiltration. J Antimicrob Chemother. 2011;66(4):880–4. doi: 10.1093/jac/dkq545. [DOI] [PubMed] [Google Scholar]
  • 107.Vazquez JA, Lynch M, Boikov D, et al. In vitro activity of a new pneumocandin antifungal, L-743,872, against azole-susceptible and -resistant Candida species. Antimicrob Agents Chemother. 1997;41(7):1612–4. doi: 10.1128/aac.41.7.1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Arathoon EG, Gotuzzo E, Noriega LM, et al. Randomized, double-blind, multicenter study of caspofungin versus amphotericin B for treatment of oropharyngeal and esophageal candidiases. Antimicrob Agents Chemother. 2002;46(2):451–7. doi: 10.1128/AAC.46.2.451-457.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Villanueva A, Arathoon EG, Gotuzzo E, et al. A randomized double-blind study of caspofungin versus amphotericin for the treatment of candidal esophagitis. Clin Infect Dis. 2001;33(9):1529–35. doi: 10.1086/323401. [DOI] [PubMed] [Google Scholar]
  • 110.Stone JA, Holland SD, Wickersham PJ, et al. Single- and multiple-dose pharmacokinetics of caspofungin in healthy men. Antimicrob Agents Chemother. 2002;46(3):739–45. doi: 10.1128/AAC.46.3.739-745.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sable CA, Nguyen BY, Chodakewitz JA, et al. Safety and tolerability of caspofungin acetate in the treatment of fungal infections. Transpl Infect Dis. 2002;4(1):25–30. doi: 10.1034/j.1399-3062.2002.01004.x. [DOI] [PubMed] [Google Scholar]
  • 112.Mistry GC, Migoya E, Deutsch PJ, et al. Single- and multiple-dose administration of caspofungin in patients with hepatic insufficiency: implications for safety and dosing recommendations. J Clin Pharmacol. 2007;47(8):951–61. doi: 10.1177/0091270007303764. [DOI] [PubMed] [Google Scholar]
  • 113.Cesaro S, Giacchino M, Locatelli F, et al. Safety and efficacy of a caspofungin-based combination therapy for treatment of proven or probable aspergillosis in pediatric hematological patients. BMC Infect Dis. 2007;7:28. doi: 10.1186/1471-2334-7-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Merlin E, Galambrun C, Ribaud P, et al. Efficacy and safety of caspofungin therapy in children with invasive fungal infections. Pediatr Infect Dis J. 2006;25(12):1186–8. doi: 10.1097/01.inf.0000246844.42159.a0. [DOI] [PubMed] [Google Scholar]
  • 115.Zaoutis TE, Jafri HS, Huang LM, et al. A prospective, multicenter study of caspofungin for the treatment of documented Candida or Aspergillus infections in pediatric patients. Pediatrics. 2009;123(3):877–84. doi: 10.1542/peds.2008-1158. [DOI] [PubMed] [Google Scholar]
  • 116.Maertens JA, Madero L, Reilly AF, et al. A randomized, double-blind, multicenter study of caspofungin versus liposomal amphotericin B for empiric antifungal therapy in pediatric patients with persistent fever and neutropenia. Pediatr Infect Dis J. 2010;29(5):415–20. doi: 10.1097/INF.0b013e3181da2171. [DOI] [PubMed] [Google Scholar]
  • 117.Neely M, Jafri HS, Seibel N, et al. Pharmacokinetics and safety of caspofungin in older infants and toddlers. Antimicrob Agents Chemother. 2009;53(4):1450–6. doi: 10.1128/AAC.01027-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Natarajan G, Lulic-Botica M, Rongkavilit C, et al. Experience with caspofungin in the treatment of persistent fungemia in neonates. J Perinatol. 2005;25(12):770–7. doi: 10.1038/sj.jp.7211380. [DOI] [PubMed] [Google Scholar]

RESOURCES