Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 14.
Published in final edited form as: Early Hum Dev. 2011 Feb 1;87(Suppl 1):S61–S65. doi: 10.1016/j.earlhumdev.2011.01.014

Pharmacokinetics of Antifungal Agents in Children

Kevin Watt 1, Daniel K Benjamin Jr 1, Michael Cohen-Wolkowiez 1
PMCID: PMC3418808  NIHMSID: NIHMS270144  PMID: 21277714

Abstract

Invasive fungal infections in immunocompromised children are common and often fatal. The first antifungal agents such as amphotericin B and fluconazole offered effective treatment, but their use was often limited by toxicity and resistance. Numerous new antifungal agents have since been developed and appear to be as effective. Most dosing and safety trials have been done in adults, and extrapolation of this data to children has proven inadequate. We reviewed the literature regarding the pharmacokinetics/pharmacodynamics (PK/PD) and safety of antifungal agents with an emphasis on the newer azoles and echinocandins. From a small but growing number of PK/PD trials, better dosing guidelines have been developed.

Keywords: antifungal agents, pharmacology, pharmacokinetics, neonates, infants, children

INTRODUCTION

Invasive fungal infections in immunocompromised children are common and often fatal. Amphotericin B deoxycholate has been a mainstay of therapy for over 50 years but its use has been limited by toxicity. The introduction of the azole class of antifungal agents offered fewer side effects; however, heavy use of fluconazole in adult patients has resulted in the selection of resistant organisms, though this is not yet widespread in children (13). Since the introduction of fluconazole, there have been a number of new azoles brought to market as well as a new class of antifungal agents, the echinocandins. Amphotericin B deoxycholate has often been the standard against which these new therapies are measured (49). This paper will review our current knowledge of antifungal pharmacokinetics/pharmacodynamics (PK/PD) in children, with an emphasis on the newer azole and echinocandin systemic antifungals.

POLYENES

The polyene macrolides are the oldest class of antifungal agents and are comprised of amphotericin B deoxycholate and its associated lipid-based formulations: amphotericin B lipid complex (ABLC) and liposomal amphotericin B. The polyenes work by binding the sterol component of cell walls which results in increased membrane permeability and cell death. Amphotericin B deoxycholate is active against Candida spp., Aspergillus spp., and zygomycetes.

Amphotericin B deoxycholate

In spite of being approved for use in adults in 1958, there have been relatively few studies of amphotericin B deoxycholate PK/PD in children. Amphotericin B was initially dosed in children at 1.0 mg/kg/day based on clinical experience and a dose escalation study involving 41 children who had undergone bone marrow transplant (10). Several subsequent PK/PD studies in infants and children, including premature infants, found extreme inter-subject variability for the half-life, volume of distribution, and clearance but overall supported dosing with 1.0–1.5 mg/kg/day (1114). A more recent population PK study of amphotericin B in 57 children ages 9 mos to 16 years with malignancy developed a model that suggested younger, lighter infants may be under-dosed at 1 mg/kg/day while older, heavier children may be overdosed at that same dose (15). Penetration into the CSF seems to be age dependent. In adults, cerebrospinal fluid (CSF) values are only 2–4% of serum concentrations (16). In contrast, in a small series of premature infants born at 27.4 (± 5) weeks gestational age (n=5), amphotericin B concentrations in CSF were 40–90% of simultaneously collected serum concentrations (11).

Lipid-based amphotericin B preparations

Lipid formulations of amphotericin B generally have a slower onset of action and are less active than amphotericin B in time-kill studies, presumably due to the required disassociation of free amphotericin B from the lipid vehicle (17). The different PK and toxicities of the lipid formulations are reflected in the dosing recommendations: amphotericin B lipid complex (ABLC) is recommended at 3–5mg/kg/day (18) and liposomal amphotericin B at 1–5 mg/kg/day (19, 20). A multicenter maximum tolerated dose study of L-amphotericin B using doses from 7.5 to 15 mg/kg/day found a non-linear plasma PK profile with a maximal concentration at 10 mg/kg/day and no demonstrable dose-limiting nephrotoxicity or infusionrelated toxicity (21).

NUCLEOSIDE ANALOGS

5-Flucytosine

5-Flucytosine (5-FC) is an antimetabolite drug with excellent activity against Candida spp, Cryptococcus neoformansand Chromomycosis spp (22, 23). Resistance develops rapidly to 5-FC mono-therapy, so clinicians have reserved it for combination therapy to augment other antifungals. 5-FC has been shown to enhance the antifungal activity of amphotericin B, especially in anatomical sites where amphotericin B penetration is often suboptimal such as CSF, heart valves, and the vitreal body (24). 5-FC is only available as an oral formulation in the United States, and is given 150 mg/kg/day in four divided doses.

5-FC has a narrow therapeutic window with elevated serum concentrations resulting in hepatic injury (25, 26), bone marrow suppression (27), and gastrointestinal intolerance (28). Because of this routine therapeutic drug monitoring is standard of care throughout the treatment course as amphotericin-related renal insufficiency may develop over time and result in decreased clearance of 5-FC. The use of 5-FC in premature neonates is discouraged. A study evaluating risk factors and mortality rates of neonatal candidiasis among extremely premature infants showed that infants with Candida meningitis who received amphotericin B in combination with 5-FC had significantly longer time to sterilization of the CSF compared to those receiving amphotericin B monotherapy (median of 17.5 versus 6 days, respectively) (29).

TRIAZOLES

The triazoles work by inhibiting the enzyme responsible for converting lanosterol to ergosterol, a key component of fungal cell membranes, resulting in cell lysis and death (8, 30, 31).

Fluconazole

Fluconazole is available in both intravenous (IV) and oral formulations with rapid absorption from GI tract with high oral availability (~92% bioavailability compared with the IV form). It has low protein binding and excellent penetration into the CSF with concentrations ~80% of those found in the serum. Penetration into other body tissues such as joint spaces, saliva, vaginal secretions, and vitreous humor of the eye also approximate or exceed levels found in the serum. Fluconazole is eliminated primarily through the kidneys and excreted largely unchanged resulting in urine concentrations 10–20 times higher than those found in blood (32, 33).

Scaling of the corresponding adult dosage of fluconazole on a body weight basis is inappropriate for children. A review of 5 separate fluconazole PK studies in 113 children, including 12 premature neonates (34) showed that fluconazole clearance is generally more rapid in children than adults, with a mean plasma half-life of approximately 20 hours compared to approximately 30 hours in adults. Therefore, to achieve comparable exposure in children, the daily fluconazole dose needs to be essentially doubled. Correct pediatric fluconazole doses should be proportionately higher than adult doses, generally 12 mg/kg/day.

Neonates require approximately 5 days to reach steady-state (35), and maintenance fluconazole doses of 12 mg/kg/day are necessary to achieve exposures similar to older children and adults (36). Thus, a loading dose of 25 mg/kg to achieve steady-state concentrations sooner than the traditional dosing scheme may be appropriate. Data from a phase 1 trial investigating a fluconazole loading dose in 12 critically ill children showed that the therapeutic target (AUC0–24 = 400 ug*h/mL) was reached with the loading dose in 72% of subjects with no related adverse events (37).

Voriconazole

Voriconazole is a synthetic derivative of fluconazole and has both fungicidal and fungistatic activity against Aspergillus (3840). It is extensively metabolized by the liver with less than 5% excreted unchanged in the urine. Voriconazole is 58% protein bound, has a large volume of distribution and penetrates well into the CSF.

Voriconazole serum concentrations demonstrate wide inter-patient variability. There are several factors that are thought to contribute to this variability including nonlinear PK of voriconazole and genetic polymorphisms of CYP2C19. Studies in both healthy volunteers (4143) and critically ill adults (44, 45) have shown nonlinear PK that is thought to be due to saturable first-pass metabolism and decreased systemic clearance. Individuals with poor metabolizing phenotypes have a roughly two-fold higher drug exposure than heterozygous extensive metabolizers and four-fold higher exposure than homozygous extensive metabolizes (46, 47).

In contrast to adults, the PK of voriconazole in children is linear at lower doses and patient weight rather than age seems to play a larger role in determining drug exposure, making extrapolation of adult data problematic. Based on PK/PD analysis in a prospective multicenter study of 39 immunocompromised children age 2–11 y, a dose of 4 mg/kg intravenously was found to be equivalent to the recommended dose of 3 mg/kg in adults (48). Similar to adults, drug elimination correlated with CYP2C19 phenotype, but children were found to have higher elimination rates than those found in adults, which at least partially explains the linear kinetics at lower doses. In a population PK analysis of 3 open label pediatric studies involving 82 children 2–11 y of age, an intravenous dose of 7 mg/kg or 200 mg orally twice daily was found to be equivalent to adult dosing (49).

Because of the nonlinear PK observed in adults and high inter-patient variability in serum concentration, voriconazole therapeutic drug monitoring is recommended. Therapeutic failures occur with voriconazole plasma levels <1 µg/ml and toxicity rises when levels exceed 5–6 µg/ml (5054).

Studies in children are more limited, but retrospective studies show that children also experience high inter-patient variability in voriconazole concentrations (55) and that mortality decreases when voriconazole serum trough concentrations >1µg/ml are achieved (56). Case studies in infants suggest that even higher voriconazole doses are needed to achieve this target trough concentration, underscoring the need for therapeutic drug monitoring (5759). Even though the data acquired thus far supports the use of therapeutic drug monitoring, prospective studies are lacking in children and need to be conducted to answer this question.

Posaconazole

Posaconazole is a second-generation triazole antifungal agent available as a suspension for oral administration. The antimicrobial spectrum of posaconazole is similar to voriconazole, but with additional activity against zygomycetes. Experience with posaconazole in children is very limited. In adults, dose-proportional increases in plasma exposure (AUC) to posaconazole were observed following single oral doses from 50 mg to 800 mg and following multi-dose administration from 50 mg to 400 mg twice daily. Steady-state plasma concentrations are attained at 7 to 10 days following multi-dose administration (60). Posaconazole is predominantly eliminated in the feces with renal clearance playing a minor role. Therefore, no dose adjustment is necessary in mild to moderate renal insufficiency. Posaconazole is fungicidal in vitro with likely time-dependent killing (61).

Ravuconazole

Ravuconazole is structurally more similar to fluconazole and voriconazole, containing a thiazole instead of a second triazole. It is often fungicidal (62, 63), has 47–74% bioavailability with linear PK, and a long half-life of approximately 100 hours (64). The drug is well-absorbed following oral administration, and its absorption is enhanced by food (63). Penetration of ravuconazole into healthy rat tissue showed that concentration of drug in the lungs was 2–6 times higher than the corresponding blood concentration (65). Ravuconazole has not been approved by the FDA. Ravuconazole was well-tolerated in healthy human subjects in single (64) and multiple doses (66). To our knowledge, there are no clinical trials of ravuconazole in children.

ECHINOCANDINS

There are three drugs in this class: caspofungin, micafungin, and anidulafungin, and they all work by inhibiting 1,3 β-D-glucan synthase, an enzyme important in fungal cell wall biosynthesis (67, 68). The echinocandins are fungicidal against Candida spp and fungistatic against Aspergillus spp, though only caspofungin carries an FDA indication for Aspergillus (68, 69). Additionally, caspofungin is the only echinocandin approved by the FDA for use in children (age >3 mos) (70). Because the echinocandins are not hepatically metabolized they have the potential for fewer drug interactions as a class than the other antifungals (71).

Caspofungin

There have been three PK/PD studies of caspofungin in infants and children that have demonstrated significant differences in PK/PD parameters than those described in adults (68, 72, 73). Extrapolating the adult dose of 50 mg/day to a per kilo dose in children ages 2–17 y (1 mg/kg/day) resulted in significantly lower AUC0–24 (46%) (68). Dosing based on body surface area, however, resulted in comparable AUC0–24. Infants and children ages 3 mos – 17y required 50 mg/m2/day (68, 72), while infants <3 mos achieved therapeutic serum concentrations with 25 mg/m2/day (73). The appropriate dosage of caspofungin in the nursery is not known.

Micafungin

Though it does not carry an FDA label for use in children, micafungin has been almost as well studied in pediatric populations as caspofungin. The first PK study of micafungin in children was a phase 1, open-label dose escalation study in 77 neutropenic children ages 2–17 y who were given doses ranging from 0.5–4 mg/kg/day (74). The authors noted linear PK with increased clearance in the 2–8 y cohort resulting in dosing recommendations of 3–4.5 mg/kg/day for 2–8 y and 2–3 mg/kg/day for those 9–17 y to achieve concentrations comparable with standard adult dosing. Importantly, the investigators found no dose-related side effects, which is in line with previous adult studies (74). In a smaller study of 19 children ages 8 mos to 15 y with confirmed invasive fungal infection, Tabata et al (75) also noted linear PK but without age-associated differences in clearance, though this may have been due to the smaller size of the study. These investigators recommended dosing of 3 mg/kg/day.

Candida is an important pathogen in premature neonates, and there is evidence in neonates, especially those with Candida meningoencephalitis, that higher doses of micafungin may be required (76). Heresi et al (77) conducted a phase 1 multi-center sequential dose study (0.75 mg/kg, 1.5 mg/kg, 3.0 mg/kg) of micafungin PK in 18 premature neonates (mean gestational age 26.4 weeks; mean birth weight 1497g) with suspected or confirmed invasive fungal infection. These investigators noted a larger volume of distribution, as might be expected in neonates with larger extracellular fluid compartments, as well as an unexpected increase in clearance compared with children (1.7 fold for ages 2–8 y) and adolescents (2.6 fold for ages 9–17 y). All doses were well tolerated. More recently, Smith et al (78), investigated higher dose micafungin (15 mg/kg/day for 5 days) in 12 preterm neonates (mean gestational age 27 weeks, mean birth weight 775g) with suspected systemic infection. This higher dose was well tolerated in all subjects. The resultant PK parameters were comparable to those in adults in a maximum tolerated dose study who were dosed at 5 mg/kg (79). Benjamin et al studied doses of 7 and 10 mg/kg/day and observed exposures similar to those required to clear meningoencephalitis in animal models. Based on these results, we recommend dosing in neonates of 10 mg/kg/day.

Anidulafungin

Anidulafungin was approved by the FDA for use in adults in 2006. Like caspofungin, anidulafungin seems to concentrate in the liver, lung, and kidney with poor CSF penetration (80). PK studies in both healthy adults and adults with invasive fungal infections showed that subjects achieved therapeutic concentrations using a 200 mg loading dose on day 1 followed by a daily dose of 100 mg/day starting on day 2 (81, 82). Benjamin et al (83) enrolled 24 neutropenic children ages 2–16 y in a multi-center dose escalation PK study. Children in the low-dose group (n=12) were loaded with 1.5 mg/kg and started on maintenance of 0.75 mg/kg/day and those in the high dose group (n=12) were loaded with 3 mg/kg followed by a maintenance dose of 1.5 mg/kg/day. Based on measured PK parameters, the low- and high-dose regimens corresponded to 50 mg/day and 100 mg/day dosing in adults respectively. Preliminary data from a phase 1 PK study of anidulafungin in neonates and infants (n=14) showed that anidulafungin maintenance doses of 1.5 mg/kg/day resulted in exposures similar to older children and adults receiving comparable weight-based dosing (84). The optimal dose in neonates, who are at greatest risk of Candida meningoencephalitis, remains to be determined.

Conclusion

With the rapid development of new antifungal agents, clinicians have more options in the treatment of invasive fungal infections. Most of the PK/PD studies performed are in adults, but as demonstrated above, extrapolation of adult data to children has proved inadequate for many of these agents. This highlights the need to undertake dedicated PK/PD studies of antifungal agents in children.

Table 1.

Pediatric Dosing of Antifungal Agents

Neonates
(0–30 d)
Infants
(31 d-2 y)
Children
(2 y-17 y)
Polyenes
Amphotericin B 0.5–1 mg/kg/day 0.5–1 mg/kg/day 0.5–1 mg/kg/day
ABLC Unk Unk 3–5 mg/kg/day
L-Amphotericin B Unk Unk 1–5 mg/kg/day
Nucleoside
analogs
5-Flucytosine 25–100 mg/kg/day
div every 12–24h
50–150
mg/kg/day div
every 6h
50–150
mg/kg/day div
every 6h
Triazoles
Fluconazole 6–12 mg/kg every
72ha
12 mg/kg/day 6–12 mg/kg/day
6–12 mg/kg every
48hb
6–12 mg/kg/dayc
Voriconazole NR Unk 14 mg/kg/day
div every 12h
Posaconazole Unk Unk 800 mg/day div
every 6–12hd
Ravuconazole Unk Unk Unk
Echinocandins
Caspofungin 25 mg/m2/day 50 mg/m2/day 70 mg/m2 load
50 mg/m2/day
Micafungin 10 mg/kg/day 1–4 mg/kg/day 1–4 mg/kg/day
Anidulafungin 1.5 mg/kg/day 1.5 mg/kg/day 1.5 mg/kg/day
a

≤ 29wks gestation, post-natal age 0–14 d

b

≤29wks gestation, post-natal age >14 d or 30–36 wks gestation, post-natal age 0–14 d

c

≥30wks gestation, post-natal age >14 d or >36 wks gestation

d

Dosing unknown for children <13 years of age

NR – not recommended

Unk – appropriate dosing unknown

Footnotes

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

CONFLICT OF INTEREST

Dr. Benjamin receives support from the United States Government for his work in pediatric and neonatal clinical pharmacology (1R01HD057956-02, 1R01FD003519-01, 1U10-HD45962-06, 1K24HD058735-01, and is the Principal Investigator of the Pediatric Trials Network, Government Contract HHSN275201000002I); the non-profit organization Thrasher Research Foundation for his work in neonatal candidiasis (http://www.thrasherresearch.org); and from industry for neonatal and pediatric drug development (http://www.dcri.duke.edu/research/coi.jsp).

Dr. Cohen-Wolkowiez receives support from NICHD for his work in pediatric and neonatal clinical pharmacology (1K23HD064814-01

Dr. Watt receives support from the United States Government for his work in pediatric research (5T32HD043029-09

REFERENCES

  • 1.Boschman CR, Bodnar UR, Tornatore MA, Obias AA, Noskin GA, Englund K, et al. Thirteen-year evolution of azole resistance in yeast isolates and prevalence of resistant strains carried by cancer patients at a large medical center. Antimicrob Agents Chemother. 1998 Apr;42(4):734–738. doi: 10.1128/aac.42.4.734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Magill SS, Swoboda SM, Shields CE, Colantuoni EA, Fothergill AW, Merz WG, et al. The epidemiology of Candida colonization and invasive candidiasis in a surgical intensive care unit where fluconazole prophylaxis is utilized: follow-up to a randomized clinical trial. Ann Surg. 2009 Apr;249(4):657–665. doi: 10.1097/SLA.0b013e31819ed914. [DOI] [PubMed] [Google Scholar]
  • 3.Nguyen MH, Peacock JE, Jr, Morris AJ, Tanner DC, Nguyen ML, Snydman DR, et al. The changing face of candidemia: emergence of non-Candida albicans species and antifungal resistance. Am J Med. 1996 Jun;100(6):617–623. doi: 10.1016/s0002-9343(95)00010-0. [DOI] [PubMed] [Google Scholar]
  • 4.Krause DS, Simjee AE, van Rensburg C, Viljoen J, Walsh TJ, Goldstein BP, et al. A randomized, double-blind trial of anidulafungin versus fluconazole for the treatment of esophageal candidiasis. Clin Infect Dis. 2004 Sep 15;39(6):770–775. doi: 10.1086/423378. [DOI] [PubMed] [Google Scholar]
  • 5.Kullberg BJ, Sobel JD, Ruhnke M, Pappas PG, Viscoli C, Rex JH, et al. Voriconazole versus a regimen of amphotericin B followed by fluconazole for candidaemia in nonneutropenic patients: a randomised non-inferiority trial. Lancet. 2005 Oct 22–28;366(9495):1435–1442. doi: 10.1016/S0140-6736(05)67490-9. [DOI] [PubMed] [Google Scholar]
  • 6.Mora-Duarte J, Betts R, Rotstein C, Colombo AL, Thompson-Moya L, Smietana J, et al. Comparison of caspofungin and amphotericin B for invasive candidiasis. N Engl J Med. 2002 Dec 19;347(25):2020–2029. doi: 10.1056/NEJMoa021585. [DOI] [PubMed] [Google Scholar]
  • 7.Reboli AC, Rotstein C, Pappas PG, Chapman SW, Kett DH, Kumar D, et al. Anidulafungin versus fluconazole for invasive candidiasis. N Engl J Med. 2007 Jun 14;356(24):2472–2482. doi: 10.1056/NEJMoa066906. [DOI] [PubMed] [Google Scholar]
  • 8.Rex JH, Bennett JE, Sugar AM, Pappas PG, van der Horst CM, Edwards JE, et al. A randomized trial comparing fluconazole with amphotericin B for the treatment of candidemia in patients without neutropenia. Candidemia Study Group and the National Institute. N Engl J Med. 1994 Nov 17;331(20):1325–1330. doi: 10.1056/NEJM199411173312001. [DOI] [PubMed] [Google Scholar]
  • 9.Rex JH, Pappas PG, Karchmer AW, Sobel J, Edwards JE, Hadley S, et al. A randomized and blinded multicenter trial of high-dose fluconazole plus placebo versus fluconazole plus amphotericin B as therapy for candidemia and its consequences in nonneutropenic subjects. Clin Infect Dis. 2003 May 15;36(10):1221–1228. doi: 10.1086/374850. [DOI] [PubMed] [Google Scholar]
  • 10.Emminger W, Lang HR, Emminger-Schmidmeier W, Peters C, Gadner H. Amphotericin B serum levels in pediatric bone marrow transplant recipients. Bone Marrow Transplant. 1991 Feb;7(2):95–99. [PubMed] [Google Scholar]
  • 11.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):791–797. doi: 10.1016/s0022-3476(05)82674-5. [DOI] [PubMed] [Google Scholar]
  • 12.Benson JM, Nahata MC. Pharmacokinetics of amphotericin B in children. Antimicrob Agents Chemother. 1989 Nov;33(11):1989–1993. doi: 10.1128/aac.33.11.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Koren G, Lau A, Klein J, Golas C, Bologa-Campeanu M, Soldin S, et al. Pharmacokinetics and adverse effects of amphotericin B in infants and children. J Pediatr. 1988 Sep;113(3):559–563. doi: 10.1016/s0022-3476(88)80653-x. [DOI] [PubMed] [Google Scholar]
  • 14.Starke JR, Mason EO, Jr, Kramer WG, Kaplan SL. Pharmacokinetics of amphotericin B in infants and children. J Infect Dis. 1987 Apr;155(4):766–774. doi: 10.1093/infdis/155.4.766. [DOI] [PubMed] [Google Scholar]
  • 15.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):671–680. doi: 10.1046/j.1365-2125.2001.01496.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Luna B, Drew RH, Perfect JR. Agents for treatment of invasive fungal infections. Otolaryngologic Clinics of North America. 2000;33:277–299. doi: 10.1016/s0030-6665(00)80005-5. [DOI] [PubMed] [Google Scholar]
  • 17.Ralph ED, Khazindar AM, Barber KR, Grant CWM. 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. Antimicrobial Agents and Chemotherapy. 1991;35:188–191. doi: 10.1128/aac.35.1.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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):5092–5098. doi: 10.1128/AAC.49.12.5092-5098.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hong Y, Shaw PJ, Nath CE, Yadav SP, Stephen KR, Earl JW, et al. Population pharmacokinetics of liposomal amphotericin B in pediatric patients with malignant diseases. Antimicrob Agents Chemother. 2006 Mar;50(3):935–942. doi: 10.1128/AAC.50.3.935-942.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kotwani RN, Gokhale PC, Bodhe PV, Kirodian BG, Kshirsagar NA, Pandya SK. A comparative study of plasma concentrations of liposomal amphotericin B (L-AMP-LRC-1) in adults, children and neonates. Int J Pharm. 2002 May 15;238(1–2):11–15. doi: 10.1016/s0378-5173(02)00066-2. [DOI] [PubMed] [Google Scholar]
  • 21.Walsh TJ, Goodman JL, Pappas P, Bekersky I, Buell DN, Roden M, et al. Safety, tolerance, and pharmacokinetics of high-dose liposomal amphotericin B (AmBisome) in patients infected with Aspergillus species and other filamentous fungi: maximum tolerate dose study. Antimicrobial Agents and Chemotherapy. 2001;45:3487–3496. doi: 10.1128/AAC.45.12.3487-3496.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bennet JE. Flucytosine. Ann Intern Med. 1977 Mar;86(3):319–321. doi: 10.7326/0003-4819-86-3-319. [DOI] [PubMed] [Google Scholar]
  • 23.Mauceri AA, Cullen SI, Vandevelde AG, Johnson JE., 3rd Flucytosine. An effective oral treatment for chromomycosis. Arch Dermatol. 1974 Jun;109(6):873–876. doi: 10.1001/archderm.109.6.873. [DOI] [PubMed] [Google Scholar]
  • 24.Denning DW, Stevens DA. Antifungal and surgical treatment of invasive aspergillosis: review of 2,121 published cases. Reviews of Infectious Diseases. 1990;12:1147–1200. doi: 10.1093/clinids/12.6.1147. [DOI] [PubMed] [Google Scholar]
  • 25.Song JC, Deresinski S. Hepatotoxicity of antifungal agents. Curr Opin Investig Drugs. 2005 Feb;6(2):170–177. [PubMed] [Google Scholar]
  • 26.Stamm AM, Diasio RB, Dismukes WE, Shadomy S, Cloud GA, Bowles CA, et al. Toxicity of amphotericin B plus flucytosine in 194 patients with cryptococcal meningitis. Am J Med. 1987 Aug;83(2):236–242. doi: 10.1016/0002-9343(87)90691-7. [DOI] [PubMed] [Google Scholar]
  • 27.Benson JM, Nahata MC. Clinical use of systemic antifungal agents. Clin Pharm. 1988 Jun;7(6):424–438. [PubMed] [Google Scholar]
  • 28.Patel R. Antifungal agents. Part I. Amphotericin B preparations and flucytosine. Mayo Clin Proc. 1998 Dec;73(12):1205–1225. doi: 10.4065/73.12.1205. [DOI] [PubMed] [Google Scholar]
  • 29.Benjamin DKJ, Stoll BJ, Fanaroff AA, McDonald SA, Oh W, R D, Duara S, et al. Neonatal candidiasis among extremely low birth weight infants: risk factors, mortality rates, and neurodevelopmental outcomes at 18 to 22 months. Pediatrics. 2006;117:84–92. doi: 10.1542/peds.2004-2292. [DOI] [PubMed] [Google Scholar]
  • 30.Anaissie EJ, Darouiche RO, Abi-Said D, Uzun O, Mera J, Gentry LO, et al. Management of invasive candidal infections: results of a prospective, randomized, multicenter study of fluconazole versus amphotericin B and review of the literature. Clin Infect Dis. 1996 Nov;23(5):964–972. doi: 10.1093/clinids/23.5.964. [DOI] [PubMed] [Google Scholar]
  • 31.Phillips JR, Karlowicz MG. Prevalence of Candida species in hospital-acquired urinary tract infections in a neonatal intensive care unit. Pediatric Infectious Disease Journal. 1997 Feb;16(2):190–194. doi: 10.1097/00006454-199702000-00005. [DOI] [PubMed] [Google Scholar]
  • 32.Debruyne D. Clinical pharmacokinetics of fluconazole in superficial and systemic mycoses. Clin Pharmacokinet. 1997 Jul;33(1):52–77. doi: 10.2165/00003088-199733010-00005. [DOI] [PubMed] [Google Scholar]
  • 33.Wildfeuer A, Laufen H, Schmalreck AF, Yeates RA, Zimmermann T. Fluconazole: comparison of pharmacokinetics, therapy and in vitro susceptibility. Mycoses. 1997 Nov;40(7–8):259–265. doi: 10.1111/j.1439-0507.1997.tb00230.x. [DOI] [PubMed] [Google Scholar]
  • 34.Brammer KW, Coates PE. Pharmacokinetics of fluconazole in pediatric patients. Eur J Clin Microbiol Infect Dis. 1994 Apr;13(4):325–329. doi: 10.1007/BF01974613. [DOI] [PubMed] [Google Scholar]
  • 35.Debruyne D. Clinical pharmacokinetics of fluconazole in superficial and systemic mycoses. Clinical Pharmacokinetics. 1997;33:52–77. doi: 10.2165/00003088-199733010-00005. [DOI] [PubMed] [Google Scholar]
  • 36.Wade KC, Wu D, Kaufman DA, Ward RM, Benjamin DK, Jr, Sullivan JE, et al. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother. 2008 Nov;52(11):4043–4049. doi: 10.1128/AAC.00569-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Piper LSP, Hornik CP, Cheifetz IM, Barrett J, Hope W, Moorthy G, Wade K, Cohen-Wolkowiez M, Benjamin DK., Jr Fluconazole Loading Dose Pharmacokinetics and Safety in Infants. Society for Pediatric Research Conference; Vancouver, Canada. May 2010; 2010. [Abstract] [Google Scholar]
  • 38.Sabo JA, Abdel-Rahman SM. Voriconazole: a new triazole antifungal. Annals of Pharmacotherapy. 2000;34:1032–1043. doi: 10.1345/aph.19237. [DOI] [PubMed] [Google Scholar]
  • 39.Manavathu EK, Cutright JL, Chandrasekar PH. In vitro susceptibility of itraconazoleresistant isolates of Aspergillus fumigatus to voriconazole. Clinical Microbiology and Infection. 1997;3(Suppl 2):81. [Google Scholar]
  • 40.Johnson EM, Szekely A, Warnock DW. In-vitro activity of voriconazole, itraconazole and amphotericin B against filamentous fungi. Journal of Antimicrobial Chemotherapy. 1998;42:741–745. doi: 10.1093/jac/42.6.741. [DOI] [PubMed] [Google Scholar]
  • 41.Purkins L, Wood N, Ghahramani P, Greenhalgh K, Allen MJ, Kleinermans D. Pharmacokinetics and safety of voriconazole following intravenous- to oral-dose escalation regimens. Antimicrob Agents Chemother. 2002 Aug;46(8):2546–2553. doi: 10.1128/AAC.46.8.2546-2553.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Purkins L, Wood N, Greenhalgh K, Allen MJ, Oliver SD. Voriconazole, a novel wide-spectrum triazole: oral pharmacokinetics and safety. Br J Clin Pharmacol. 2003 Dec;56(Suppl 1):10–16. doi: 10.1046/j.1365-2125.2003.01993.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Patterson BaPC., editor. UK-109496, a novel, wide spectrum triazole derivative for the treatment of fungal infections: pharmacokinetics in man [abstract F78]. Program and abstracts of the 35th Interscience Conference on Antimicrobial Agents and Chemotherapy; Washington, DC: American Society for Microbiology; 1995. [Google Scholar]
  • 44.Myrianthefs P, Markantonis SL, Evaggelopoulou P, Despotelis S, Evodia E, Panidis D, et al. Monitoring plasma voriconazole levels following intravenous administration in critically ill patients: an observational study. Int J Antimicrob Agents. May;35(5):468–472. doi: 10.1016/j.ijantimicag.2009.12.021. [DOI] [PubMed] [Google Scholar]
  • 45.Lazarus HM, Blumer JL, Yanovich S, Schlamm H, Romero A. Safety and pharmacokinetics of oral voriconazole in patients at risk of fungal infection: a dose escalation study. J Clin Pharmacol. 2002 Apr;42(4):395–402. [PubMed] [Google Scholar]
  • 46.Hyland R, Jones BC, Smith DA. Identification of the cytochrome P450 enzymes involved in the N-oxidation of voriconazole. Drug Metab Dispos. 2003 May;31(5):540–547. doi: 10.1124/dmd.31.5.540. [DOI] [PubMed] [Google Scholar]
  • 47.Ikeda Y, Umemura K, Kondo K, Sekiguchi K, Miyoshi S, Nakashima M. Pharmacokinetics of voriconazole and cytochrome P450 2C19 genetic status. Clin Pharmacol Ther. 2004 Jun;75(6):587–588. doi: 10.1016/j.clpt.2004.02.002. [DOI] [PubMed] [Google Scholar]
  • 48.Walsh TJ, Karlsson MO, Driscoll T, Arguedas AG, Adamson P, Saez-Llorens X, et al. Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration. Antimicrob Agents Chemother. 2004 Jun;48(6):2166–2172. doi: 10.1128/AAC.48.6.2166-2172.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Karlsson MO, Lutsar I, Milligan PA. Population pharmacokinetic analysis of voriconazole plasma concentration data from pediatric studies. Antimicrob Agents Chemother. 2009 Mar;53(3):935–944. doi: 10.1128/AAC.00751-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Boyd AE, Modi S, Howard SJ, Moore CB, Keevil BG, Denning DW. Adverse reactions to voriconazole. Clin Infect Dis. 2004 Oct 15;39(8):1241–1244. doi: 10.1086/424662. [DOI] [PubMed] [Google Scholar]
  • 51.Denning DW, Ribaud P, Milpied N, Caillot D, Herbrecht R, Thiel E, et al. Efficacy and safety of voriconazole in the treatment of acute invasive aspergillosis. Clin Infect Dis. 2002 Mar 1;34(5):563–571. doi: 10.1086/324620. [DOI] [PubMed] [Google Scholar]
  • 52.Imhof A, Schaer DJ, Schanz U, Schwarz U. Neurological adverse events to voriconazole: evidence for therapeutic drug monitoring. Swiss Med Wkly. 2006 Nov 11;136(45–46):739–742. doi: 10.4414/smw.2006.11547. [DOI] [PubMed] [Google Scholar]
  • 53.Smith J, Safdar N, Knasinski V, Simmons W, Bhavnani SM, Ambrose PG, et al. Voriconazole therapeutic drug monitoring. Antimicrob Agents Chemother. 2006 Apr;50(4):1570–1572. doi: 10.1128/AAC.50.4.1570-1572.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Trifilio S, Pennick G, Pi J, Zook J, Golf M, Kaniecki K, et al. Monitoring plasma voriconazole levels may be necessary to avoid subtherapeutic levels in hematopoietic stem cell transplant recipients. Cancer. 2007 Apr 15;109(8):1532–1535. doi: 10.1002/cncr.22568. [DOI] [PubMed] [Google Scholar]
  • 55.Pasqualotto AC, Shah M, Wynn R, Denning DW. Voriconazole plasma monitoring. Arch Dis Child. 2008 Jul;93(7):578–581. doi: 10.1136/adc.2007.118844. [DOI] [PubMed] [Google Scholar]
  • 56.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]
  • 57.Muldrew KM, Maples HD, Stowe CD, Jacobs RF. Intravenous voriconazole therapy in a preterm infant. Pharmacotherapy. 2005 Jun;25(6):893–898. doi: 10.1592/phco.2005.25.6.893. [DOI] [PubMed] [Google Scholar]
  • 58.Santos RP, Sanchez PJ, Mejias A, Benjamin DK, Jr, Walsh TJ, Patel S, et al. Successful medical treatment of cutaneous aspergillosis in a premature infant using liposomal amphotericin B, voriconazole and micafungin. Pediatr Infect Dis J. 2007 Apr;26(4):364–366. doi: 10.1097/01.inf.0000258698.98370.89. [DOI] [PubMed] [Google Scholar]
  • 59.Maples HD, Stowe CD, Saccente SL, Jacobs RF. Voriconazole serum concentrations in an infant treated for Trichosporon beigelii infection. Pediatr Infect Dis. J. 2003 Nov;22(11):1022–1024. doi: 10.1097/01.inf.0000095167.38306.76. [DOI] [PubMed] [Google Scholar]
  • 60.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):2788–2795. doi: 10.1128/AAC.47.9.2788-2795.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Groll AH, Mickiene D, Petraitis V, Petraitiene R, Ibrahim KH, Piscitelli SC, et al. Compartmental pharmacokinetics and tissue distribution of the antifungal echinocandin lipopeptide micafungin (FK463) in rabbits. Antimicrob Agents Chemother. 2001 Dec;45(12):3322–3327. doi: 10.1128/AAC.45.12.3322-3327.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Moore CB, Walls CM, Denning DW. In vitro activity of the new triazole BMS-207147 against Aspergillus species in comparison with itraconazole and amphotericin B. Antimicrobial Agents and Chemotherapy. 2000;44:441–443. doi: 10.1128/aac.44.2.441-443.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Arikan S, Rex JH. Ravuconazole. Current Opinion in Investigational Drugs. 2002;3:555–561. [PubMed] [Google Scholar]
  • 64.Olsen SJ, Mummaneni V, Rolan P, Norton J, Grasela DM, editors. Ravuconazole single ascending oral dose study in healthy subjects. Program and abstracts of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; Toronto, Ontario: Washington, DC. 2000. 2000 *****; [Google Scholar]
  • 65.Mikamo H, Yin XH, Hayasaki Y, Shimamura Y, Uesugi K, Fukayama N, et al. Penetration of ravuconazole, a new triazole antifungal, into rat tissues. Chemotherapy. 2002;48:7–9. doi: 10.1159/000048580. [DOI] [PubMed] [Google Scholar]
  • 66.Faix RG, Polley TZ, Grasela TH. A randomized, controlled trial of parenteral clindamycin in neonatal necrotizing enterocolitis. J Pediatr. 1988 Feb;112(2):271–277. doi: 10.1016/s0022-3476(88)80069-6. [DOI] [PubMed] [Google Scholar]
  • 67.Ernst EJ. Investigational antifungal agents. Pharmacotherapy. 2001 Aug;21(8 Pt 2):165S–174S. doi: 10.1592/phco.21.12.165s.34509. [DOI] [PubMed] [Google Scholar]
  • 68.Walsh TJ, Viviani MA, Arathoon E, Chiou C, Ghannoum M, Groll AH, et al. New targets and delivery systems for antifungal therapy. Med Mycol. 2000;38(Suppl 1):335–347. [PubMed] [Google Scholar]
  • 69.Groll AH, Piscitelli SC, Walsh TJ. Antifungal pharmacodynamics: concentration-effect relationships in vitro and in vivo. Pharmacotherapy. 2001 Aug;21(8 Pt 2):133S–148S. doi: 10.1592/phco.21.12.133s.34507. [DOI] [PubMed] [Google Scholar]
  • 70.Merck Caspofungin Product Label. Administration FaD, editor. 2010 [Google Scholar]
  • 71.Lipp HP. Antifungal agents--clinical pharmacokinetics and drug interactions. Mycoses. 2008;51(Suppl 1):7–18. doi: 10.1111/j.1439-0507.2008.01523.x. [DOI] [PubMed] [Google Scholar]
  • 72.Neely M, Jafri HS, Seibel N, Knapp K, Adamson PC, Bradshaw SK, et al. Pharmacokinetics and safety of caspofungin in older infants and toddlers. Antimicrob Agents Chemother. 2009 Apr;53(4):1450–1456. doi: 10.1128/AAC.01027-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Saez-Llorens X, Macias M, Maiya P, Pineros J, Jafri HS, Chatterjee A, et al. Pharmacokinetics and safety of caspofungin in neonates and infants less than 3 months of age. Antimicrob Agents Chemother. 2009 Mar;53(3):869–875. doi: 10.1128/AAC.00868-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Seibel NL, Schwartz C, Arrieta A, Flynn P, Shad A, Albano E, et al. Safety, tolerability, and pharmacokinetics of Micafungin (FK463) in febrile neutropenic pediatric patients. Antimicrob Agents Chemother. 2005 Aug;49(8):3317–3324. doi: 10.1128/AAC.49.8.3317-3324.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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):1706–1711. doi: 10.1248/bpb.29.1706. [DOI] [PubMed] [Google Scholar]
  • 76.Hope WW, Mickiene D, Petraitis V, Petraitiene R, Kelaher AM, Hughes JE, 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):163–171. doi: 10.1086/524063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Heresi GP, Gerstmann DR, Reed MD, van den Anker JN, Blumer JL, Kovanda L, et al. The pharmacokinetics and safety of micafungin, a novel echinocandin, in premature infants. Pediatr Infect Dis J. 2006 Dec;25(12):1110–1115. doi: 10.1097/01.inf.0000245103.07614.e1. [DOI] [PubMed] [Google Scholar]
  • 78.Smith PB, Walsh TJ, Hope W, Arrieta A, Takada A, Kovanda LL, et al. Pharmacokinetics of an elevated dosage of micafungin in premature neonates. Pediatr Infect Dis J. 2009 May;28(5):412–415. doi: 10.1097/INF.0b013e3181910e2d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sirohi B, Powles RL, Chopra R, Russell N, Byrne JL, Prentice HG, et al. A study to determine the safety profile and maximum tolerated dose of micafungin (FK463) in patients undergoing haematopoietic stem cell transplantation. Bone Marrow Transplant. 2006 Jul;38(1):47–51. doi: 10.1038/sj.bmt.1705398. [DOI] [PubMed] [Google Scholar]
  • 80.Damle B, Stogniew M, Dowell J. Pharmacokinetics and tissue distribution of anidulafungin in rats. Antimicrob Agents Chemother. 2008 Jul;52(7):2673–2676. doi: 10.1128/AAC.01596-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dowell JA, Knebel W, Ludden T, Stogniew M, Krause D, Henkel T. Population pharmacokinetic analysis of anidulafungin, an echinocandin antifungal. J Clin Pharmacol. 2004 Jun;44(6):590–598. doi: 10.1177/0091270004265644. [DOI] [PubMed] [Google Scholar]
  • 82.Estes KE, Penzak SR, Calis KA, Walsh TJ. Pharmacology and antifungal properties of anidulafungin, a new echinocandin. Pharmacotherapy. 2009 Jan;29(1):17–30. doi: 10.1592/phco.29.1.17. [DOI] [PubMed] [Google Scholar]
  • 83.Benjamin DK, Jr, Driscoll T, Seibel NL, Gonzalez CE, Roden MM, Kilaru R, 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):632–638. doi: 10.1128/AAC.50.2.632-638.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cohen-Wolkowiez M, Benjamin DK., Jr Abstract: Safety and Pharmacokinetics (PK) of Multiple-Dose Anidulafungin in Neonates. In: Piper L, Moran C, Liu P, Aram J, Kashuba A, Capparelli E, Walsh TJ, Hope WW, Smith PB, editors. Interscience Conference on Antimicrobial Agents and Chemotherapy; September 12–15,2010; Boston, MA. 2010. [Google Scholar]

RESOURCES