Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 1.
Published in final edited form as: Drugs. 2014 Jun;74(8):891–909. doi: 10.1007/s40265-014-0227-3

Pharmacokinetics and Pharmacodynamics of Antifungals in Children: Clinical Implications

Julie Autmizguine 1, Jeffrey T Guptill 1, Michael Cohen-Wolkowiez 1, Daniel K Benjamin Jr 1, Edmund V Capparelli 2
PMCID: PMC4073603  NIHMSID: NIHMS599936  PMID: 24872147

Abstract

Invasive fungal disease (IFD) remains life-threatening in premature infants and immunocompromised children despite the recent development of new antifungal agents. Optimal dosing of antifungals is one of the few factors clinicians can control to improve outcomes of IFD. However, dosing in children cannot be extrapolated from adult data because IFD pathophysiology, immune response, and drug disposition differ from adults. We critically examined the literature on pharmacokinetics (PK) and pharmacodynamics (PD) of antifungal agents and highlight recent developments in treating pediatric IFD.

To match adult exposure in pediatric patients, dosing adjustment is necessary for almost all antifungals. In young infants, the maturation of renal and metabolic functions occurs rapidly and can significantly influence drug exposure. Fluconazole clearance doubles from birth to 28 days of life and, beyond the neonatal period, agents like fluconazole, voriconazole, and micafungin require higher dosing than in adults due to faster clearance in children. As a result, dosing recommendations are specific to bracketed ranges of age.

Pharmacodynamics principles of antifungals mostly rely on in vitro and in vivo models but very few pharmacodynamics studies specifically address IFD in children. Exposure-response relationship may differ in younger children compared with adults, especially in infants with invasive candidiasis who are at higher risk of disseminated disease and meningoencephalitis, and by extension severe neurodevelopmental impairment. Micafungin is the only antifungal agent for which a specific target of exposure was proposed based on a neonatal hematogenous Candida meningoencephalitis animal model.

In this review, we found that pediatric data on drug disposition of newer triazoles and echinocandins are lacking, dosing of older antifungals such as fluconazole and amphotericin B products still need optimization in young infants, and that target PK/PD indices need to be clinically validated for almost all antifungals in children. A better understanding of age-specific PK and PD of new antifungals in infants and children will help improve clinical outcomes of IFD by informing dosing and identifying future research areas.

Introduction

Invasive fungal diseases (IFD) cause significant mortality and morbidity in children. Candida sp. occur more commonly in children than adults and are a major cause of IFD [1] with a high mortality of 34% in very-low-birth-weight infants [2]. Invasive aspergillosis (IA) is another important cause of IFD in immunocompromised children, resulting in unacceptably high mortality despite antifungal therapy (nearly 50%) [3].

Therapeutic options for IFD are evolving, and several antifungal classes are available to clinicians. Pharmacokinetics (PK), pharmacodynamics (PD), and safety data are predominantly available in adults. However, IFD pathophysiology may differ in children; for example, one of the characteristics of neonatal candidiasis is the high frequency of meningoencephalitis reported in 8 to 28% of neonates with invasive candidiasis, likely due to immature immune system and more permeable blood brain barrier [4-6]. This incidence, however, is underreported given the difficulties in growing Candida in the microbiology laboratory and lack of available brain tissue samples for culture. In addition to pathophysiology differences, change in PK in children compared with that in adults might lead to suboptimal drug exposure or increase in toxicity. In this review, we critically examined the literature on PK and PD of systemic antifungal agents in the pediatric population. A better understanding of these pharmacological concepts will help optimize and personalize antifungal therapy in children and identify areas of future research. The following sources were searched: MEDLINE, clinicaltrials.gov, dailymed.nlm.nih.gov, ema.europa.eu and international proceedings and abstracts from the earliest record to 15 November 2013. The search strategy included the following key words: ‘pharmacokinetics’, ‘pharmacodynamics’, ‘antifungal’, amphotericin B’, ‘liposomal amphotericin B’, ‘deoxycholate amphotericin B’, ‘amphotericin B lipid complex’, ‘amphotericin B colloidal dispersion’, ‘fluconazole’, ‘itraconaozle’, ‘voriconazole’, ‘posaconazole’, ‘triazoles’, ‘ravuconazole’, ‘isavuconazole’, ‘albaconazole’, ‘echinocandins’, ‘micafungin’, ‘caspofungin’, ‘anidulafungin’, ‘aminocandin’, ‘5-flucytosine’, ‘flucytosine’, ‘children’, ‘infants’, ‘neonates’. Electronic searches were supplemented by hand-searching the reference lists of previous systematic reviews. The search was restricted to trials published in English.

1. Polyenes

The polyene macrolide class includes amphotericin B deoxycholate (AmB) and newer lipid-based formulations: amphotericin B lipid complex (ABLC), amphotericin B colloidal dispersion (ABCD), and liposomal amphotericin B (L-AmB). Although these agents are among the oldest class of antifungals and are associated with frequent toxicity, they still play a major role in the treatment of IFD in children [7]. The biggest advantage of polyenes is their wide spectrum of activity.

1.1. Amphotericin B Deoxycholate

By binding ergosterol, a component of the fungus cell wall, AmB increases membrane permeability and induces cell death [8, 9]. In vitro fungicidal activity has been demonstrated against a wide spectrum of fungi, including Candida sp., Aspergillus sp., Zygomycetes, and dimorphic fungi [10]. Although occasional resistance has been reported for multiple fungi species, clinically significant resistance is rare and is mostly encountered in A. terreus, C. lusitaniae, Trichosporon sp., Scedosporium sp., and Fusarium sp. [11-14].

AmB activity is concentration-dependent with a prolonged post-antifungal effect (PAFE>12h) [15]. In vitro, its fungicidal effect was observed when maximum concentration (Cmax) was 4 times greater than minimum inhibitory concentration (MIC) for C. albicans, whereas in candidiasis murine models, maximal activity occurred at Cmax:MIC of 2.4 [10, 16, 17]. In vitro susceptibility testing has been correlated with clinical outcome in adults with invasive aspergillosis, but no correlation was established in adults with invasive candidiasis [18, 19]. No PK/PD indices are validated in children.

Due to poor oral absorption [20], systemic AmB is administered parenterally. AmB circulates in plasma highly bound to lipoproteins and is taken up by reticulo-endothelial organs, especially the liver [21]. In adults, cerebrospinal fluid (CSF) penetration is limited (2–4% of serum concentrations) [22]. Immaturity in the blood-brain barrier can lead to enhanced central nervous system (CNS) penetration in premature infants where CSF: plasma ratios can reach 40– 90% [23]. AmB is slowly excreted in urine and bile, and metabolism pathways are not well defined [21]. PK in children is characterized by a lower volume of distribution (V) (0.4–3.1 L/kg) and faster clearance (CL) (0.03–0.22 L/kg/h) compared with adults (V: 4L/kg; CL: 0.03 L/kg/h) (Table 1) [23-27]. Although most PK studies in children support a dosage regimen of 0.5–1 mg/kg/day [23-25, 27], a population PK analysis suggested that younger children might be underexposed with 1 mg/kg/day while older children may be overdosed at the same regimen [26].

Table 1.

Summary of pharmacokinetic and pharmacodynamic parameters

References Route of
elimination
Protein
binding
Bio-
availabi
lity
N Age Dosage AUC0-24a
(mg*h/L)
Cmax
(mg/L)
Half-
life
(h)
CL
(L/kg/h)b
V
(L/kg)b
PK/PD end
point
AmphotericinB
[10, 17, 23-27,
106, 167, 186]
Kidney;
possible
metabolic
pathway
unknown
NR NR 13 0–2 m
GA 24–40 wk
0.5 mg/kg IV
q24h
1.0
(0.5–
4.0)
15
(5–
82)
1
(0.46–
4.34)/1.73m2
1.5
(0.1–17.5)
Cmax:MIC


91c 3 wk – 18 y 0.25–1 mg/kg IV
q24h
NR 2.9 7–63 0.020–0.828 0.4–9.4


>90% <5% 22 Adults 0.6–1 mg/kg
IV q24h
0.5–2 24h –
15d
0.02–0.03 4.0

Liposomal Amphotericin B
[38, 187, 188] Kidney;
possible
metabolic
pathway
unknown
12 3 y (0.3–9) 10 mg/kg IV q1
week
AUCinf:
255.3d
3.0 56 0.059 4.2 Cmax:MIC

>90% NR 39 7 y (0.2–17) 0.8–5.9 mg/kg IV
q24h
NR 11.4–
44.2e
7f 0.020 0.2

44 Adults 43 y 7.5–15 mg/kg IV
q24h
692–554 76–105 6–
10.5
0.02 0.2–0.3

Amphotericin B Lipid Complex
[39, 143, 183] Kidney;
possible
metabolic
pathway
unknown
28 27 d (8–89)
GA 24–41 wk
2.5–5 mg/kg IV
q24h
NR NR 395 0.420 11.1 Cmax:MIC

NR NR 6 10 y (4–17) 2.5 mg/kg IV
q24h
11.9 2.1 NR 0.218 NR

48 Adults 0.6–5 mg/kg IV
q24h
3.29–
15.26
0.9–2.7 8–187 0.114–0.408 20.7–131

Fluconazole
[60, 64-66, 85,
122]
Kidney 63 1–88 d
GA 23–40 wk
3–12 mg/kg IV
q24h
347–496 NR 26–80 0.008–0.022 0.9–1.5 AUC0-24:
MIC

NR NR 88 2–16 y 2–8 mg/kg IV
q24h
AUCinf
73.9–
230.9c
NR 21–22 0.022–
0.037g
0.7–0.95

12% 90% 400 Adults 100–400 mg PO
q24h
76.8h 4.1–8.1 30 0.01 0.7

Itraconazole
[74, 76, 78, 79,
83]
CYP3A4
(main)
25 0.5–5 y 2.5 mg/kg PO
q12h
5 mg/kg PO q24h
6.9–16.1 0.5–1.0 31–47 0.714i NR AUC:MIC
Cmax:MIC
Cmin:MIC
NR NR
44 5–18 y 2.5 mg/kg PO
q12h
5 mg/kg PO q24h
7.1–23.0 0.6–1.5 28–
104
0.601–0.073 5.1–15.5

99.8% 55% 32 Adults 100 mg PO q24h
200 mg PO q12h
5.3–39.3
AUC0-12
22.6
0.4–2.3 34–64 0.326i 11.4i

Voriconazole
[62, 97, 98,
189-191]
CYP2C19,
CYP2C9,
CYP3A4
152 2–<12y 3–8 mg/kg IV
q12h
4.7–162 1.48–
19.2
3.1–
29.2
0.142–0.348 1.8–1.9 AUC0–
24:MIC
44–91%
70 2–<12y 4–18 mg/kg PO
q12h
1.70–
203.0
0.51–
18.0
NR NR NR

NR 26 12–17 y LD: 6 mg/kg IV
q12h
MD: 4 mg/kg IV
q12h
AUC0-12
27.9
(6.24–
95.3)
3.72
(1.71–
9.99)
NR 0.143g 1.1j
NR
22 12–17 y 300 mg PO q12h AUC0-12
18.7
(1.17–
49.7)
2.84
(0.18–
5.88)
NR NR NR

58% 96% 33 Adults LD: 6 mg/kg IV
q12h
MD: 4 mg/kg IV
q12h
37.6 4.6 NR 0.106 g 0.8j


34 200 mg PO q12h 12.8 2.45 NR NR

Posaconazole
[110, 112, 113,
115, 168]
Hepatic
glucuro-
nidation
NR 38 0.7–15.4 y 4–12 mg/kg PO
q12h
800 mg/day PO in
div doses
NR NR NR NR NR AUC:MIC
T>MIC

>98% 8–47% 36 Adults 50–400 mg
POq12h
8.3–73.1 0.4–4.2 20–31 0.147–
0.199k
4.9–6.9k

Caspofungin
[87, 92, 148,
153, 192]
Hydrolysis,
N-
acetylation
18 0–3 m 25 mg/m2 IV q24h NR 10.9–
11.1
NR NR

NR NR 10 3–24 m LD: 70 mg/m2 IV
MD: 50 mg/m2 IV
q24h
131.2 17.6 8.8kl 0.367m NR

35 2–11 y 50 mg/m2 IV q24h 146.0 2.41 NR NR NR

33 12–17y 50 mg/m2 IV q24h 117 12.9 11.2 l 0.344m NR

97% Poor >120 Adults 50–70 mg/m2 IV
q24h
86.9–
129.6
8.7–
14.0
10 0.009 0.1

Micafungin
[119, 123, 125,
126, 138, 140]
Arylsulfatas
e
catechol-O-
methyltrans–
ferase
43 2–119d
GA 24–40 wk
0.75–15 mg/kg
q24h
19–643.2 2.5–
48.1
6.9–
20.1
0.015 0.147 AUC0-24:
MIC
NR NR
73 2–17 y 0.5–4 mg/kg q24h 27.9–
301.9
6.4–
43.5
12.2–
13.5
0.014–0.243 0.26–0.42

>99% Poor 62 Adults 12.5–200 mg 11.9–
210.6
1.1–
22.6
12–20 0.014–0.021 0.23–0.33

Anidulafungin
[52, 105, 163,

164]
Chemical
degradation
8 2-28 d
GA 26-39 wk
LD: 3 mg/kg
MD: 1.5 mg/kg q24h
30.4–
108.9
2.2–6.7 NR 0.013–0.049 0.5–4.4 AUC:MIC
Cmax:MIC

NR NR 7 50–451d
GA 24–40 wk
LD: 3 mg/kg
MD: 1.5 mg/kg
q24h
30.3–
278.0
2.2–
14.9
NR 0.005–0.049 0.2–4.4

12 2–16 y LD: 1.5–3 mg/kg
MD: 0.75–1.5
mg/kg q24h
16.5–
155.7
0.91–
12.3
13.9–
38.9
0.0094–
0.0446
0.319–
0.962

13 13–16y LD: 1.5–3 mg/kg
MD: 0.75–1.5
mg/kg q24h
31.8–
134.1
3.1–
8.66
12.0–
38.9
0.0095–
0.0311
0.163–
0.803

>99% Poor 225 Adults LD: 100–200 mg
MD: 50–
100mg/kg q24h
42.3–
111.8
3.6–8.6 26–52 0.013 0.474

Flucytosine
[23, 120, 178] Kidney NR NR 13 2–55 d 25-100
mg/kg q24h
NR 27.7 7.4 2.052 L/h/1.73 m2 1.1 AUC:MIC
Ttau>MIC

2.9–4% 78–89% >64 Adults 25 mg/kg PO
q4h–
100 mg/kg q24h
576 70–80 2–5 0.096 1.4

AUC=area under the concentration-time curve; AUC0-24=area under the concentration-time curve from 0 to 24h; AUCinf=area under the concentration-time curve from 0 to infinity; AUC:MIC=AUC over minimal inhibitory concentration ratio; Cmax=maximal concentration; Cmin=minimal concentration; CL=clearance rate; GA=gestational age; IV=intravenous; LD=loading dose; MD=maintenance dose; MIC=minimal inhibitory concentration; N=number of subjects, can be from multiple studies; NR=not reported; PK/PD=pharmacokinetics/pharmacodynamics; PO=oral; q=every; Ttau= time of dosing interval; V=volume of distribution.

a

AUC0-24h unless specified otherwise.

b

Adult parameters are weight-normalized dose for a standard adult’s weight of 70 kg.

c

One neonate (17 days old) is excluded with CL of 0.009 L/h/kg; V of 9.4 L/kg and t1/2 of 693 h.

d

AUC 0 to infinity.

e

Predicted Cmax based on a population pharmacokinetic model in 11 children.

f

Half-life estimated based on the following equation: half-life=0.693*V/CL.

g

Clearance estimated based on the following equation: CL=dose/AUC.

h

AUC 0-24h after fluconazole oral dosing of 100 mg at steady state.

i

Clearance and volume after IV administration.

j

Volume estimated with the following equation: V=dose/Cmax after 7 days of treatment with 4 mg/kg IV q12h.

k

Apparent clearance or volume of distribution.

l

β-phase half-life.

m

Clearance in l/h/m2.

Similar to adults, AmB use in children is associated with infusion-related toxicities (fever, chills rigors), nephrotoxicity, and electrolyte disturbances [25]. AmB is often believed to be better tolerated in children than in adults due to decreased number of co-morbidities and comedication. However, in infants, renal toxicity rates vary across studies from 0 to 44% whereas a rate of 53% was described in adults with invasive aspergillosis [28-31]. On the other hand, up to 50% of adults experience fever during infusions whereas this toxicity is rarely described in infants [28, 30, 32].

1.2. Lipid-based Amphotericin B Preparations

Lipid-associated formulations have the same mechanism of action and antifungal spectrum as AmB, but higher dosages are required for equivalent antifungal efficacy in vitro and in animal models [33-35]. In a murine candidiasis model, AmB was 5–8-fold more potent than L-Amb and ABLC in the kidneys [33]. Nonetheless, in adults with hematologic malignancies, no difference in therapeutic efficacy has been demonstrated between the 2 formulations [36, 37]. In a cohort of children (median age 7 years) with proven IFD caused by C. albicans, Aspergillus sp., or Scedosporium sp., Cmax:MIC was statistically higher in children with a complete response compared with children with partial response (median Cmax:MIC of 68 vs. 40, p=0.02) [38]. Other than in this small cohort, no further correlation between PK and PD has been established in children.

Compared with AmB, the newer lipid-based formulations have different pharmacological properties. ABLC at a dosage of 2.5 mg/kg/day showed a lower Cmax (2.1 mg/L) and faster CL (0.218 L/kg/h) than conventional AmB in children with invasive candidiasis (Cmax of 2.9 mg/L and CL of 0.040 L/kg/h after dosing of 1–1.5 mg/kg/day of AmB) [24, 39]. ABLC was also investigated in neonates with invasive candidiasis (age 8–89 days; gestational age [GA] of 24–41 weeks), showing a clearance rate at the upper range of adults values (0.4 L/kg/h) [40, 41]. This neonatal population analysis led to dosing recommendations of 2.5–5 mg/kg/day (Table 2). In contrast to ABLC, a population PK study of L-Amb in 39 children (age 0.2–17 years) revealed a higher Cmax (11.4–44.2 mg/L) compared with similar AmB (Table 1) [38]. In this study, LAmB also had a lower V and CL, presumably due to a slower tissue distribution [38]. PK estimates for each of these lipid-based formulations were nonetheless comparable to adult values. No published data were found on ABCD PK in children.

Table 2.

Dosing recommendations of antifungal agents in children

Drug Dosing Therapeutic drug
monitoring
References
Neonates
(0–30 days)
Infants
(31 days–2 years)
Children
(2 years–17 years)
Polyenes
Amphotericin B 0.6−1 mg/kg IV q24h 0.6−1 mg/kg IV q24h 0.6−1 mg/kg IV q24h No [23-27, 167]
ABLC 2.5−5 mg/kg q24h Unknown 5 mg/kg IV q24h No [39, 50, 51, 183]
ABCD unknown Unknown Unknown No
L-AmB 3-5 mg/kg q24h 3–5 mg/kg IV q24h 3–6 mg/kg IV q24h No [47-49, 193]
Triazoles
Fluconazole Loading dose 25 mg/kg
12 mg/kg q24h
(3–6 mg/kg twice weekly
for prophylaxis)
12 mg/kg IV q24h
(3 mg/kg IV q24h for
prophylaxis)
6-12 mg/kg IV or PO q24h
(3 mg/kg q24h for
prophylaxis)
No [64-66]
Itraconazole unknown Unknown 2.5–5 mg/kg PO q12h
(2.5 mg/kg PO q24h for
prophylaxis)
Trough >0.5 mg/Lc [57, 74-76, 78]
Voriconazole unknown Unknown 7–9 mg/kg IV q12ha
200 mg PO q12h
(2.5 mg/kg PO q12h for
prophylaxis)
Trough 1.0-5.5 mg/L [62, 91, 99, 190]
Posaconazole unknown Unknown 200–400 mg PO q6–12 hb Prophylaxis : trough>0.5 mg/Lc
Treatment : trough >1 mg/Lc
[168]
Ravuconazole
Albaconazole
Isavuconazole
unknown Unknown unknown unknown
Echinocandins [148-150, 153,
155]
Caspofungin 25 mg/m2 IV q24h 50 mg/m2 IV q24h Load with 70 mg/m2 IV once,
then 50 mg/m2 IV q24h
No
Micafungin 10 mg/kg IV q24h 3–4 mg/kg IV q24h 2–4 mg/kg IV q24h
(1 mg/kg IV q24h for
prophylaxis)
No [2, 125, 135, 138,
141]
Anidulafungin Load with 3 mg/kg IV
once,
then 1.5 mg/kg IV q24h
Load with 3 mg/kg IV
once,
then 1.5 mg/kg IV
q24h
Load with 3 mg/kg IV once,
then 1.5 mg/kg IV q24h
No [105, 163, 164]
Aminocandin unknown unknown unknown unknown
Nucleoside analogs
5-Flucytosine unknown unknown unknown Cmax : 20−50 mg/Lc [120, 181]

ABCD=amphotericin B colloidal dispersion; ABLC=amphotericin B lipid complex; AUC=area under the concentration-time curve; AUC:MIC=AUC over minimal inhibitory concentration ratio; Cmax= maximal concentration; div=divided dose; EMA=European Medicines Agency; FDA=Food and Drug Administration; IV=intravenous; L-AmB=liposomal amphotericin B; MIC=minimal inhibitory concentration; PD=pharmacodynamics; PO=oral; NR=not reported; q=every; T>MIC=time concentrations are over the MIC.

a

Dosing for <12 years old. For children ≥ 12 years old, adult dosing is recommended.

b

Dosing unknown for children <12 years of age.

c

Target concentrations in adults.

There have been concerns that amphotericin lipid formulations penetrate kidneys and CNS at a lesser extent than AmB [42]. For infants, this characteristic is of special interest because Candida commonly disseminates in those two compartments. In animals, Candida kidney infection models showed decreased penetration and potency of lipid formulations compared to AmB [33, 42]. Despite these findings, neonates treated with ABLC demonstrated urine amphotericin concentrations higher than the MIC of many Candida isolates [41]. In the CNS, studies in animal models demonstrated that AmB and L-AmB had higher penetration than ABLC and ABCD, consistent with clinical findings in which neonates treated with ABLC had low or undetectable amphotericin concentrations in CSF [41, 43]. The clinical significance of these findings is not well established. The largest comparative effectiveness study between AmB and lipid-formulation in infants consisted of a cohort study (730 infants, <120 days old) in which there was increased mortality and therapeutic failure with lipid-formulations compared with AmB [44]. These findings are consistent with lower lipid products penetration to the kidneys and CNS in animal models, but this observational study failed to adjust for renal function and other clinical data and these results could also be due to confounding by indication.

In terms of safety, the lipid-based formulations offer the advantage of reduced toxicity compared with AmB, with the exception of ABCD for which the frequency of infusion-related symptoms in adults is similar [45]. In infants, uncontrolled studies demonstrated favorable safety for L-Amb with mild increases in liver enzymes (0–37%) and serum creatinine (0–5%), and decrease in potassium (0–5%) [46-49]. No serious AEs were reported in those studies For ABLC, 2 large safety studies were conducted in children [50, 51]. The first study involved 111 children (21 days –16 years) who received ABLC at a dosing of 5 mg/kg/day and demonstrated no drug-related adverse events [50]. The second involved 548 children (0–20 years) who received 5 mg/kg/day of ABLC and showed an increase of serum creatinine of 2.5x baseline in 8.8% of children [51].

Given extensive clinical experience with AmB and broad-spectrum activity, amphotericin B compounds are widely used for IFD therapy in pediatric populations. Lipid-based formulations PK data are limited in infants, and there are concerns of insufficient penetration in the urinary tract and CNS in this population [44]. As a result, AmB is generally the first line polyene agent in young infants unless urine infection and meningoencephalitis are excluded. Further comparative data on efficacy and safety of AmB vs lipid-based formulations are needed in infants. Appropriate plasma concentrations have not been defined for safety and efficacy, and therapeutic drug monitoring (TDM) is therefore not recommended.

2. Triazoles

Triazole agents inhibit the cytochrome P450 (CYP) that synthesizes ergosterol, which is a major cell membrane component of most fungi [52]. This mechanism of action inhibits cell growth and replication. In humans, because triazole agents are inhibitors of CYP enzymes (particularly CYP3A4), this class is prone to significant drug-drug interactions. Despite this characteristic, clinical efficacy of triazoles has been demonstrated for the prophylaxis and treatment of many IFD.

2.1. First-generation Triazoles

Fluconazole and itraconazole constitute the first generation in this class. They are active against Candida sp., Cryptococcus neoformans, and dimorphic fungi. Exceptions include C. glabrata and krusei, which are often resistant to fluconazole (MIC90 of 32 mg/L and 64 mg/L, respectively) [53]. Only itraconazole provides coverage of Aspergillus sp. Both agents are available as oral and parenteral formulations.

2.1.1. Fluconazole

Fluconazole exhibits in vitro fungistatic activity [54], which does not correlate well with concentration and is more dependent on duration of exposure (time-dependent). In vivo infection models demonstrated that a ratio of area under the concentration-time curve (AUC) over MIC (AUC:MIC) of 18 was associated with 80% survival in the murine candidiasis models [55, 56]. In adults with Candida infection, a ratio (AUC: MIC) <11.5, and MIC≤64 mg/L were associated with increased mortality or therapeutic failure [57, 58]. For a Candida sp. with an MIC of 32 mg/L, AUC:MIC >12 corresponds to an AUC0-24 of 400 mg*h/L, which is often cited as the exposure target for adults. In immunocompromised patients and premature infants, an AUC0-24 of 800 mg*h/L is typically targeted, probably to cover a broader range of Candida spp. MICs. In children, there is no established relationship between PK and PD, and exposure target is usually extrapolated from adults (AUC0-24 from 400–800 mg*h/L).

Fluconazole has high oral bioavailability (92%) [59]. Consistent with its low protein-binding, fluconazole shows good penetration into tissues and body fluids, especially in the urine, and concentrations in CSF achieve 80% of blood levels [60, 61]. Fluconazole does not undergo significant metabolism and is predominantly excreted unchanged in urine. After filtration, fluconazole is highly reabsorbed by the kidney [62]. Dosing should be reduced in subjects with renal impairment; however, continuous renal replacement therapy might result in lower concentrations than expected because of the lack of renal reabsorption in this setting [63].

In neonates, a population PK model developed with a cohort of 55 infants <120 days of age (23–40-week-gestation) revealed the importance of gestational age and postnatal age in fluconazole disposition [64]. Based on this PK model, simulations predicted that a daily dose of 12 mg/kg in infants <30 weeks of gestation achieved a median 24h-AUC close to the target of 800 mg*h/liter, whereas infants >30 weeks of gestation achieved a lower median 24h-AUC of 400 mg*h/L [64]. In contrast, dosing of 6 mg/kg daily in adults is sufficient to achieve a 24h-AUC of 400 mg*h/L in more than 85% of the subjects [57]. In the population PK model involving infants described above, fluconazole achieved steady state therapeutic AUC in 5-7 days [64]. As a result, administration of a loading dose has been suggested in vulnerable populations such as infants [7]. In a small cohort of 10 infants < 60 days old, a fluconazole loading dose of 25 mg/kg was safe and achieved the therapeutic target more rapidly than traditional dosing which may lead to decreased morbidity [65].

Beyond the neonatal period, the PK in children is characterized by a higher CL (0.030 L/kg/h) and a larger V (0.95 L/kg in children aged 2–12 years) compared with adults (CL of 0.016 L/kg/h and V of 0.7 L/kg) as demonstrated in a study involving 100 children (ages 0–18 years) (Table 1) [60, 66]. Consequently, children require a proportionately higher dose to match adult exposures. Finally, although limited, assessment of oral absorption after 2 years of age suggested similar bioavailability as adults (>80%) [66].

Fluconazole is a potent inhibitor of CYP2C9 and CYP3A4 leading to increased plasma concentration of other drugs metabolized by those metabolic enzymes when co-administered with fluconazole. Such compounds include tacrolimus or cyclosporine, which are commonly used in the pediatric population at high risk for invasive fungal infection [67]. Beside safety concerns related to drug-drug interactions, fluconazole is well tolerated in children as evidenced by a safety analysis in 562 children (0–17 years of age) following oral and intravenous administration [68]. The most common treatment-related adverse events (AEs) were gastrointestinal symptoms (7.7%) and skin rash (1.2%). In this study, overall, 18 of 562 children (3.2%) discontinued fluconazole due to AEs [68].

Fluconazole is widely used for the prevention and treatment of invasive candidiasis in children and infants. It is also used for treatment and secondary prophylaxis of cryptococcal meningitis. PK and safety are well defined across a wide age range. Moreover, its reliable penetration into CSF and the urinary tract makes fluconazole a useful agent against neonatal invasive candidiasis [69, 70]. Children generally require higher dosing per unit of weight to match adult exposure. However, target exposure is extrapolated from adults, and age-specific PD indices have not been established.

2.1.2. Itraconazole

Similar to fluconazole, itraconazole is fungistatic against yeast-like fungi [71], while only itraconazole has fungicidal activity against molds [72]. Given the time-dependent PD demonstrated for both yeasts and Aspergillus sp. [71, 73], separating the daily dose in 2 appears more appropriate (Table 2) [74]. PD modeling revealed that AUC:MIC, Cmax:MIC, and minimum plasma concentration (Cmin):MIC equally correlated with antifungal efficacy for the treatment of oropharyngeal candidiasis in human immunodeficiency virus (HIV)-infected children [74].

Itraconazole circulates in blood highly protein-bound and undergoes extensive hepatic metabolism into several metabolites. One of those metabolites, hydroxyl-itraconazole, has similar activity to the parent drug. Itraconazole is excreted through the liver and kidneys. Oral absorption is variable, but acidic gastric environment, food, and administration in oral solutions enhance its absorption [75]. Dosing of 5 mg/kg/day with oral solution results in lower exposure in children compared with adults (mean AUC0-24 of 8.7 mg.h/L vs. 22.7 mg.h/L, and mean Cmax of 0.6 mg/L vs. 1.5 mg/L) [76, 77]. These differences were not found with lower dosing (2.5 mg/kg/dose twice a day), which produced similar exposure in adults and children [78]. These findings highlight the non-linearity of itraconazole’s PK profile and the possible dose-dependent bioavailability. PK parameters in children >5 years of age are otherwise relatively similar to adults [74, 76, 79].

Itraconazole is well tolerated in children, and the most commonly reported AEs are gastrointestinal symptoms (8–12%) [74, 80]. In adults, trough concentrations above 17 mg/L (measured by bioassay) are significantly associated with toxicity (mainly fluid retention and gastrointestinal symptoms) [81]. The main safety concern for itraconazole is the potential for drug-drug interactions due to inhibition of CYP3A enzymes [82]. Co-medication with itraconazole may result in increased plasma concentration of drugs metabolized by CYP3A4 enzymes, such as cisapride or oral midazolam [83]. Enhanced vincristine neurotoxicity is a well-documented drug interaction with itraconazole in both adults and children [84, 85]. In a retrospective study of 20 children with acute lymphoblastic leukemia, those receiving vincristine in combination with azole treatment (predominantly itraconazole) experienced significantly more peripheral neurotoxicity (p<0.05) [85].

Itraconazole use is limited in children due to erratic, dose-dependent oral bioavailability, high PK variability, and the availability of more reliable alternatives [86]. Given an established target trough concentrations in adults, TDM is standard practice (target trough concentrations >0.5 mg/L when measured by HPLC [57] and <17 mg/L when measured by bioassay [81]). No age-specific target concentrations have been identified in children.

2.2. Second-generation Triazoles

Second generation triazoles are active against a wide spectrum of clinically important fungi including yeast, molds, and dimorphic fungi. Members of this class of triazoles include voriconazole, posaconazole, and newer compounds such as ravuconazole.

2.2.1. Voriconazole

Voriconazole is available in oral and intravenous formulations and is the primary therapy for invasive aspergillosis. It is structurally similar to fluconazole but has extended antifungal spectrum against Aspergillus sp. Despite its broad-spectrum activity against yeast and molds, voriconazole is not active against zygomycetes [12, 87]. In vitro, fungicidal and fungistatic activity against Aspergillus sp. and Candida sp., respectively, are time-dependent [88, 89]. Near maximal effect against Candida sp. was observed at concentrations 3 times the MIC at different time points [89]. In murine candidiasis models, AUC: MIC was strongly predictive of treatment success with a suggested target of free concentration AUC0-24:MIC of 20 [90]. Consistent with voriconazole time-dependent effect, Cmin >1-2 mg/L was a good predictor of successful clinical outcome in both adults and children [91, 92]. In children, Cmin <1 mg/L was associated with increased odds of death (odds ratio [OR] 2.6; 95% confidence interval [CI] 1.6, 4.8) [91].

Voriconazole is 58% protein-bound, distributes well into tissues and CSF [93, 94], and is extensively metabolized by hepatic CYP2C19. Allelic variations contribute to high inter-subject PK variability in adults and children [95, 96]. In contrast to adults, children have a linear PK at dosing of 3–4 mg/kg intravenously (IV) [97]. Elimination becomes nonlinear over the range of 4 mg/kg to 8 mg/kg every 12 hours [98]. Children <12 years of age have greater clearance of voriconazole and require almost twice the dose to match adult systemic exposure (7–9 vs. 4 mg/kg IV twice a day) [95, 98, 99]. Children also have a lower oral bioavailability than adults (45–65% vs. 96%) [95, 98]. This difference is not completely understood but may relate to greater first-pass metabolism in children.

Voriconazole side effects include visual disturbances; elevated hepatic transaminases, and skin photosensitization (13–30%) [97, 98, 100, 101]. In adults, trough concentrations above 4–5.5 mg/L correlate with toxicity [6, 102, 103]. In a small pediatric cohort, oral administration of more than 6 mg/kg/dose twice a day was associated with an increased risk of phototoxic skin reactions, but no correlation was described with trough concentrations [101]. For other forms of toxicity, no relationship to exposure has been determined in children [104].

Multiple pediatric PK studies recently helped determine optimal dosing in children down to 1 year of age, but voriconazole currently has European Medicines Agency (EMA) and Food and Drug Administration (FDA) labeling for children over 2 and 12 years of age, respectively [105, 106]. In infants, PK and safety still need to be characterized, and voriconazole use is discouraged in this population. Efficacy has been well established in adults with aspergillosis, and it is the primary recommended therapy for invasive aspergillosis [107]. TDM is recommended, given a high inter-subject variability, and has proven useful in adults for which a randomized controlled trial showed better outcome and reduced toxicity with target Cmin between 1 and 5.5 mg/L [108]. In children, Cmin≥1 mg/L was also retrospectively associated with decreased mortality from IFD, but as opposed to adults, no upper bound of target trough concentration has been identified, and this question warrants further study [91].

2.2.2. Posaconazole

Posaconazole has broad antifungal activity against the majority of yeasts and azole-resistant Candida sp. In addition to Aspergillus sp., posaconazole is also active against other molds, including zygomycetes, as opposed to voriconazole. Similar to other triazoles, posaconazole is fungicidal in vitro with time-dependent killing against most Candida species and molds [63, 75]. In animal models, the duration of exposure to plasma concentrations above the MIC is the most important parameter for optimum efficacy [109].

Posaconazole is only available as an oral formulation and should be taken with high-fat meals to enhance absorption [110]. Protein-binding is high, and the primary route of elimination is through feces with renal clearance playing a minor role [111]. Posaconazole prophylaxis and treatment have been described in children, but the PK is not well characterized. No specific dose recommendations exist, and its use in children is off-label [112-115]. A twice-daily dosing algorithm based on allometric scaling (body weight-based) delivered adequate exposure in 12 children with chronic granulomatous disease (mean trough concentration of 1.54 mg/L) [115]. In adults, the PK is linear, and the long half-life (25 hours) produces stable plasma concentrations over time [110]. Steady-state concentrations are achieved 7–10 days after initiating therapy, suggesting that this agent might be suboptimal for induction therapy or a loading dose may be required. Posaconazole is well tolerated, and the most common side effect is gastrointestinal symptoms in 25% of patients [113, 114].

Exposure-response relationship analyses in adults suggest the need for TDM with a target trough concentration of 0.5 mg/L to 1 mg/L for prophylaxis and therapy, respectively [116, 117]. There is no correlation between exposure and toxicity [116].

Posaconazole PK/PD in children is scarce and needs better characterization. It is not licensed for use in children under 12 years of age by regulatory agencies [17, 35]. An ongoing trial will address posaconazole dosing in children (NCT01716234). Availability of an intravenous formulation will attract interest from the pediatric community; however, PK studies will need to be conducted to evaluate optimal dosing in children.

2.2.3. Other Triazoles

Ravuconazole, isavuconazole, and albaconazole are the newest antifungal triazoles with in vitro and in vivo fungicidal activity against a wide spectrum of clinically important fungi. They are active against Candida sp. including fluconazole-resistant strains, Cryptococcus sp., and Aspergillus sp. [90, 118-121]. Activity against Fusarium sp., Scedosporium sp., and Mucor sp. remains limited [53, 121, 122]. Similar to other triazoles, animal models show that antifungal efficacy is time-dependent [90, 123]. All 3 agents are available orally and have high oral bioavailability, but only ravuconazole and isavuconazole are available intravenously [124, 125]. Clinical experience is still limited in adults, but PK studies reveal a prolonged half-life from 75– 117 hours [124, 126, 127]. These new triazoles are well tolerated, and the most frequently reported AEs are headaches, rhinitis, and gastrointestinal symptoms [124, 126, 127]. No clinical trials have been completed in children, and so these agents are not currently recommended.

3. Echinocandins

Agents from the echinocandins class are only available in parenteral form and include micafungin, caspofungin, anidulafungin and the more recent aminocandin. They act by inhibiting (1,3)-β-D-glucan synthase, a fungus-specific enzyme crucial to the biosynthesis of glucan in the fungal cell wall [128]. They exhibit fungicidal activity against most Candida spp, including fluconazole-resistant species (C. albicans, C. parapsilosis, C. glabrata, C. tropicalis, and C. krusei) and fungistatic activity against Aspergillus sp. [129, 130]. Echinocandins are not active against Cryptococcus neoformans, zygomycetes, and dimorphic fungi. Advantages of echinocandins include low toxicity and minimal drug interactions due to the fungus-specific mechanism of action. Disadvantages include the lack of oral formulation and the reduced activity against non-Candida species.

3.1. Micafungin

Micafungin displays concentration-dependent fungicidal killing, and in animal models, efficacy correlates best with AUC:MIC ratios [131]. Analysis of adult clinical data for the treatment of invasive candidiasis found that an AUC:MIC ratio over 3,000 predicts better mycological response [127]. In neonates who are at greater risk of meningoencephalitis caused by Candida sp., a different exposure target (AUC0-24 =166.5 mg*/L) has been proposed based on a rabbit model of neonatal hematogenous Candida meningoencephalitis [132].

Micafungin is highly bound to protein and distributes well into tissues including lung, liver, and spleen. Although penetration into the CNS and eye are generally limited in adults, high-dose micafungin successfully treated neonatal animal models of hematogenous Candida meningoencephalitis [132, 133]. Micafungin undergoes only limited phase 1 metabolism, and the parent drug is mostly excreted via the biliary system [134].

Micafungin PK is well described in pediatric patients aged 4 months to 16 years, and 229 patients were included in population PK analyses in support of recent U.S. pediatric labeling [135]. The half-life is approximately 12 hours, and the PK is linear at doses of 1–3 mg/kg [136]. Several PK studies show an age effect demonstrated by an inverse relationship between weight and CL, such that as body weight decreases, higher dosages of micafungin (on a mg/kg basis) are required to achieve equivalent drug exposure [137]. In a study of 77 neutropenic children, doses of 3–4.5 mg/kg/day for 2–8-year-olds and 2–3 mg/kg/day for 9–17-year-olds achieved adult exposure [138].

Four PK studies demonstrated that younger infants (GA of 24–40 weeks and postnatal age of 2–119 days) have higher clearances and therefore need higher dosing than children and adults [123, 126, 139, 140]. Moreover, this population requires reliable micafungin CNS penetration because of a higher risk of Candida meningoencephalitis. Based on a population PK model, PK simulation analyses showed that 83% of neonates receiving 10 mg/kg/day would achieve an AUC0-24h of 166.5 mg*/L, a specific target for neonatal Candida meningoencephalitis [125, 132]. These preliminary PK/PD analyses led to the dose selection for an ongoing phase 3, randomized controlled trial comparing micafungin to amphotericin B deoxycholate (NCT00815516).

Pooled data from 6 pediatric studies show that micafungin has a favorable safety profile [141]. The most common treatment-related AEs are transient and include liver enzyme elevations and decreased potassium (3%) [141]. In a trial comparing micafungin and L-Amb in 106 children with invasive candidiasis, both drugs had equivalent efficacy, but micafungin was better tolerated [142]. No maximum tolerated dose has been identified, and high doses of 15 mg/kg have been safely used in premature neonates [140].

Overall, micafungin dosing is well-defined in children, and regulatory agencies have labeled its use in the pediatric population [143, 144]. The EMA product license includes dosing of 2 mg/kg/day for all children <16 years of age, including neonates. On the other hand, the U.S. FDA product label includes dosing of 2 and 3 mg/kg/day in children <30 kg for treatment of invasive candidiasis and esophageal candidiasis, respectively. Unlike EMA, FDA label restricts its use to children ≥ 4months of age. For infants ≤ 3 months, PK studies and age-specific exposure targets suggest that dosing up to 10 mg/kg/day are required. Clinical efficacy and safety data with newly proposed dosing in this population needs further evaluation.

3.2. Caspofungin

Caspofungin is recommended for therapy of invasive candidiasis, salvage therapy of invasive aspergillosis, and as empirical therapy in febrile neutropenia in children >3 months of age. It demonstrates in vitro fungicidal activity against Candida sp. with a concentration-dependent effect and prolonged PAFE (12 hours) [15, 75, 145]. In vitro PD properties suggest optimal killing when concentration is 4x the MIC [145, 146]. Murine candidiasis models demonstrate reduced activity against C. parapsilosis and C. guillermondii. The clinical significance of this reduced activity is unclear, but clinicians should be aware of the possibility of reduced efficacy. Caspofungin is generally classified as fungistatic against Aspergillus sp, however, evidence of killing in vitro was shown by change in cells staining pattern but this phenomenon did not translate into reduction in the number of CFU from standard killing curve measurements [147]. In a PK trial of 32 children aged 3 months to 17 years with confirmed or suspected IFD (Candida sp. and Aspergillus sp.), the exposure-response analyses revealed no association with clinical outcome and PK/PD parameters [148].

Caspofungin is metabolized by the liver with a half-life of 7–10 hours and demonstrates linear PK in children.[149, 150] Similar to other agents in this class, caspofungin urine concentrations are low [151]. However, this poor renal penetration has not translated into treatment failure of candiduria in adults [152]. Specific to caspofungin, weight-based dosing does not provide consistent steady-state concentrations across ages, whereas dosing based on body surface area does [150]. In infants and children with neutropenia, 50 mg/m2/day dosing produces systemic exposures similar to adults receiving 50 mg/day [149, 150]. In young infants, although the PK has not been fully characterized, a study involving 18 infants <3 months old with suspected invasive candidiasis showed that 25 mg/m2/day was required to achieve adult plasma levels [153].

Dose-limiting toxicities have not been seen in adults [154]. Multiple clinical trials in children found that caspofungin is well tolerated; fever, rash, hypokalemia, and elevated liver enzymes are the most frequently reported AEs but do not usually require therapy discontinuation [148, 150, 153, 155, 156]. The incidence of AEs does not correlate with concentration [148]. In young infants, safety data are limited to 2 small cohorts (total N=22) in which AEs were described without a comparator group [157, 158].

Caspofungin is recommended for pediatric IFD and is particularly useful against invasive candidiasis caused by azole-resistant isolates. Its PK has been adequately characterized in children down to 3 months of age. However, PK, safety, and efficacy are inadequately defined in young infants [157, 158].

3.3. Anidulafungin

The spectrum of anidulafungin activity is wide with fungicidal effect against Candida sp. including fluconazole-resistant strains, and fungistatic effect against Aspergillus sp. [118, 159-161]. Fungicidal activity was demonstrated in neutropenic murine models for which both Cmax:MIC and AUC:MIC ratios strongly predict successful treatment of systemic candidiasis [162]. In Aspergillus animal models, anidulafungin therapy reduces Aspergillus antigenemia but is unable to clear the infection, consistant with its fungistatic activity [118].

Anidulafungin is highly protein-bound (>99%) but achieves tissue concentrations above MIC in animal models [51]. Anidulafungin demonstrates linear PK and a longer half-life than other echinocandins (20 hours) [163]. It has a unique elimination pathway among the echinocandins, consisting of slow, non-enzymatic degradation to inactive metabolites. When anidulafungin is given at the same weight-adjusted dose (1.5–3 mg/kg loading dose, 0.75–1.5 mg/kg/day maintenance dose), children 2–17 years old with neutropenia achieve exposures similar to adults [163]. In infants and neonates, 1.5 mg/kg/day produces plasma concentrations comparable to adults [164]. Both prior PK trials demonstrate that anidulafungin is well tolerated with no observed drug-related serious AEs.

Despite available PK data and a favorable safety profile demonstrated in 2 pediatric trials, clinical experience in children remains insufficient to recommend it for use in this population. Anidulafungin is not labeled for children under 16 years in the U.S and <18y in Europe [165, 166].

3.4. Aminocandins

Aminocandin is the newest echinocandin and is available as an intravenous formulation. Similarly to other agents of this class, it is active against Candida sp. and Aspergillus sp. in vivo [167, 168]. Fungicidal effect against Candida sp. best correlates with peak:MIC demonstrating a concentration-dependent activity [168]. Animal models suggest that extended dosing interval (7– 10 days) is effective as treatment and prophylaxis of invasive candidiasis [169]. In healthy volunteers, a phase 1 study also demonstrated a long half-life (48–58 hours) indicating that dosing could be less frequent than once a day [135]. In this small cohort, aminocandin was well tolerated, but no other clinical trials have been published in adults or children.

3.5. Second generation echinochandins

ASP9726, a novel second generation echinocandin is under development. As opposed to other echinocandins, ASP9726 showed Aspergillus hyphal growth inhibition and improved MIC against Candida parapsilosis and echinocandin resistant-Candida in vitro [170]. To our knowledge, no ASP9726 clinical trials have been registered in clinicaltrials.gov at the time of submission.

4. Nucleoside Analogs

5-Flucytosine (5-FC) is an antimetabolite drug that causes RNA miscoding and inhibits DNA synthesis [171]. 5-FC has activity against Candida sp. and Cryptococcus neoformans [172, 173]. In vitro and in vivo testing demonstrates that flucytosine is fungistatic against yeasts with concentration-independent pharmacodynamics [174, 175]. Moreover, 5-FC exhibits PAFE up to 10 hours [176]. It is never used as monotherapy given rapid emergence of resistance. Its use is limited for the induction therapy of cryptococcal meningitis for which 5-FC in combination with AmB was shown to be efficacious [177].

Only the oral formulation is available in the United States. Oral bioavailability is high in adults (75–95%), and a trial comparing IV and oral 5-FC in adults with HIV-associated cryptococcal meningitis did not detect a difference in fungicidal activity [178]. Protein-binding is negligible, and distribution into tissues and body fluids is reliable. 5-FC is mainly eliminated in active form in urine. The elimination half-life is 3–5 hours, and administration in 3–4 daily doses is required [179]. Data on 5-FC PK in children are limited, but drug clearance appears slower in children compared with adults [23, 180]; as a result, the adult dose of 100 mg/kg/day might lead to overexposure. In addition, the elimination half-life in neonates is nearly twice as long as in adults (4 hours vs. 7 hours) [23, 180], suggesting that the dosing interval should be longer (8 hours to 24 hours).

Dose-limiting toxicities described in up to 44% of adults (hematologic, gastrointestinal, and hepatic) limit the use of 5-FC [181, 182]. Safe target plasma concentrations in adults are established at 40–60 mg/L, but no such data are available for children [181].

Given the narrow therapeutic window, TDM is standard of care for 5-FC with target peak concentrations of 20–50 mg/L [183]. However, target concentrations have not been established in children. Concentration-independent PD suggests that lower dosages could be considered in future research. Clinical use of 5-FC is limited mainly to combination therapy with AmB for the treatment of cryptococcal meningitis. Because of limited PK, safety, and efficacy data, the use of 5-FC in infants is discouraged.

5. Novel Antifungals Agents under development

Two antifungals agents are under development and would each represent a new class of antifungals. T2307, a novel arylamidine showed in vitro and in vivo activities against Candida species, Cryptococcus neoformans, and Aspergillus species [184].The second agent is kakeromycin, which demonstrated good in vitro activity against the same fungal pathogens than T2307 [185]. For those 2 potential antifungal drugs, no clinical data are available thus far.

Conclusion

Effective and safe antifungal therapy depends on optimal drug dosing. With the recent development of new antifungal therapies, specific data in children are needed because prior extrapolations of adult dosing to children have often proved wrong. Thanks to regulatory initiatives and suitable trial designs, the number of PK studies in children has increased and clarified some dosing issues in this vulnerable population. Based on our review of the literature, we present a summary of antifungal dosing recommendations in Table 2. Whenever dose ranges are presented, clinicians should consider the benefits of early initiation of high dose therapy in improving outcomes in the setting of severe IFD.

Despite better evidence on antifungal therapy, morbidity and mortality related to IFD remain unacceptably high and dosing should be optimized, especially in infants. Future research should focus on determining drug disposition of newer antifungals such as newer triazoles and echinocandins in children as they become available for adults. Future research questions should also relate to optimize dosing of older agents such as fluconazole in neonates for which there is a need to better support the use of a loading dose. We also need better comparison of safety and effectiveness of AmB and lipid formulations of amphotericin B in infants. Finally, clinical validation of PK/PD indices determined in animal models are sparse in humans, even more so in children. Ongoing efforts to characterize the PK and PD of antifungal agents, especially in younger populations, will help inform dosing and improve clinical outcomes of IFD.

Key Points.

  • Antifungal pharmacokinetics in children frequently differ from adults, necessitating dosing adjustment to match adult exposure.

  • Antifungal target plasma concentrations are still largely extrapolated from adult data; therefore, pharmacokinetics/pharmacodynamics indices specific to children must be better defined.

Acknowledgment

Julie Autmizguine receives support from Training Award, Fonds Irma-Levasseur, Pediatric Department, Sainte-Justine University Hospital Center, Montreal, QC, Canada.

Jeffrey T. Guptill receives support from the American Academy of Neurology Foundation – Myasthenia Gravis Foundation of America (Clinician-Scientist Development Award) and from industry for drug development consulting (www.dcri.duke.edu/research/coi.jsp).

Michael Cohen-Wolkowiez receives support for research from the National Institutes of Health (NIH) (1K23HD064814), the National Center for Advancing Translational Sciences of the NIH (UL1TR001117), the Food and Drug Administration (1U01FD004858-01), the Biomedical Advanced Research and Development Authority (BARDA) (HHSO100201300009C), the nonprofit organization Thrasher Research Fund (www.thrasherresearch.org), and from industry for drug development in adults and children (www.dcri.duke.edu/research/coi.jsp).

Daniel K. Benjamin Jr. receives support from the United States government for his work in pediatric and neonatal clinical pharmacology (1R01HD057956-05, 1K24HD058735-05, UL1TR001117, and NICHD contract HHSN275201000003I) and the nonprofit organization Thrasher Research Fund for his work in neonatal candidiasis (www.thrasherresearch.org); he also receives research support from industry for neonatal and pediatric drug development (www.dcri.duke.edu/research/coi.jsp).

Edmund V. Capparelli receives research support from the United States government (U54 HD071600-01) and consulting fees from Trius, Cerexa Pharmaceuticals, Abbott, Cempra, and Theravance.

References

  • [1].Kao AS, Brandt ME, Pruitt WR, et al. The epidemiology of candidemia in two United States cities: results of a population-based active surveillance. Clin Infect Dis. 1999 Nov;29(5):1164–70. doi: 10.1086/313450. [DOI] [PubMed] [Google Scholar]
  • [2].Benjamin DK, Jr., Stoll BJ, Gantz MG, et al. Neonatal candidiasis: epidemiology, risk factors, and clinical judgment. Pediatrics. 2010 Oct;126(4):e865–73. doi: 10.1542/peds.2009-3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Burgos A, Zaoutis TE, Dvorak CC, et al. Pediatric invasive aspergillosis: a multicenter retrospective analysis of 139 contemporary cases. Pediatrics. 2008 May;121(5):e1286–94. doi: 10.1542/peds.2007-2117. [DOI] [PubMed] [Google Scholar]
  • [4].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 Jan;117(1):84–92. doi: 10.1542/peds.2004-2292. [DOI] [PubMed] [Google Scholar]
  • [5].Fernandez M, Moylett EH, Noyola DE, Baker CJ. Candidal meningitis in neonates: a 10-year review. Clin Infect Dis. 2000 Aug;31(2):458–63. doi: 10.1086/313973. [DOI] [PubMed] [Google Scholar]
  • [6].Friedman S, Richardson SE, Jacobs SE, O’Brien K. Systemic Candida infection in extremely low birth weight infants: short term morbidity and long term neurodevelopmental outcome. Pediatr Infect Dis J. 2000 Jun;19(6):499–504. doi: 10.1097/00006454-200006000-00002. [DOI] [PubMed] [Google Scholar]
  • [7].Pappas PG, Kauffman CA, Andes D, et al. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis. 2009 Mar 1;48(5):503–35. doi: 10.1086/596757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Brajtburg J, Powderly WG, Kobayashi GS, Medoff G. Amphotericin B: current understanding of mechanisms of action. Antimicrob Agents Chemother. 1990 Feb;34(2):183–8. doi: 10.1128/aac.34.2.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Sokol-Anderson M, Sligh JE, Jr., Elberg S, Brajtburg J, Kobayashi GS, Medoff G. Role of cell defense against oxidative damage in the resistance of Candida albicans to the killing effect of amphotericin B. Antimicrob Agents Chemother. 1988 May;32(5):702–5. doi: 10.1128/aac.32.5.702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Klepser ME, Wolfe EJ, Jones RN, Nightingale CH, Pfaller MA. Antifungal pharmacodynamic characteristics of fluconazole and amphotericin B tested against Candida albicans. Antimicrob Agents Chemother. 1997 Jun;41(6):1392–5. doi: 10.1128/aac.41.6.1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Pfaller MA, Messer SA, Hollis RJ. Strain delineation and antifungal susceptibilities of epidemiologically related and unrelated isolates of Candida lusitaniae. Diagn Microbiol Infect Dis. 1994 Nov;20(3):127–33. doi: 10.1016/0732-8893(94)90106-6. [DOI] [PubMed] [Google Scholar]
  • [12].Sabatelli F, Patel R, Mann PA, et al. In vitro activities of posaconazole, fluconazole, itraconazole, voriconazole, and amphotericin B against a large collection of clinically important molds and yeasts. Antimicrob Agents Chemother. 2006 Jun;50(6):2009–15. doi: 10.1128/AAC.00163-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Sutton DA, Sanche SE, Revankar SG, Fothergill AW, Rinaldi MG. In vitro amphotericin B resistance in clinical isolates of Aspergillus terreus, with a head-to-head comparison to voriconazole. J Clin Microbiol. 1999 Jul;37(7):2343–5. doi: 10.1128/jcm.37.7.2343-2345.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Walsh TJ, Melcher GP, Rinaldi MG, et al. Trichosporon beigelii, an emerging pathogen resistant to amphotericin B. J Clin Microbiol. 1990 Jul;28(7):1616–22. doi: 10.1128/jcm.28.7.1616-1622.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Ernst EJ, Klepser ME, Pfaller MA. Postantifungal effects of echinocandin, azole, and polyene antifungal agents against Candida albicans and Cryptococcus neoformans. Antimicrob Agents Chemother. 2000 Apr;44(4):1108–11. doi: 10.1128/aac.44.4.1108-1111.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].EMA Cancidas : EPAR - Product Information. 2013.
  • [17].EMA Noxafil : EPAR - Product Information. 2012 [Google Scholar]
  • [18].Lass-Florl C, Kofler G, Kropshofer G, et al. In-vitro testing of susceptibility to amphotericin B is a reliable predictor of clinical outcome in invasive aspergillosis. J Antimicrob Chemother. 1998 Oct;42(4):497–502. doi: 10.1093/jac/42.4.497. [DOI] [PubMed] [Google Scholar]
  • [19].Park BJ, Arthington-Skaggs BA, Hajjeh RA, et al. Evaluation of amphotericin B interpretive breakpoints for Candida bloodstream isolates by correlation with therapeutic outcome. Antimicrob Agents Chemother. 2006 Apr;50(4):1287–92. doi: 10.1128/AAC.50.4.1287-1292.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Kravetz HM, Andriole VT, Huber MA, Utz JP. Oral administration of solubilized amphotericin B. N Engl J Med. 1961 Jul 27;265:183–4. doi: 10.1056/NEJM196107272650409. [DOI] [PubMed] [Google Scholar]
  • [21].Christiansen KJ, Bernard EM, Gold JW, Armstrong D. Distribution and activity of amphotericin B in humans. J Infect Dis. 1985 Nov;152(5):1037–43. doi: 10.1093/infdis/152.5.1037. [DOI] [PubMed] [Google Scholar]
  • [22].Luna B, Drew RH, Perfect JR. Agents for treatment of invasive fungal infections. Otolaryngol Clin North Am. 2000 Apr;33(2):277–99. doi: 10.1016/s0030-6665(00)80005-5. [DOI] [PubMed] [Google Scholar]
  • [23].Baley JE, Meyers C, Kliegman RM, Jacobs MR, Blumer JL. Pharmacokinetics, outcome of treatment, and toxic effects of amphotericin B and 5-fluorocytosine in neonates. J Pediatr. 1990 May;116(5):791–7. doi: 10.1016/s0022-3476(05)82674-5. [DOI] [PubMed] [Google Scholar]
  • [24].Benson JM, Nahata MC. Pharmacokinetics of amphotericin B in children. Antimicrob Agents Chemother. 1989 Nov;33(11):1989–93. doi: 10.1128/aac.33.11.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Koren G, Lau A, Klein J, et al. Pharmacokinetics and adverse effects of amphotericin B in infants and children. J Pediatr. 1988 Sep;113(3):559–63. doi: 10.1016/s0022-3476(88)80653-x. [DOI] [PubMed] [Google Scholar]
  • [26].Nath CE, McLachlan AJ, Shaw PJ, Gunning R, Earl JW. Population pharmacokinetics of amphotericin B in children with malignant diseases. Br J Clin Pharmacol. 2001 Dec;52(6):671–80. doi: 10.1046/j.1365-2125.2001.01496.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Starke JR, Mason EO, Jr., Kramer WG, Kaplan SL. Pharmacokinetics of amphotericin B in infants and children. J Infect Dis. 1987 Apr;155(4):766–74. doi: 10.1093/infdis/155.4.766. [DOI] [PubMed] [Google Scholar]
  • [28].Wingard JR, Kubilis P, Lee L, et al. Clinical significance of nephrotoxicity in patients treated with amphotericin B for suspected or proven aspergillosis. Clin Infect Dis. 1999 Dec;29(6):1402–7. doi: 10.1086/313498. [DOI] [PubMed] [Google Scholar]
  • [29].Holler B, Omar SA, Farid MD, Patterson MJ. Effects of fluid and electrolyte management on amphotericin B-induced nephrotoxicity among extremely low birth weight infants. Pediatrics. 2004 Jun;113(6):e608–16. doi: 10.1542/peds.113.6.e608. [DOI] [PubMed] [Google Scholar]
  • [30].Le J, Adler-Shohet FC, Nguyen C, Lieberman JM. Nephrotoxicity associated with amphotericin B deoxycholate in neonates. Pediatr Infect Dis J. 2009 Dec;28(12):1061–3. doi: 10.1097/INF.0b013e3181af6201. [DOI] [PubMed] [Google Scholar]
  • [31].Linder N, Klinger G, Shalit I, et al. Treatment of candidaemia in premature infants: comparison of three amphotericin B preparations. J Antimicrob Chemother. 2003 Oct;52(4):663–7. doi: 10.1093/jac/dkg419. [DOI] [PubMed] [Google Scholar]
  • [32].Goodwin SD, Cleary JD, Walawander CA, Taylor JW, Grasela TH., Jr. Pretreatment regimens for adverse events related to infusion of amphotericin B. Clin Infect Dis. 1995 Apr;20(4):755–61. doi: 10.1093/clinids/20.4.755. [DOI] [PubMed] [Google Scholar]
  • [33].Andes D, Safdar N, Marchillo K, Conklin R. Pharmacokinetic-pharmacodynamic comparison of amphotericin B (AMB) and two lipid-associated AMB preparations, liposomal AMB and AMB lipid complex, in murine candidiasis models. Antimicrob Agents Chemother. 2006 Feb;50(2):674–84. doi: 10.1128/AAC.50.2.674-684.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Ralph ED, Khazindar AM, Barber KR, Grant CW. Comparative in vitro effects of liposomal amphotericin B, amphotericin B-deoxycholate, and free amphotericin B against fungal strains determined by using MIC and minimal lethal concentration susceptibility studies and time-kill curves. Antimicrob Agents Chemother. 1991 Jan;35(1):188–91. doi: 10.1128/aac.35.1.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Merck 2012. NOXAFIL (posaconazole) [package insert]
  • [36].Subira M, Martino R, Gomez L, Marti JM, Estany C, Sierra J. Low-dose amphotericin B lipid complex vs. conventional amphotericin B for empirical antifungal therapy of neutropenic fever in patients with hematologic malignancies--a randomized, controlled trial. Eur J Haematol. 2004 May;72(5):342–7. doi: 10.1111/j.1600-0609.2004.00239.x. [DOI] [PubMed] [Google Scholar]
  • [37].Fleming RV, Kantarjian HM, Husni R, et al. Comparison of amphotericin B lipid complex (ABLC) vs. ambisome in the treatment of suspected or documented fungal infections in patients with leukemia. Leuk Lymphoma. 2001 Feb;40(5-6):511–20. doi: 10.3109/10428190109097650. [DOI] [PubMed] [Google Scholar]
  • [38].Hong Y, Shaw PJ, Nath CE, et al. Population pharmacokinetics of liposomal amphotericin B in pediatric patients with malignant diseases. Antimicrob Agents Chemother. 2006 Mar;50(3):935–42. doi: 10.1128/AAC.50.3.935-942.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Walsh TJ, Whitcomb P, Piscitelli S, et al. Safety, tolerance, and pharmacokinetics of amphotericin B lipid complex in children with hepatosplenic candidiasis. Antimicrob Agents Chemother. 1997 Sep;41(9):1944–8. doi: 10.1128/aac.41.9.1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Adedoyin A, Bernardo JF, Swenson CE, et al. Pharmacokinetic profile of ABELCET (amphotericin B lipid complex injection): combined experience from phase I and phase II studies. Antimicrob Agents Chemother. 1997 Oct;41(10):2201–8. doi: 10.1128/aac.41.10.2201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Wurthwein G, Groll AH, Hempel G, Adler-Shohet FC, Lieberman JM, Walsh TJ. Population pharmacokinetics of amphotericin B lipid complex in neonates. Antimicrob Agents Chemother. 2005 Dec;49(12):5092–8. doi: 10.1128/AAC.49.12.5092-5098.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Groll AHMD, Petraitis V, Petraitiene R, Roussillion K, Hemmings M, Lyman LA, Walsh TJ, editors. Disposition and efficacy of amphotericin B formulations in a kidney target model of invasive candidiasis. 41st Interscience Conference Antimicrobial Agents Chemotherapy; Chicago, IL. 2001. [Google Scholar]
  • [43].Groll AH, Giri N, Petraitis V, et al. Comparative efficacy and distribution of lipid formulations of amphotericin B in experimental Candida albicans infection of the central nervous system. J Infect Dis. 2000 Jul;182(1):274–82. doi: 10.1086/315643. [DOI] [PubMed] [Google Scholar]
  • [44].Ascher SB, Smith PB, Watt K, et al. Antifungal therapy and outcomes in infants with invasive Candida infections. Pediatr Infect Dis J. 2012 May;31(5):439–43. doi: 10.1097/INF.0b013e3182467a72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Bowden R, Chandrasekar P, White MH, et al. A double-blind, randomized, controlled trial of amphotericin B colloidal dispersion versus amphotericin B for treatment of invasive aspergillosis in immunocompromised patients. Clin Infect Dis. 2002 Aug 15;35(4):359–66. doi: 10.1086/341401. [DOI] [PubMed] [Google Scholar]
  • [46].Cetin H, Yalaz M, Akisu M, Hilmioglu S, Metin D, Kultursay N. The efficacy of two different lipid-based amphotericin B in neonatal Candida septicemia. Pediatr Int. 2005 Dec;47(6):676–80. doi: 10.1111/j.1442-200x.2005.02135.x. [DOI] [PubMed] [Google Scholar]
  • [47].Juster-Reicher A, Flidel-Rimon O, Amitay M, Even-Tov S, Shinwell E, Leibovitz E. High-dose liposomal amphotericin B in the therapy of systemic candidiasis in neonates. Eur J Clin Microbiol Infect Dis. 2003 Oct;22(10):603–7. doi: 10.1007/s10096-003-0993-4. [DOI] [PubMed] [Google Scholar]
  • [48].Juster-Reicher A, Leibovitz E, Linder N, et al. Liposomal amphotericin B (AmBisome) in the treatment of neonatal candidiasis in very low birth weight infants. Infection. 2000 Jul-Aug;28(4):223–6. doi: 10.1007/s150100070040. [DOI] [PubMed] [Google Scholar]
  • [49].Scarcella A, Pasquariello MB, Giugliano B, Vendemmia M, de Lucia A. Liposomal amphotericin B treatment for neonatal fungal infections. Pediatr Infect Dis J. 1998 Feb;17(2):146–8. doi: 10.1097/00006454-199802000-00013. [DOI] [PubMed] [Google Scholar]
  • [50].Walsh TJ, Seibel NL, Arndt C, et al. Amphotericin B lipid complex in pediatric patients with invasive fungal infections. Pediatr Infect Dis J. 1999 Aug;18(8):702–8. doi: 10.1097/00006454-199908000-00010. [DOI] [PubMed] [Google Scholar]
  • [51].Damle B, Stogniew M, Dowell J. Pharmacokinetics and tissue distribution of anidulafungin in rats. Antimicrob Agents Chemother. 2008 Jul;52(7):2673–6. doi: 10.1128/AAC.01596-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Pfizer . New York: Diflucan [package insert] USA2013. [Google Scholar]
  • [53].Carrillo AJ, Guarro J. In vitro activities of four novel triazoles against Scedosporium spp. Antimicrob Agents Chemother. 2001 Jul;45(7):2151–3. doi: 10.1128/AAC.45.7.2151-2153.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Lewis RE, Lund BC, Klepser ME, Ernst EJ, Pfaller MA. Assessment of antifungal activities of fluconazole and amphotericin B administered alone and in combination against Candida albicans by using a dynamic in vitro mycotic infection model. Antimicrob Agents Chemother. 1998 Jun;42(6):1382–6. doi: 10.1128/aac.42.6.1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Louie A, Drusano GL, Banerjee P, et al. Pharmacodynamics of fluconazole in a murine model of systemic candidiasis. Antimicrob Agents Chemother. 1998 May;42(5):1105–9. doi: 10.1128/aac.42.5.1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Andes D, van Ogtrop M. Characterization and quantitation of the pharmacodynamics of fluconazole in a neutropenic murine disseminated candidiasis infection model. Antimicrob Agents Chemother. 1999 Sep;43(9):2116–20. doi: 10.1128/aac.43.9.2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Rex JH, Pfaller MA, Galgiani JN, et al. Development of interpretive breakpoints for antifungal susceptibility testing: conceptual framework and analysis of in vitro-in vivo correlation data for fluconazole, itraconazole, and candida infections. Subcommittee on Antifungal Susceptibility Testing of the National Committee for Clinical Laboratory Standards. Clin Infect Dis. 1997 Feb;24(2):235–47. doi: 10.1093/clinids/24.2.235. [DOI] [PubMed] [Google Scholar]
  • [58].Baddley JW, Patel M, Fau - Bhavnani SM, Bhavnani Sm Fau - Moser SA, Moser Sa Fau - Andes DR, Andes DR. Association of fluconazole pharmacodynamics with mortality in patients with candidemia. 20080826 DCOM- 20081023(1098-6596 (Electronic)) [DOI] [PMC free article] [PubMed]
  • [59].Seay RE, Larson TA, Toscano JP, Bostrom BC, O’Leary MC, Uden DL. Pharmacokinetics of fluconazole in immune-compromised children with leukemia or other hematologic diseases. Pharmacotherapy. 1995 Jan-Feb;15(1):52–8. [PubMed] [Google Scholar]
  • [60].Brammer KW, Farrow PR, Faulkner JK. Pharmacokinetics and tissue penetration of fluconazole in humans. Rev Infect Dis. 1990 Mar-Apr;12(Suppl 3):S318–26. doi: 10.1093/clinids/12.supplement_3.s318. [DOI] [PubMed] [Google Scholar]
  • [61].Tucker RM, Williams PL, Arathoon EG, et al. Pharmacokinetics of fluconazole in cerebrospinal fluid and serum in human coccidioidal meningitis. Antimicrob Agents Chemother. 1988 Mar;32(3):369–73. doi: 10.1128/aac.32.3.369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Gross AS, McLachlan AJ, Minns I, Beal JB, Tett SE. Simultaneous administration of a cocktail of markers to measure renal drug elimination pathways: absence of a pharmacokinetic interaction between fluconazole and sinistrin, p-aminohippuric acid and pindolol. Br J Clin Pharmacol. 2001 Jun;51(6):547–55. doi: 10.1046/j.1365-2125.2001.01390.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Soczo G, Kardos G, McNicholas PM, et al. Correlation of posaconazole minimum fungicidal concentration and time kill test against nine Candida species. J Antimicrob Chemother. 2007 Nov;60(5):1004–9. doi: 10.1093/jac/dkm350. [DOI] [PubMed] [Google Scholar]
  • [64].Wade KC, Wu D, Kaufman DA, et al. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother. 2008 Nov;52(11):4043–9. doi: 10.1128/AAC.00569-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Piper L, Smith PB, Hornik CP, et al. Fluconazole loading dose pharmacokinetics and safety in infants. Pediatr Infect Dis J. 2011 May;30(5):375–8. doi: 10.1097/INF.0b013e318202cbb3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Brammer KW, Coates PE. Pharmacokinetics of fluconazole in pediatric patients. Eur J Clin Microbiol Infect Dis. 1994 Apr;13(4):325–9. doi: 10.1007/BF01974613. [DOI] [PubMed] [Google Scholar]
  • [67].Osowski CL, Dix SP, Lin LS, Mullins RE, Geller RB, Wingard JR. Evaluation of the drug interaction between intravenous high-dose fluconazole and cyclosporine or tacrolimus in bone marrow transplant patients. Transplantation. 1996 Apr 27;61(8):1268–72. doi: 10.1097/00007890-199604270-00026. [DOI] [PubMed] [Google Scholar]
  • [68].Novelli V, Holzel H. Safety and tolerability of fluconazole in children. Antimicrob Agents Chemother. 1999 Aug;43(8):1955–60. doi: 10.1128/aac.43.8.1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Driessen M, Ellis JB, Cooper PA, et al. Fluconazole vs. amphotericin B for the treatment of neonatal fungal septicemia: a prospective randomized trial. Pediatr Infect Dis J. 1996 Dec;15(12):1107–12. doi: 10.1097/00006454-199612000-00011. [DOI] [PubMed] [Google Scholar]
  • [70].Manzoni P, Stolfi I, Pugni L, et al. A multicenter, randomized trial of prophylactic fluconazole in preterm neonates. N Engl J Med. 2007 Jun 14;356(24):2483–95. doi: 10.1056/NEJMoa065733. [DOI] [PubMed] [Google Scholar]
  • [71].Fung-Tomc JC, White TC, Minassian B, Huczko E, Bonner DP. In vitro antifungal activity of BMS-207147 and itraconazole against yeast strains that are non-susceptible to fluconazole. Diagn Microbiol Infect Dis. 1999 Oct;35(2):163–7. doi: 10.1016/s0732-8893(99)00063-2. [DOI] [PubMed] [Google Scholar]
  • [72].Manavathu EK, Cutright JL, Chandrasekar PH. Organism-dependent fungicidal activities of azoles. Antimicrob Agents Chemother. 1998 Nov;42(11):3018–21. doi: 10.1128/aac.42.11.3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Burgess DS, Hastings RW, Summers KK, Hardin TC, Rinaldi MG. Pharmacodynamics of fluconazole, itraconazole, and amphotericin B against Candida albicans. Diagn Microbiol Infect Dis. 2000 Jan;36(1):13–8. doi: 10.1016/s0732-8893(99)00097-8. [DOI] [PubMed] [Google Scholar]
  • [74].Groll AH, Wood L, Roden M, et al. Safety, pharmacokinetics, and pharmacodynamics of cyclodextrin itraconazole in pediatric patients with oropharyngeal candidiasis. Antimicrob Agents Chemother. 2002 Aug;46(8):2554–63. doi: 10.1128/AAC.46.8.2554-2563.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Espinel-Ingroff A. Comparison of In vitro activities of the new triazole SCH56592 and the echinocandins MK-0991 (L-743,872) and LY303366 against opportunistic filamentous and dimorphic fungi and yeasts. J Clin Microbiol. 1998 Oct;36(10):2950–6. doi: 10.1128/jcm.36.10.2950-2956.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].de Repentigny L, Ratelle J, Leclerc JM, et al. Repeated-dose pharmacokinetics of an oral solution of itraconazole in infants and children. Antimicrob Agents Chemother. 1998 Feb;42(2):404–8. doi: 10.1128/aac.42.2.404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Prentice AG, Warnock DW, Johnson SA, Taylor PC, Oliver DA. Multiple dose pharmacokinetics of an oral solution of itraconazole in patients receiving chemotherapy for acute myeloid leukaemia. J Antimicrob Chemother. 1995 Oct;36(4):657–63. doi: 10.1093/jac/36.4.657. [DOI] [PubMed] [Google Scholar]
  • [78].Schmitt C, Perel Y, Harousseau JL, et al. Pharmacokinetics of itraconazole oral solution in neutropenic children during long-term prophylaxis. Antimicrob Agents Chemother. 2001 May;45(5):1561–4. doi: 10.1128/AAC.45.5.1561-1564.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Hardin TC, Graybill JR, Fetchick R, Woestenborghs R, Rinaldi MG, Kuhn JG. Pharmacokinetics of itraconazole following oral administration to normal volunteers. Antimicrob Agents Chemother. 1988 Sep;32(9):1310–3. doi: 10.1128/aac.32.9.1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Foot AB, Veys PA, Gibson BE. Itraconazole oral solution as antifungal prophylaxis in children undergoing stem cell transplantation or intensive chemotherapy for haematological disorders. Bone Marrow Transplant. 1999 Nov;24(10):1089–93. doi: 10.1038/sj.bmt.1702023. [DOI] [PubMed] [Google Scholar]
  • [81].Lestner JM, Roberts SA, Moore CB, Howard SJ, Denning DW, Hope WW. Toxicodynamics of itraconazole: implications for therapeutic drug monitoring. Clin Infect Dis. 2009 Sep 15;49(6):928–30. doi: 10.1086/605499. [DOI] [PubMed] [Google Scholar]
  • [82].Bermudez M, Fuster JL, Llinares E, Galera A, Gonzalez C. Itraconazole-related increased vincristine neurotoxicity: case report and review of literature. J Pediatr Hematol Oncol. 2005 Jul;27(7):389–92. doi: 10.1097/01.mph.0000172751.06286.5b. [DOI] [PubMed] [Google Scholar]
  • [83].Merck 2013. CANCIDAS (caspofungin acetate) [package insert]
  • [84].Benjamin DK, Jr., Deville JG, Azie N, et al. Safety and pharmacokinetic profiles of repeated-dose micafungin in children and adolescents treated for invasive candidiasis. Pediatr Infect Dis J. 2013 Nov;32(11):e419–25. doi: 10.1097/INF.0b013e31829efd14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Koks CH, Meenhorst PL, Hillebrand MJ, Bult A, Beijnen JH. Pharmacokinetics of fluconazole in saliva and plasma after administration of an oral suspension and capsules. Antimicrob Agents Chemother. 1996 Aug;40(8):1935–7. doi: 10.1128/aac.40.8.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Abdel-Rahman SM, Jacobs RF, Massarella J, et al. Single-dose pharmacokinetics of intravenous itraconazole and hydroxypropyl-beta-cyclodextrin in infants, children, and adolescents. Antimicrob Agents Chemother. 2007 Aug;51(8):2668–73. doi: 10.1128/AAC.00297-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Espinel-Ingroff A, Johnson E, Hockey H, Troke P. Activities of voriconazole, itraconazole and amphotericin B in vitro against 590 moulds from 323 patients in the voriconazole Phase III clinical studies. J Antimicrob Chemother. 2008 Mar;61(3):616–20. doi: 10.1093/jac/dkm518. [DOI] [PubMed] [Google Scholar]
  • [88].Johnson EM, Szekely A, Warnock DW. In-vitro activity of voriconazole, itraconazole and amphotericin B against filamentous fungi. J Antimicrob Chemother. 1998 Dec;42(6):741–5. doi: 10.1093/jac/42.6.741. [DOI] [PubMed] [Google Scholar]
  • [89].Klepser ME, Malone D, Lewis RE, Ernst EJ, Pfaller MA. Evaluation of voriconazole pharmacodynamics using time-kill methodology. Antimicrob Agents Chemother. 2000 Jul;44(7):1917–20. doi: 10.1128/aac.44.7.1917-1920.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Andes D, Marchillo K, Stamstad T, Conklin R. In vivo pharmacodynamics of a new triazole, ravuconazole, in a murine candidiasis model. Antimicrob Agents Chemother. 2003 Apr;47(4):1193–9. doi: 10.1128/AAC.47.4.1193-1199.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Neely M, Rushing T, Kovacs A, Jelliffe R, Hoffman J. Voriconazole pharmacokinetics and pharmacodynamics in children. Clin Infect Dis. 2010 Jan 1;50(1):27–36. doi: 10.1086/648679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Troke PF, Hockey HP, Hope WW. Observational study of the clinical efficacy of voriconazole and its relationship to plasma concentrations in patients. Antimicrob Agents Chemother. 2011 Oct;55(10):4782–8. doi: 10.1128/AAC.01083-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lutsar I, Roffey S, Troke P. Voriconazole concentrations in the cerebrospinal fluid and brain tissue of guinea pigs and immunocompromised patients. Clin Infect Dis. 2003 Sep 1;37(5):728–32. doi: 10.1086/377131. [DOI] [PubMed] [Google Scholar]
  • [94].Weiler S, Fiegl D, MacFarland R, et al. Human tissue distribution of voriconazole. Antimicrob Agents Chemother. 2011 Feb;55(2):925–8. doi: 10.1128/AAC.00949-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Karlsson MO, Lutsar I, Milligan PA. Population pharmacokinetic analysis of voriconazole plasma concentration data from pediatric studies. Antimicrob Agents Chemother. 2009 Mar;53(3):935–44. doi: 10.1128/AAC.00751-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Narita A, Muramatsu H, Sakaguchi H, et al. Correlation of CYP2C19 phenotype with voriconazole plasma concentration in children. J Pediatr Hematol Oncol. 2013 Jul;35(5):e219–23. doi: 10.1097/MPH.0b013e3182880eaa. [DOI] [PubMed] [Google Scholar]
  • [97].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 Jun;48(6):2166–72. doi: 10.1128/AAC.48.6.2166-2172.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Walsh TJ, Driscoll T, Milligan PA, et al. Pharmacokinetics, safety, and tolerability of voriconazole in immunocompromised children. Antimicrob Agents Chemother. 2010 Oct;54(10):4116–23. doi: 10.1128/AAC.00896-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Friberg LE, Ravva P, Karlsson MO, Liu P. Integrated population pharmacokinetic analysis of voriconazole in children, adolescents, and adults. Antimicrob Agents Chemother. 2012 Jun;56(6):3032–42. doi: 10.1128/AAC.05761-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Walsh TJ, Lutsar I, Driscoll T, et al. Voriconazole in the treatment of aspergillosis, scedosporiosis and other invasive fungal infections in children. Pediatr Infect Dis J. 2002 Mar;21(3):240–8. doi: 10.1097/00006454-200203000-00015. [DOI] [PubMed] [Google Scholar]
  • [101].Bernhard S, Kernland Lang K, Ammann RA, et al. Voriconazole-induced phototoxicity in children. Pediatr Infect Dis J. 2012 Jul;31(7):769–71. doi: 10.1097/INF.0b013e3182566311. [DOI] [PubMed] [Google Scholar]
  • [102].Suzuki Y, Tokimatsu I, Sato Y, et al. Association of sustained high plasma trough concentration of voriconazole with the incidence of hepatotoxicity. Clin Chim Acta. 2013 Jun 6;424C:119–22. doi: 10.1016/j.cca.2013.05.025. [DOI] [PubMed] [Google Scholar]
  • [103].Pascual A, Calandra T, Bolay S, Buclin T, Bille J, Marchetti O. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis. 2008 Jan 15;46(2):201–11. doi: 10.1086/524669. [DOI] [PubMed] [Google Scholar]
  • [104].Pieper S, Kolve H, Gumbinger HG, Goletz G, Wurthwein G, Groll AH. Monitoring of voriconazole plasma concentrations in immunocompromised paediatric patients. J Antimicrob Chemother. 2012 Nov;67(11):2717–24. doi: 10.1093/jac/dks258. [DOI] [PubMed] [Google Scholar]
  • [105].EMA Vfend: EPAR - Product information. 2013.
  • [106].Roerig VFEND (voriconazole) 2013. [package insert]
  • [107].Herbrecht R, Denning DW, Patterson TF, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002 Aug 8;347(6):408–15. doi: 10.1056/NEJMoa020191. [DOI] [PubMed] [Google Scholar]
  • [108].Park WB, Kim NH, Kim KH, et al. The effect of therapeutic drug monitoring on safety and efficacy of voriconazole in invasive fungal infections: a randomized controlled trial. Clin Infect Dis. 2012 Oct;55(8):1080–7. doi: 10.1093/cid/cis599. [DOI] [PubMed] [Google Scholar]
  • [109].Petraitiene R, Petraitis V, Groll AH, et al. Antifungal activity and pharmacokinetics of posaconazole (SCH 56592) in treatment and prevention of experimental invasive pulmonary aspergillosis: correlation with galactomannan antigenemia. Antimicrob Agents Chemother. 2001 Mar;45(3):857–69. doi: 10.1128/AAC.45.3.857-869.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Courtney R, Pai S, Laughlin M, Lim J, Batra V. Pharmacokinetics, safety, and tolerability of oral posaconazole administered in single and multiple doses in healthy adults. Antimicrob Agents Chemother. 2003 Sep;47(9):2788–95. doi: 10.1128/AAC.47.9.2788-2795.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Courtney R, Sansone A, Smith W, et al. Posaconazole pharmacokinetics, safety, and tolerability in subjects with varying degrees of chronic renal disease. J Clin Pharmacol. 2005 Feb;45(2):185–92. doi: 10.1177/0091270004271402. [DOI] [PubMed] [Google Scholar]
  • [112].Doring M, Muller C, Johann PD, et al. Analysis of posaconazole as oral antifungal prophylaxis in pediatric patients under 12 years of age following allogeneic stem cell transplantation. BMC Infect Dis. 2012;12:263. doi: 10.1186/1471-2334-12-263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Krishna G, Sansone-Parsons A, Martinho M, Kantesaria B, Pedicone L. Posaconazole plasma concentrations in juvenile patients with invasive fungal infection. Antimicrob Agents Chemother. 2007 Mar;51(3):812–8. doi: 10.1128/AAC.00454-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Segal BH, Barnhart LA, Anderson VL, Walsh TJ, Malech HL, Holland SM. Posaconazole as salvage therapy in patients with chronic granulomatous disease and invasive filamentous fungal infection. Clin Infect Dis. 2005 Jun 1;40(11):1684–8. doi: 10.1086/430068. [DOI] [PubMed] [Google Scholar]
  • [115].Welzen ME, Bruggemann RJ, Van Den Berg JM, et al. A twice daily posaconazole dosing algorithm for children with chronic granulomatous disease. Pediatr Infect Dis J. 2011 Sep;30(9):794–7. doi: 10.1097/INF.0b013e3182195808. [DOI] [PubMed] [Google Scholar]
  • [116].Jang SH, Colangelo PM, Gobburu JV. Exposure-response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther. 2010 Jul;88(1):115–9. doi: 10.1038/clpt.2010.64. [DOI] [PubMed] [Google Scholar]
  • [117].Ullmann AJ, Lipton JH, Vesole DH, et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med. 2007 Jan 25;356(4):335–47. doi: 10.1056/NEJMoa061098. [DOI] [PubMed] [Google Scholar]
  • [118].Roberts J, Schock K, Marino S, Andriole VT. Efficacies of two new antifungal agents, the triazole ravuconazole and the echinocandin LY-303366, in an experimental model of invasive aspergillosis. Antimicrob Agents Chemother. 2000 Dec;44(12):3381–8. doi: 10.1128/aac.44.12.3381-3388.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Ramos G, Cuenca-Estrella M, Monzon A, Rodriguez-Tudela JL. In-vitro comparative activity of UR-9825, itraconazole and fluconazole against clinical isolates of Candida spp. J Antimicrob Chemother. 1999 Aug;44(2):283–6. doi: 10.1093/jac/44.2.283. [DOI] [PubMed] [Google Scholar]
  • [120].Miller JL, Schell WA, Wills EA, et al. In vitro and in vivo efficacies of the new triazole albaconazole against Cryptococcus neoformans. Antimicrob Agents Chemother. 2004 Feb;48(2):384–7. doi: 10.1128/AAC.48.2.384-387.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Capilla J, Ortoneda M, Pastor FJ, Guarro J. In vitro antifungal activities of the new triazole UR-9825 against clinically important filamentous fungi. Antimicrob Agents Chemother. 2001 Sep;45(9):2635–7. doi: 10.1128/AAC.45.9.2635-2637.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Cuenca-Estrella M, Gomez-Lopez A, Mellado E, Garcia-Effron G, Monzon A, Rodriguez-Tudela JL. In vitro activity of ravuconazole against 923 clinical isolates of nondermatophyte filamentous fungi. Antimicrob Agents Chemother. 2005 Dec;49(12):5136–8. doi: 10.1128/AAC.49.12.5136-5138.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Lepak AJ, Marchillo K, Vanhecker J, Diekema D, Andes DR. Isavuconazole Pharmacodynamic Target Determination for Candida species in an In vivo Murine Disseminated Candidiasis Model. Antimicrob Agents Chemother. 2013 Sep 3; doi: 10.1128/AAC.01354-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Gupta AK, Leonardi C, Stoltz RR, Pierce PF, Conetta B. A phase I/II randomized, double-blind, placebo-controlled, dose-ranging study evaluating the efficacy, safety and pharmacokinetics of ravuconazole in the treatment of onychomycosis. J Eur Acad Dermatol Venereol. 2005 Jul;19(4):437–43. doi: 10.1111/j.1468-3083.2005.01212.x. [DOI] [PubMed] [Google Scholar]
  • [125].Schmitt-Hoffmann A, Roos B, Heep M, et al. Single-ascending-dose pharmacokinetics and safety of the novel broad-spectrum antifungal triazole BAL4815 after intravenous infusions (50, 100, and 200 milligrams) and oral administrations (100, 200, and 400 milligrams) of its prodrug, BAL8557, in healthy volunteers. Antimicrob Agents Chemother. 2006 Jan;50(1):279–85. doi: 10.1128/AAC.50.1.279-285.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Benjamin DK, Jr., Smith PB, Arrieta A, et al. Safety and pharmacokinetics of repeat-dose micafungin in young infants. Clin Pharmacol Ther. 2010 Jan;87(1):93–9. doi: 10.1038/clpt.2009.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].van Rossem K, Lowe JA. A Phase 1, randomized, open-label crossover study to evaluate the safety and pharmacokinetics of 400 mg albaconazole administered to healthy participants as a tablet formulation versus a capsule formulation. Clin Pharmacol. 2013;5:23–31. doi: 10.2147/CPAA.S39600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Hatano K, Morishita Y, Nakai T, Ikeda F. Antifungal mechanism of FK463 against Candida albicans and Aspergillus fumigatus. J Antibiot (Tokyo) 2002 Feb;55(2):219–22. doi: 10.7164/antibiotics.55.219. [DOI] [PubMed] [Google Scholar]
  • [129].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 Jan;44(1):57–62. doi: 10.1128/aac.44.1.57-62.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Ikeda F, Wakai Y, Matsumoto S, et al. Efficacy of FK463, a new lipopeptide antifungal agent, in mouse models of disseminated candidiasis and aspergillosis. Antimicrob Agents Chemother. 2000 Mar;44(3):614–8. doi: 10.1128/aac.44.3.614-618.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Petraitis V, Petraitiene R, Groll AH, et al. Comparative antifungal activities and plasma pharmacokinetics of micafungin (FK463) against disseminated candidiasis and invasive pulmonary aspergillosis in persistently neutropenic rabbits. Antimicrob Agents Chemother. 2002 Jun;46(6):1857–69. doi: 10.1128/AAC.46.6.1857-1869.2002. [Comparative Study] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].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 Jan 1;197(1):163–71. doi: 10.1086/524063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Okugawa S, Ota Y, Tatsuno K, Tsukada K, Kishino S, Koike K. A case of invasive central nervous system aspergillosis treated with micafungin with monitoring of micafungin concentrations in the cerebrospinal fluid. Scand J Infect Dis. 2007;39(4):344–6. doi: 10.1080/00365540600951333. [DOI] [PubMed] [Google Scholar]
  • [134].Astellas [Accessed 2011];Mycamine (micafungin)[package insert] 2011 Available from: http://www.astellas.us/docs/mycamine.pdf.
  • [135].Sandage BCG, Najarian N, Lowther J, editors. Pharmacokinetics and fungicidal activity of aminocandin (HMR3270), a novel echinocandin in healthy volunteers. 15th European Congress of Clinical Microbiology and Infectious Diseases; Copenhagen, Denmark. 2005 April 2-5 2005. [Google Scholar]
  • [136].Tabata K, Katashima M, Kawamura A, Tanigawara Y, Sunagawa K. Linear pharmacokinetics of micafungin and its active metabolites in Japanese pediatric patients with fungal infections. Biol Pharm Bull. 2006 Aug;29(8):1706–11. doi: 10.1248/bpb.29.1706. [DOI] [PubMed] [Google Scholar]
  • [137].Hope WW, Seibel NL, Schwartz CL, et al. Population pharmacokinetics of micafungin in pediatric patients and implications for antifungal dosing. Antimicrob Agents Chemother. 2007 Oct;51(10):3714–9. doi: 10.1128/AAC.00398-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Seibel NL, Schwartz C, Arrieta A, et al. Safety, tolerability, and pharmacokinetics of Micafungin (FK463) in febrile neutropenic pediatric patients. Antimicrob Agents Chemother. 2005 Aug;49(8):3317–24. doi: 10.1128/AAC.49.8.3317-3324.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Hope WW, Smith PB, Arrieta A, et al. Population pharmacokinetics of micafungin in neonates and young infants. Antimicrob Agents Chemother. Jun;54(6):2633–7. doi: 10.1128/AAC.01679-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Smith PB, Walsh TJ, Hope W, et al. Pharmacokinetics of an elevated dosage of micafungin in premature neonates. Pediatr Infect Dis J. 2009 May;28(5):412–5. doi: 10.1097/INF.0b013e3181910e2d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Arrieta AC, Maddison P, Groll AH. Safety of micafungin in pediatric clinical trials. Pediatr Infect Dis J. 2011 Jun;30(6):e97–e102. doi: 10.1097/INF.0b013e3182127eaf. [DOI] [PubMed] [Google Scholar]
  • [142].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 Sep;27(9):820–6. doi: 10.1097/INF.0b013e31817275e6. [DOI] [PubMed] [Google Scholar]
  • [143].Astellas Pharma I . Tokyo, Japan: 2013. Mycamine [package insert] [Google Scholar]
  • [144].EMA Mycamine : EPAR - Product Information. 2013.
  • [145].Ernst EJ, Klepser ME, Ernst ME, Messer SA, Pfaller MA. In vitro pharmacodynamic properties of MK-0991 determined by time-kill methods. Diagn Microbiol Infect Dis. 1999 Feb;33(2):75–80. doi: 10.1016/s0732-8893(98)00130-8. [DOI] [PubMed] [Google Scholar]
  • [146].Faix RG. Systemic Candida infections in infants in intensive care nurseries: high incidence of central nervous system involvement. J Pediatr. 1984 Oct;105(4):616–22. doi: 10.1016/s0022-3476(84)80433-3. [DOI] [PubMed] [Google Scholar]
  • [147].Bowman JC, Hicks PS, Kurtz MB, et al. The antifungal echinocandin caspofungin acetate kills growing cells of Aspergillus fumigatus in vitro. Antimicrob Agents Chemother. 2002 Sep;46(9):3001–12. doi: 10.1128/AAC.46.9.3001-3012.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Li CC, Sun P, Dong Y, et al. Population pharmacokinetics and pharmacodynamics of caspofungin in pediatric patients. Antimicrob Agents Chemother. 2011 May;55(5):2098–105. doi: 10.1128/AAC.00905-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Neely M, Jafri HS, Seibel N, et al. Pharmacokinetics and safety of caspofungin in older infants and toddlers. Antimicrob Agents Chemother. 2009 Apr;53(4):1450–6. doi: 10.1128/AAC.01027-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Walsh TJ, Adamson PC, Seibel NL, et al. Pharmacokinetics, safety, and tolerability of caspofungin in children and adolescents. Antimicrob Agents Chemother. 2005 Nov;49(11):4536–45. doi: 10.1128/AAC.49.11.4536-4545.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Balani SK, Xu X, Arison BH, et al. Metabolites of caspofungin acetate, a potent antifungal agent, in human plasma and urine. Drug Metab Dispos. 2000 Nov;28(11):1274–8. [PubMed] [Google Scholar]
  • [152].Sobel JD, Bradshaw SK, Lipka CJ, Kartsonis NA. Caspofungin in the treatment of symptomatic candiduria. Clin Infect Dis. 2007 Mar 1;44(5):e46–9. doi: 10.1086/510432. [DOI] [PubMed] [Google Scholar]
  • [153].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 Mar;53(3):869–75. doi: 10.1128/AAC.00868-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Cornely OA, Vehreschild JJ, Vehreschild MJ, et al. Phase II dose escalation study of caspofungin for invasive Aspergillosis. Antimicrob Agents Chemother. 2011 Dec;55(12):5798–803. doi: 10.1128/AAC.05134-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].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 Mar;123(3):877–84. doi: 10.1542/peds.2008-1158. [DOI] [PubMed] [Google Scholar]
  • [156].Franklin JA, McCormick J, Flynn PM. Retrospective study of the safety of caspofungin in immunocompromised pediatric patients. Pediatr Infect Dis J. 2003 Aug;22(8):747–9. doi: 10.1097/01.inf.0000078164.80233.14. [DOI] [PubMed] [Google Scholar]
  • [157].Natarajan G, Lulic-Botica M, Rongkavilit C, Pappas A, Bedard M. Experience with caspofungin in the treatment of persistent fungemia in neonates. J Perinatol. 2005 Dec;25(12):770–7. doi: 10.1038/sj.jp.7211380. [DOI] [PubMed] [Google Scholar]
  • [158].Odio CM, Araya R, Pinto LE, et al. Caspofungin therapy of neonates with invasive candidiasis. Pediatr Infect Dis J. 2004 Dec;23(12):1093–7. [PubMed] [Google Scholar]
  • [159].Zhanel GG, Karlowsky JA, Harding GA, et al. In vitro activity of a new semisynthetic echinocandin, LY-303366, against systemic isolates of Candida species, Cryptococcus neoformans, Blastomyces dermatitidis, and Aspergillus species. Antimicrob Agents Chemother. 1997 Apr;41(4):863–5. doi: 10.1128/aac.41.4.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Petraitiene R, Petraitis V, Groll AH, et al. Antifungal activity of LY303366, a novel echinocandin B, in experimental disseminated candidiasis in rabbits. Antimicrob Agents Chemother. 1999 Sep;43(9):2148–55. doi: 10.1128/aac.43.9.2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Pfaller MA, Boyken L, Hollis RJ, Messer SA, Tendolkar S, Diekema DJ. In vitro activities of anidulafungin against more than 2,500 clinical isolates of Candida spp., including 315 isolates resistant to fluconazole. J Clin Microbiol. 2005 Nov;43(11):5425–7. doi: 10.1128/JCM.43.11.5425-5427.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Andes D, Diekema DJ, Pfaller MA, et al. In vivo pharmacodynamic characterization of anidulafungin in a neutropenic murine candidiasis model. Antimicrob Agents Chemother. 2008 Feb;52(2):539–50. doi: 10.1128/AAC.01061-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].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 Feb;50(2):632–8. doi: 10.1128/AAC.50.2.632-638.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].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 May;89(5):702–7. doi: 10.1038/clpt.2011.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].EMA ECALTA (anidulafungin): EPAR - Product Information. 2013.
  • [166].Roerig ERAXIS (anidulafungin) [package insert] 2012.
  • [167].Warn PA, Sharp A, Morrissey G, Denning DW. Activity of aminocandin (IP960; HMR3270) compared with amphotericin B, itraconazole, caspofungin and micafungin in neutropenic murine models of disseminated infection caused by itraconazole-susceptible and -resistant strains of Aspergillus fumigatus. Int J Antimicrob Agents. 2010 Feb;35(2):146–51. doi: 10.1016/j.ijantimicag.2009.09.029. [DOI] [PubMed] [Google Scholar]
  • [168].Andes D, Marchillo K, Lowther J, Bryskier A, Stamstad T, Conklin R. In vivo pharmacodynamics of HMR 3270, a glucan synthase inhibitor, in a murine candidiasis model. Antimicrob Agents Chemother. 2003 Apr;47(4):1187–92. doi: 10.1128/AAC.47.4.1187-1192.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Najvar LK, Bocanegra R, Wiederhold NP, et al. Therapeutic and prophylactic efficacy of aminocandin (IP960) against disseminated candidiasis in mice. Clin Microbiol Infect. 2008 Jun;14(6):595–600. doi: 10.1111/j.1469-0691.2008.01994.x. [DOI] [PubMed] [Google Scholar]
  • [170].Morikawa H, Tomishima M, Kayakiri N, et al. Synthesis and antifungal activity of ASP9726, a novel echinocandin with potent Aspergillus hyphal growth inhibition. Bioorg Med Chem Lett. 2014 Feb 15;24(4):1172–5. doi: 10.1016/j.bmcl.2013.12.116. [DOI] [PubMed] [Google Scholar]
  • [171].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]
  • [172].Pfaller MA, Messer SA, Coffman S. In vitro susceptibilities of clinical yeast isolates to a new echinocandin derivative, LY303366, and other antifungal agents. Antimicrob Agents Chemother. 1997 Apr;41(4):763–6. doi: 10.1128/aac.41.4.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Johnson E, Espinel-Ingroff A, Szekely A, Hockey H, Troke P. Activity of voriconazole, itraconazole, fluconazole and amphotericin B in vitro against 1763 yeasts from 472 patients in the voriconazole phase III clinical studies. Int J Antimicrob Agents. 2008 Dec;32(6):511–4. doi: 10.1016/j.ijantimicag.2008.05.023. [DOI] [PubMed] [Google Scholar]
  • [174].Lewis RE, Klepser ME, Pfaller MA. In vitro pharmacodynamic characteristics of flucytosine determined by time-kill methods. Diagn Microbiol Infect Dis. 2000 Feb;36(2):101–5. doi: 10.1016/s0732-8893(99)00125-x. [DOI] [PubMed] [Google Scholar]
  • [175].Anaissie EJ, Karyotakis NC, Hachem R, Dignani MC, Rex JH, Paetznick V. Correlation between in vitro and in vivo activity of antifungal agents against Candida species. J Infect Dis. 1994 Aug;170(2):384–9. doi: 10.1093/infdis/170.2.384. [DOI] [PubMed] [Google Scholar]
  • [176].Scalarone GM, Mikami Y, Kurita N, Yazawa K, Miyaji M. The postantifungal effect of 5-fluorocytosine on Candida albicans. J Antimicrob Chemother. 1992 Feb;29(2):129–36. doi: 10.1093/jac/29.2.129. [DOI] [PubMed] [Google Scholar]
  • [177].van der Horst CM, Saag MS, Cloud GA, et al. Treatment of cryptococcal meningitis associated with the acquired immunodeficiency syndrome. National Institute of Allergy and Infectious Diseases Mycoses Study Group and AIDS Clinical Trials Group. N Engl J Med. 1997 Jul 3;337(1):15–21. doi: 10.1056/NEJM199707033370103. [DOI] [PubMed] [Google Scholar]
  • [178].Brouwer AE, van Kan HJ, Johnson E, et al. Oral versus intravenous flucytosine in patients with human immunodeficiency virus-associated cryptococcal meningitis. Antimicrob Agents Chemother. 2007 Mar;51(3):1038–42. doi: 10.1128/AAC.01188-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Schonebeck J, Polak A, Fernex M, Scholer HJ. Pharmacokinetic studies on the oral antimycotic agent 5-fluorocytosine in individuals with normal and impaired kidney function. Chemotherapy. 1973;18(6):321–36. doi: 10.1159/000221279. [DOI] [PubMed] [Google Scholar]
  • [180].Soltani M, Tobin CM, Bowker KE, Sunderland J, MacGowan AP, Lovering AM. 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 Dec;28(6):574–7. doi: 10.1016/j.ijantimicag.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • [181].Francis P, Walsh TJ. Evolving role of flucytosine in immunocompromised patients: new insights into safety, pharmacokinetics, and antifungal therapy. Clin Infect Dis. 1992 Dec;15(6):1003–18. doi: 10.1093/clind/15.6.1003. [DOI] [PubMed] [Google Scholar]
  • [182].Stamm AM, Diasio RB, Dismukes WE, et al. Toxicity of amphotericin B plus flucytosine in 194 patients with cryptococcal meningitis. Am J Med. 1987 Aug;83(2):236–42. doi: 10.1016/0002-9343(87)90691-7. [DOI] [PubMed] [Google Scholar]
  • [183].Andes D, Pascual A, Marchetti O. Antifungal therapeutic drug monitoring: established and emerging indications. Antimicrob Agents Chemother. 2009 Jan;53(1):24–34. doi: 10.1128/AAC.00705-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Mitsuyama J, Nomura N, Hashimoto K, et al. In vitro and in vivo antifungal activities of T-2307, a novel arylamidine. Antimicrob Agents Chemother. 2008 Apr;52(4):1318–24. doi: 10.1128/AAC.01159-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Matsumoto YKK, Kato S, Iwami M, editors. A Novel Antifungal Agent Discovered from Streptomyces sp. Sea Sand. ICAAC; Denver, Colorado. 2013.Sep 10-13, 2013. [Google Scholar]
  • [186].Atkinson AJ, Jr., Bennett JE. Amphotericin B pharmacokinetics in humans. Antimicrob Agents Chemother. 1978 Feb;13(2):271–6. doi: 10.1128/aac.13.2.271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Mehta P, Vinks A, Filipovich A, et al. High-dose weekly AmBisome antifungal prophylaxis in pediatric patients undergoing hematopoietic stem cell transplantation: a pharmacokinetic study. Biol Blood Marrow Transplant. 2006 Feb;12(2):235–40. doi: 10.1016/j.bbmt.2005.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Walsh TJ, Goodman JL, Pappas P, et al. Safety, tolerance, and pharmacokinetics of high-dose liposomal amphotericin B (AmBisome) in patients infected with Aspergillus species and other filamentous fungi: maximum tolerated dose study. Antimicrob Agents Chemother. 2001 Dec;45(12):3487–96. doi: 10.1128/AAC.45.12.3487-3496.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Driscoll TA, Frangoul H, Nemecek ER, et al. Comparison of pharmacokinetics and safety of voriconazole intravenous-to-oral switch in immunocompromised adolescents and healthy adults. Antimicrob Agents Chemother. 2011 Dec;55(12):5780–9. doi: 10.1128/AAC.05010-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Driscoll TA, Yu LC, Frangoul H, et al. Comparison of pharmacokinetics and safety of voriconazole intravenous-to-oral switch in immunocompromised children and healthy adults. Antimicrob Agents Chemother. 2011 Dec;55(12):5770–9. doi: 10.1128/AAC.00531-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Michael C, Bierbach U, Frenzel K, et al. Voriconazole pharmacokinetics and safety in immunocompromised children compared to adult patients. Antimicrob Agents Chemother. 2010 Aug;54(8):3225–32. doi: 10.1128/AAC.01731-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Stone JA, Holland SD, Wickersham PJ, et al. Single- and multiple-dose pharmacokinetics of caspofungin in healthy men. Antimicrob Agents Chemother. 2002 Mar;46(3):739–45. doi: 10.1128/AAC.46.3.739-745.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Weitkamp JH, Poets CF, Sievers R, et al. Candida infection in very low birth-weight infants: outcome and nephrotoxicity of treatment with liposomal amphotericin B (AmBisome) Infection. 1998 Jan-Feb;26(1):11–5. doi: 10.1007/BF02768745. [DOI] [PubMed] [Google Scholar]

RESOURCES