Fluconazole is an antifungal agent with reported evidence for its prophylactic effect against systemic fungal infection in preterm infants. The aim of this study was to build a population pharmacokinetic model to evaluate the pharmacokinetic characteristics of intravenous and oral fluconazole in preterm infants with the current prophylactic fluconazole dosing regimen.
KEYWORDS: fluconazole, infant, premature, patient-specific modeling, pharmacokinetics
ABSTRACT
Fluconazole is an antifungal agent with reported evidence for its prophylactic effect against systemic fungal infection in preterm infants. The aim of this study was to build a population pharmacokinetic model to evaluate the pharmacokinetic characteristics of intravenous and oral fluconazole in preterm infants with the current prophylactic fluconazole dosing regimen. A pharmacokinetic model was developed using 301 fluconazole concentrations from 75 preterm infants with a baseline body weight (WT) ranging from 0.5 to 1.5 kg and an estimated glomerular filtration rate (eGFR) ranging from 12.9 to 58.5 ml/min/1.73 m2. Eligible infants received an intravenous or oral dose of 3 mg/kg of body weight of fluconazole, twice weekly with a ≥72-h dose interval, for 4 weeks. The model was qualified with basic goodness-of-fit diagnostics, visual predictive checks, and bootstrapping. The fluconazole pharmacokinetics was well described with a one-compartment linear model with a proportional residual error. The population clearance (CL) and volume of distribution (V) were derived as 0.0197 × (WT/1.00)0.746 × (eGFR/25.0)0.463 × exp(η) and 1.04 × WT × exp(η), respectively. Such covariate analyses augment the awareness of the need for personalized dosing in preterm infants. (This study has been registered at ClinicalTrials.gov under identifier NCT01683760.)
INTRODUCTION
Prophylactic uses of antifungal agents for the prevention of systemic fungal infections are used in both adult patients with immunodeficiency or cancer and preterm infants with immature immune systems (1–5). Such uses of antifungal agents do not necessarily eliminate infections but reportedly decrease the number of events of invasive systemic candidiasis, leading to a reduction in overall mortality by the suppression of Candida colonization (6–12). The widely used classes of antifungal agents for the prevention of systemic candidiasis are triazoles and polyenes (7, 13).
Numerous prophylactic uses of fluconazole, a synthetic triazole, have been reported to reduce the rate of invasive candidiasis in preterm infants with extremely low birth weight (ELBW) via the suppression of Candida colonization (6, 9, 11, 14, 15). Out of the pharmacokinetic-pharmacodynamic (PK-PD) indices for measuring antifungal activity, the ratio of the fluconazole dose to the MIC, termed the dose/MIC ratio, and the ratio of the area under the curve over 24 h after dose at steady state (AUC24) to the MIC, termed the AUC/MIC ratio, are known to best reflect the antifungal activity of fluconazole (16–20). In adults, maintained exposure with a daily dose of 100 mg was considered equivalent to an AUC24 of 100 mg · h/liter; the AUC/MIC and dose/MIC ratios give rise to identical numerical values (19). A minimum efficacious exposure of fluconazole can be estimated based on the increased success rate of therapeutic response and a dose/MIC ratio of >50 among adult patients with candidemia (19, 21). For fluconazole-susceptible Candida species based on the Clinical and Laboratory Standards Institute (CLSI) sensitivity breakpoint at an MIC of ≤8 mg/liter, the predicted minimum efficacious exposure of fluconazole required to ensure at least 70% efficacy for the treatment of invasive systemic candidiasis to meet an AUC/MIC ratio of >50 corresponds to an AUC24 of 400 mg · h/liter (21–25). This is equivalent to the susceptible dose-dependent species with MIC breakpoints of 32 mg/liter to meet an AUC/MIC ratio of >12, which is often cited as efficacious exposure for adults (26).
A daily dose of 400 mg is recommended in drug labeling for adults undergoing bone marrow transplantation requiring fluconazole prophylaxis; however, retrospective analyses from randomized controlled trials (RCTs) have shown that reduced doses of 100 to 200 mg gave a similar incidence of breakthrough infections (27, 28). When the dose equivalency scheme from adults is applied, daily doses of 100 and 200 mg resulting in an AUC24 of 100 to 200 mg · h/liter would correspond to the susceptible dose-dependent MICs of 2 and 4 mg/liter, respectively, for an AUC/MIC ratio of >50 (23). Due to the limited experience of PK-PD studies in preterm infants receiving fluconazole for preventive purposes, such presumptions are inevitable, and observational evidence remains to be provided (9). Thus far, the suggested dosing regimens for the early prevention of invasive systemic candidiasis in infants from Monte Carlo simulations of 3 and 6 mg/kg of body weight twice weekly are expected to meet AUC/MIC ratios of >50 for MICs of 2 and 4 mg/liter by maintaining the fluconazole concentration above 2 mg/liter for more than 40% during a ≥72-h interval with an AUC24 of 50 to 100 mg · h/liter (8, 19).
Although numerous findings support the effectiveness of fluconazole prophylaxis through RCTs, there are also controversial affirmations with debate on whether it truly reduces the mortality and morbidity rates among very-low-birth-weight (VLBW) infants with a birth weight (WT) of ≤1.5 kg (11, 12, 14, 15, 29–31). The link between fluconazole prophylaxis and its efficacy is still missing, and whether the lack of efficacy in systemic candidiasis prevention is due to other factors, such as insufficient exposure required to elicit the prophylactic effect, still remains unanswered. Therefore, a personalized approach through studies on the PK characteristics of fluconazole prophylaxis in the preterm infant population is necessary, because such information cannot be extrapolated from adults or full-term pediatric data (25).
Based on these understandings, this study aims to characterize the fluconazole PK in VLBW infants who have received fluconazole prophylaxis therapy, through population PK (PopPK) model building and evaluation.
RESULTS
Data set and baseline subject demographics.
Eighty eligible preterm infants were enrolled, with five in the intensive sampling group; however, five infants were excluded from the present study due to withdrawal of consent prior to study drug administration. In total, 303 fluconazole plasma concentrations from the remaining 75 preterm infants (with 3 in the intensive sampling group) were included in the population PK analysis (see Fig. SA1 in the supplemental material). The baseline median (interquartile range) values for gestational age (GA), postmenstrual age (PMA), postnatal age (PNA), WT, and estimated glomerular filtration rate (eGFR) were 30.3 (28.0 to 31.3) weeks, 30.6 (28.4 to 31.6) weeks, 3.0 (3.0 to 4.0) days, 1.1 (0.9 to 1.3) kg, and 26.7 (22.9 to 32.3) ml/min/1.73 m2, respectively (Table 1).
TABLE 1.
Baseline demographic characteristics of the subjects
aData are medians (interquartile ranges) or frequencies (percentages) unless otherwise specified.
bEstimated using the bedside Schwartz formula for infants, eGFR = 0.413 × HT/CRE.
Population pharmacokinetic analysis.
A one-compartment linear model structure with the bioavailability (F) estimate for the oral and intravenous administration of fluconazole adequately described the concentration-time profiles in preterm infants (Table 2 and Fig. SA2). The proportional error model included the between-subject variability (BSV) for the clearance (CL), the volume of distribution (V), and the first-order absorption rate constant for oral dose (Ka). The effect of WT on the CL and V was selected with estimated exponents and simple allometry, respectively. As a result of numerous covariate screenings, creatinine (CRE), GA, PMA, and PNA were ruled out from the significant covariates for CL and V (Table 2). Meanwhile, the exponential model described the gradual increase of CL; eGFR in the power term was a significant covariate for CL (Fig. 1 and Table 3) (32). In general, WT and eGFR had a positive relationship with the fluconazole CL, as expected.
TABLE 2.
Model-building processa
| Model | New model | Model description | Population model | OFV | ΔOFV |
|---|---|---|---|---|---|
| Reference model | |||||
| — | 100 | 1-compartment | CL = θCL × eη1 | 306.759 | |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 100 | 101 | 2-compartment | CL = θCL × eη1 | 306.758 | −0.001 |
| V2 = θV2 × (WT/1.00) × eη2 | |||||
| WT on V | V3 = θV3 × (WT/1.00) × eη3 | ||||
| 100 | 102 | 1-compartment, IIV on CL | CL = θCL × eη1 | 152.974 | −153.785 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 100 | 103 | 1-compartment, IIV on V | CL = θCL × eη1 | 225.421 | −51.338 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 100 | 104 | 1-compartment, IIV on Ka | CL = θCL × eη1 | 288.064 | −18.695 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 102 | 105 | 1-compartment, IIV on CL, V | CL = θCL × eη1 | 126.160 | −26.814 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 102 | 106 | 1-compartment, IIV on CL, Ka | CL = θCL × eη1 | 114.974 | −38.000 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 103 | 107 | 1-compartment, IIV on V, Ka | CL = θCL × eη1 | 242.456 | −12.965 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 106 | 108 | 1-compartment, IIV on CL, V, Ka | CL = θCL × eη1 | 96.156 | −18.818 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 108 | 109 | 1-compartment, IIV on CL, V, F1 | CL = θCL × eη1 | 126.033 | +29.877 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| 108 | 110 | 1-compartment, IIV on CL, V, Ka, F1 | CL = θCL × eη1 | 96.156 | 0 |
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| Base model | 1-compartment, IIV on CL, V, Ka | CL = θCL × eη1 | |||
| WT on V | V = θV × (WT/1.00) × eη2 | ||||
| Reference model | |||||
| 108 | 8001 | WT on V | V = θV × (WT/1.00) × eη2 | 96.156 | |
| 8001 | 8002 | WT on V | CL = θCL × (eGFR/25)θeGFR × eη1 | 46.094 | −50.062 |
| eGFR on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8001 | 8003 | WT on V, CL | CL = θCL × (WT/1.00)θWT × eη1 | 44.430 | −51.726 |
| V = θV × (WT/1.00) × eη2 | |||||
| 8003 | 8004 | WT on V | CL = θCL × (WT/1.00)θWT × (GA/30)θGA × eη1 | 42.148 | −2.282 |
| WT, GA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8003 | 8005 | WT on V | CL = θCL × (WT/1.00)θWT × (PNA/5)θPNA × eη1 | 34.770 | −9.660 |
| WT, PNA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8003 | 8006 | WT on V | CL = θCL × (WT/1.00)θWT × (PMA/30)θPMA × eη1 | 30.282 | −14.148 |
| WT, PMA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8003 | 8007 | WT on V | CL = θCL × (WT/1.00)θWT × (eGFR/25)θeGFR × eη1 | 17.211 | −27.219 |
| WT, eGFR on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8007 | 8008 | WT on V | CL = θCL × (WT/1.00)θWT × (eGFR/25)θeGFR × (GA/30)θGA × eη1 | 14.251 | −2.960 |
| WT, eGFR, GA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8007 | 8009 | WT on V | CL = θCL × (WT/1.00)θWT × (eGFR/25)θeGFR × (PNA/5)θPNA × eη1 | 17.491 | +0.280 |
| WT, eGFR, PNA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8007 | 8010 | WT on V | CL = θCL × (WT/1.00)θWT × (eGFR/25)θeGFR × (PMA/30)θPNA × eη1 | 13.930 | −3.281 |
| WT, eGFR, PMA on CL | V = θV × (WT/1.00) × eη2 | ||||
| 8003 | 8011 | WT on V | CL = θCL × (WT/1.00)θWT × (CRE/0.6)θeGFR × eη1 | 44.430 | 0 |
| WT, CRE on CL | V = θV × (WT/1.00) × eη2 | ||||
| Final model | WT on V | CL = θCL × (WT/1.00)θWT × (eGFR/25)θeGFR × eη1 | |||
| WT, eGFR on CL | V = θV × (WT/1.00) × eη2 | ||||
Abbreviations: CL, clearance; IIV, interindividual variability; WT, weight; eGFR, estimated glomerular filtration rate; PNA, postnatal age in days; GA, gestational age in weeks; PMA, postmenstrual age in weeks; V, volume of distribution; OFV, objective function value; ΔOFV, change in the OFV compared to the reference model; eη1, exp(η1).
FIG 1.
Schematic diagram of the final model. PO, oral; IV, intravenous.
TABLE 3.
Parameter estimates of the final population pharmacokinetic model of prophylactic fluconazole in preterm infantsa
| Parameter | Description (unit) | Estimation result {estimate [%RSEb ] or (%CV)} | Bootstrap resultc [median value (95% CI)] |
|---|---|---|---|
| Ka = θ1 | |||
| θ1 | First-order absorption rate constant (1/h) | 0.538 [18.5] | 0.536 (0.389, 0.729) |
| CL = θ2 × (WT)θ7 × (eGFR/25)θ8 × exp(η2) | |||
| θ2 | Clearance (liters/h) | 0.0197 [6.99] | 0.0194 (0.0174, 0.0217) |
| θ7 | Change in CL due to WT difference (liters/h) | 0.746 [29.2] | 0.741 (0.318, 1.004) |
| θ8 | Change in CL due to eGFR difference (liters/h) | 0.463 [24.4] | 0.456 (0.281, 0.683) |
| V = θ3 × (WT) × exp(η3) | |||
| θ3 | Vol of distribution (liters) | 1.04 [8.23] | 1.02 (0.882, 1.15) |
| F = θ4 | |||
| θ4 | Relative bioavailability of first-order absorption (%) | 0.909 [7.03] | 0.896 (0.806, 0.986) |
| Interindividual variance | |||
| ω12 | BSV of Ka | 1.44 (28.2) | 1.40 (0.758, 2.11) |
| ω22 | BSV of CL | 0.0552 (33.0) | 0.0502 (0.0239, 0.0840) |
| ω32 | BSV of V | 0.0447 (51.6) | 0.0403 (0.0111, 0.0808) |
| Residual variance | |||
| σ22 | Residual error (proportional) | 0.282 (7.61) | 0.279 (0.242, 0.312) |
Abbreviations: BSV, between-subject variability; %CV, coefficient of variation expressed as a percentage; CI, confidence interval; CRE, creatinine level; eGFR, estimated glomerular filtration rate; RSE, relative standard error.
%RSE = (standard error/parameter estimate) × 100.
Determined from a parametric bootstrap resampling of 500 replicates.
The goodness-of-fit (GOF) and visual predictive check (VPC) plots and the bootstrap resampling of the final model showed good adequacy between the observed and predicted fluconazole concentrations obtained from preterm infants (Fig. 2 and 3). The final model parameter estimates had good precision, as presented in Table 3, with the 95% confidence intervals (CIs) generated by the bootstrap-resampled simulated trials. The percent relative standard error around the parameter point estimates was in the range of 7.0 to 29.2%, along with the maximum interindividual variance of 51.6% for V, shown as the percent coefficient of variation (%CV). Overall, the final PK model was robust at describing the PK characteristics of oral and intravenous fluconazole administrations in VLBW preterm infants.
FIG 2.
Basic goodness-of-fit plot of the final pharmacokinetic model. Solid lines, line of identity; dotted lines, line of reference (y = 0); gray lines, line of locally weighted scatterplot smoothing (LOWESS).
FIG 3.
Visual predictive check results for oral (a) and intravenous (b) administrations. Solid red lines, median from the time-concentration profile at steady state; solid blue lines, lower and upper limits of the 95% confidence interval; shaded areas in red, 95% confidence intervals for the median of the simulated concentrations; shaded areas in blue, 5th and 95th percentiles of the simulated concentrations. All y axes are presented in linear scales.
DISCUSSION
The controversy over the effectiveness of fluconazole prophylaxis in preterm infants remains to be followed up with a further understanding of the relationship between the PK, proposed PD indices, and clinical outcomes. In underobserved populations such as preterm infants, a population pharmacokinetic approach is a known stepping-stone toward personalized pharmacotherapy. In this study, we have constructed an adequate PK model for preterm infants who receive a routine prophylactic fluconazole regimen (Table 3 and Fig. 1).
The goal for dose selection in infant populations primarily lies upon understanding the sources of within- and between-subject variabilities in terms of their PK characteristics. The major candidate sources of PK variability are within growth and maturation in infants, contributing order-of-magnitude differences in body weight compared to adults (33, 34). In a study by Rhodin et al., WT and PMA were used as the maturation factors to describe human renal function (35). Therefore, to disentangle competing covariate influences, WT was used as a surrogate for growth as the first covariate to explain the interindividual differences in a systemic approach (36, 37). The magnitude of power for the CL model was estimated rather than using fixed time-related or physiological variables scaled with a WT exponent of 0.25 or 0.75, respectively (38). The estimated power term within the population for CL (0.746) was almost identical to the allometric 3/4 power model when the eGFR was accounted for as the covariate. Unlike the population PK model previously reported by Wade et al., where GA and PNA were used to describe CL in infants with a WT range of 0.5 to 7.1 kg, the birth WT was included as the model’s most significant covariate (Table 2) (39). No other surrogate for maturation, GA, PNA, or PMA, further influenced the CL of this population, which consequently implies that in VLBW infants, like this population (premature infants with birth WT of ≤1.5 kg), the increase in WT influences the maturation of the kidney, which has the largest effect on the CL of fluconazole (40). The second covariate used to explain the fluconazole CL in the study infants was the eGFR calculated with the original bedside Schwartz formula for children (41). Considering the variables height (HT) and CRE included in the formula, HT may seem redundant for the effect of growth along with WT, which has already been included as a covariate. However, when CRE was examined within the covariate analyses, a similar improvement in the model fit was observed (Table 2). Therefore, it seems feasible to assume that there is no correlation between WT and CRE; hence, they are two independent covariates.
Fluconazole is used as a marker for passive distal tubular reabsorption due to its extensive tubular reabsorption (42, 43). In terms of glomerular filtration, the acquisition of renal function is completed during postnatal development, whereas tubular function matures more progressively with greater efficiency in infants for some mechanisms than in adults (44, 45). Drug transporter expression is also known to differ in an age-dependent manner for hepatic and intestinal drug transporters (46). Therefore, the contribution of immature passive renal drug transporter compositions can lead to a lesser extent of reabsorption of fluconazole into the system, which may contribute to the insufficient exposure observed compared to that anticipated within the premature infant population (34). Another maturational alteration compared to adults and older children is the decreased plasma proteins in the neonatal population, which may lead to an increase in the unbound drug concentration (47). For drugs such as phenobarbital, the percent difference in protein binding in neonates is close to 50% (48). Collectively, approaches different from those for adults are bound to be applied in preterm infants who are constantly undergoing maturation of the organs, which affects the PK of drugs administered through personalized optimum pharmacotherapy.
One drawback of the present study is that significant covariates for the CL of fluconazole in the neonate population within this study were WT and eGFR, and the weight range was 0.5 to 1.5 kg, with a maximum calculated eGFR of 58.5 ml/min/1.73 m2. In a previous population PK study in young infants with a WT range of 0.5 to 7.1 kg, fluconazole CL was best described by WT, CRE, PNA, and GA at birth (39). Another population PK study in extremely-low-birth-weight (ELBW) infants with a WT of ≤0.75 kg found that CRE was the only predictor of fluconazole CL (49). The differences between the selected covariates and those previous studies may be due to the narrower ranges of candidate covariates for the observed population and the WT range of the subjected infants included within the analyses. Second, not only WT and HT but also CRE data were not an exact match for each dosing and PK sampling time point. In addition, we included only 75 infants in the analysis. However, the PK parameters derived from the final model were in agreement with those derived by Wade et al. (39). Addressing these concerns would require further study with a larger sample size. Third, although the original bedside Schwartz formula is a validated method for the estimation of GFR in a predominantly nonchronic kidney disease population regardless of birth weight or term age due to the direct impact of the maternal CRE on the serum CRE in infants during the first 14 days of life, substantial limitations on the estimation of GFR lie within (41, 50). In addition, cystatin C and beta-trace protein are known to reflect neonatal renal function more accurately, with minimal amounts crossing the placenta, unlike CRE (51). Neither cystatin C nor beta-trace protein levels were available in the present study, hence the use of HT and CRE for the eGFR calculation. However, such a drawback can also be seen as an advantage, in that the eGFR with the Schwartz equation can easily be calculated in clinical practice with a routine safety laboratory item and growth measurements.
With the final PK model adequately describing the observed plasma concentration of prophylactic fluconazole in the preterm infant population, this study suggests that WT at birth and eGFR are predictors for fluconazole CL in the observed population. Such work is expected to be useful for the further evaluation of the PK-PD indices proposed thus far for prophylactic fluconazole use in preterm infants, complemented with observations of long-term adverse effects, such as possible concerns of adrenocortical insufficiency, neurodevelopment, and hyperbilirubinemia (52–54).
MATERIALS AND METHODS
Study design.
This study (ClinicalTrials.gov identifier NCT01683760) was conducted in full accordance with the principles stipulated in the Declaration of Helsinki as amended in 2008 (Seoul, Republic of Korea) (55) and International Conference on Harmonization (ICH) guidelines on good clinical practice (GCP) (56). The study protocol was reviewed and approved by the Seoul National University Hospital Institutional Review Board (IRB) (IRB no. H-1207-024-416).
A prospective, single-center, open-label, pharmacokinetic study was performed in preterm infants. Voluntary informed written consent for participation in this study was obtained from the legal guardians of the preterm infants hospitalized at the neonatal intensive care unit (NICU) at Seoul National University Children’s Hospital who required fluconazole prophylaxis for the prevention of systemic fungal infection and who had birth weights of ≤1.5 kg. Infants who received an antifungal agent for treatment purposes or with biological mothers who had been treated with an antifungal agent due to intrauterine fungal infection during pregnancy were excluded from the study.
Starting from the third day after birth, 80 eligible preterm infants received 3 mg/kg fluconazole (Diflucan; Pfizer Inc., Seoul, Republic of Korea), orally through an orogastric tube or intravenously if a venous catheter was present, with at least a 72-h interval twice weekly for a total of 4 weeks. Of the 80 subjects, 8 with a central venous catheter, including an umbilical venous catheter, were allocated to the intensive PK sampling group, and the rest (72 subjects) were allocated to the sparse sampling group (see Fig. SA1 in the supplemental material). Serial blood samples (0.5 ml) were obtained to determine the plasma fluconazole concentration predose and 0.5, 1, 3, 10, 24, 48, and 72 h after the third dosing for the intensive sampling group. In the sparse sampling group, one predose and three randomly ordered extra blood samples of 0.5 ml from scheduled laboratory analyses were used from each infant during intensive care after the third dosing of fluconazole.
Determination of the plasma fluconazole concentration.
Plasma fluconazole concentrations were determined using a validated method, as follows. Briefly, the Agilent 1260 high-performance liquid chromatography (HPLC) system (Agilent, Palo Alto, CA, USA) was equipped with a Luna C18 column (5 μm, 2.0 by 100 mm; Phenomenex, Torrance, CA, USA). This HPLC system was coupled with an Applied Biosystems API Qtrap 4000 tandem mass spectrometry (MS/MS) instrument (Applied Biosystems, Foster City, CA, USA). Quantification was conducted using an electrospray ionization (ESI) source in the positive mode and the following conditions: curtain gas of 20, gas 1 (nebulizer gas) of 50, gas 2 (heater gas) of 50, charged aerosol detection gas on high, TurboIonSpray (IS) voltage of 5,500 V, entrance potential (EP) of 10 V, collision energy (CE) of 25 V, probe temperature of 500°C, and scan time of 250 ms. HPLC separation was performed using an isocratic mobile phase composed of 10 mM ammonium formate in distilled water (0.1% formic acid) and 0.1% formic acid in acetonitrile (65:35, vol/vol) on a Luna guard column (2 μm, 4.0 by 2.0 mm; Phenomenex, Torrance, CA, USA). The flow rate was 0.3 ml/min, and the injection volume was 2 μl, with a 3-min run time. The multiple-reaction monitoring mode (MRM) was used for MS/MS detection at m/z 307.129 > 238.200 for fluconazole and at m/z 311.178 > 242.200 for fluconazole-d4 (internal standard). The range of quantification was 0.001 to 2 mg/liter. The accuracy (expressed as the percent difference from the theoretical concentration) of the quality control samples used during the sample analysis was in the range of 91.7 to 95.4%, with a precision (expressed as a coefficient of variation) of ≤5.2%.
Population pharmacokinetic analysis.
Plasma concentration-time records obtained from the eligible preterm infants were used for population PK analysis. The first-order conditional estimation method with the η-ε interaction option (FOCE-I) was used throughout the model-building process. Both single- and multicompartmental models were tested to describe the distribution of fluconazole PK. Different model parameterization scenarios were tested using the nonlinear mixed-effects method in NONMEM version 7.4.0 (Icon Development Solutions, Ellicott City, MD, USA) (57). Individual model parameters were obtained by a Bayesian estimation implemented in NONMEM with a covariance step. The interindividual variabilities of the PK parameters were tested in the model, along with various residual-error models for the intraindividual variabilities.
GA and postmenstrual ages (PMA) in weeks, postnatal age (PNA) in days, body weight (WT), height (HT), and the following baseline measures of laboratory tests were included in the covariate screening: alanine transaminase (ALT) and aspartate transaminase (AST) levels, serum creatinine level (CRE), estimated glomerular filtration rate (eGFR) (using the bedside Schwartz formula for children, 0.413 × HT/CRE), and blood urea nitrogen level (BUN) (41). For time-varying observations such as WT and CRE, more than one value per infant was included since the baseline observation was over the fluconazole treatment period. The covariates were screened based on a forward-selection significance level of 0.05 and a backward-deletion criterion of 0.01 for the objective function values (OFVs).
Successful minimization and standard basic model diagnostic plots, including individual fittings, goodness-of-fit (GOF) plots, and visual predictive check (VPC) plots for observed and predicted plasma fluconazole concentrations, were examined to assess the model fitting and the performance of the predictions. In addition, the precision of the parameter estimates was evaluated using 500 bootstrap-resampled simulated trials from the original data set that were sequentially estimated using the same final model for comparison of the medians and 95% confidence intervals (CIs) (2.5th and 97.5th percentiles) to those of the final model parameter estimates. The bootstrap procedures and VPC plot construction were performed with Perl-speaks-NONMEM (PsN) (version 4.7.0) (58).
Supplementary Material
ACKNOWLEDGMENTS
This research was supported by a grant (12172MFDS231) from the Ministry of Food and Drug Safety of Korea. Y.K.K. received a scholarship from the BK21-plus education program provided by the National Research Foundation of Korea grant funded by the Korean Government.
We gratefully acknowledge Chan Hyung Kim for his support on the model refinement.
Footnotes
Supplemental material for this article may be found at https://doi.org/10.1128/AAC.01960-18.
REFERENCES
- 1.Baden LR, Swaminathan S, Angarone M, Blouin G, Camins BC, Casper C, Cooper B, Dubberke ER, Engemann AM, Freifeld AG, Greene JN, Ito JI, Kaul DR, Lustberg ME, Montoya JG, Rolston K, Satyanarayana G, Segal B, Seo SK, Shoham S, Taplitz R, Topal J, Wilson JW, Hoffmann KG, Smith C. 2016. Prevention and treatment of cancer-related infections, version 2.2016, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 14:882–913. doi: 10.6004/jnccn.2016.0093. [DOI] [PubMed] [Google Scholar]
- 2.Havlir DV, Dube MP, McCutchan JA, Forthal DN, Kemper CA, Dunne MW, Parenti DM, Kumar PN, White AC Jr, Witt MD, Nightingale SD, Sepkowitz KA, MacGregor RR, Cheeseman SH, Torriani FJ, Zelasky MT, Sattler FR, Bozzette SA. 1998. Prophylaxis with weekly versus daily fluconazole for fungal infections in patients with AIDS. Clin Infect Dis 27:1369–1375. doi: 10.1086/515018. [DOI] [PubMed] [Google Scholar]
- 3.Just-Nubling G, Gentschew G, Meissner K, Odewald J, Staszewski S, Helm EB, Stille W. 1991. Fluconazole prophylaxis of recurrent oral candidiasis in HIV-positive patients. Eur J Clin Microbiol Infect Dis 10:917–921. doi: 10.1007/BF02005444. [DOI] [PubMed] [Google Scholar]
- 4.Vanden Bossche H, Koymans L, Moereels H. 1995. P450 inhibitors of use in medical treatment: focus on mechanisms of action. Pharmacol Ther 67:79–100. doi: 10.1016/0163-7258(95)00011-5. [DOI] [PubMed] [Google Scholar]
- 5.Vazquez L. 2016. Antifungal prophylaxis in immunocompromised patients. Mediterr J Hematol Infect Dis 8:e2016040. doi: 10.4084/MJHID.2016.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Healy CM, Campbell JR, Zaccaria E, Baker CJ. 2008. Fluconazole prophylaxis in extremely low birth weight neonates reduces invasive candidiasis mortality rates without emergence of fluconazole-resistant Candida species. Pediatrics 121:703–710. doi: 10.1542/peds.2007-1130. [DOI] [PubMed] [Google Scholar]
- 7.Lollis TR, Bradshaw WT. 2014. Fungal prophylaxis in neonates: a review article. Adv Neonatal Care 14:17–23. doi: 10.1097/ANC.0000000000000048. [DOI] [PubMed] [Google Scholar]
- 8.Wade KC, Benjamin DK Jr, Kaufman DA, Ward RM, Smith PB, Jayaraman B, Adamson PC, Gastonguay MR, Barrett JS. 2009. Fluconazole dosing for the prevention or treatment of invasive candidiasis in young infants. Pediatr Infect Dis J 28:717–723. doi: 10.1097/INF.0b013e31819f1f50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kim SY, Lee SJ, Kim MJ, Song ES, Choi YY. 2007. Fluconazole prophylaxis in high-risk, very low birth weight infants. Korean J Pediatr 50:636–642. doi: 10.3345/kjp.2007.50.7.636. [DOI] [Google Scholar]
- 10.Pea F, Lewis RE. 2018. Overview of antifungal dosing in invasive candidiasis. J Antimicrob Chemother 73:i33–i43. doi: 10.1093/jac/dkx447. [DOI] [PubMed] [Google Scholar]
- 11.Parikh TB, Nanavati RN, Patankar CV, Rao S, Bisure K, Udani RH, Mehta P. 2007. Fluconazole prophylaxis against fungal colonization and invasive fungal infection in very low birth weight infants. Indian Pediatr 44:830–837. [PubMed] [Google Scholar]
- 12.Benjamin DK Jr, Hudak ML, Duara S, Randolph DA, Bidegain M, Mundakel GT, Natarajan G, Burchfield DJ, White RD, Shattuck KE, Neu N, Bendel CM, Kim MR, Finer NN, Stewart DL, Arrieta AC, Wade KC, Kaufman DA, Manzoni P, Prather KO, Testoni D, Berezny KY, Smith PB, Fluconazole Prophylaxis Study Team. 2014. Effect of fluconazole prophylaxis on candidiasis and mortality in premature infants: a randomized clinical trial. JAMA 311:1742–1749. doi: 10.1001/jama.2014.2624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Watt K, Manzoni P, Cohen-Wolkowiez M, Rizzollo S, Boano E, Jacqz-Aigrain E, Benjamin DK. 2013. Triazole use in the nursery: fluconazole, voriconazole, posaconazole, and ravuconazole. Curr Drug Metab 14:193–202. [PMC free article] [PubMed] [Google Scholar]
- 14.Kaufman D, Boyle R, Hazen KC, Patrie JT, Robinson M, Donowitz LG. 2001. Fluconazole prophylaxis against fungal colonization and infection in preterm infants. N Engl J Med 345:1660–1666. doi: 10.1056/NEJMoa010494. [DOI] [PubMed] [Google Scholar]
- 15.Manzoni P, Stolfi I, Pugni L, Decembrino L, Magnani C, Vetrano G, Tridapalli E, Corona G, Giovannozzi C, Farina D, Arisio R, Merletti F, Maule M, Mosca F, Pedicino R, Stronati M, Mostert M, Gomirato G, Italian Task Force for the Study and Prevention of Neonatal Fungal Infections, Italian Society of Neonatology. 2007. A multicenter, randomized trial of prophylactic fluconazole in preterm neonates. N Engl J Med 356:2483–2495. doi: 10.1056/NEJMoa065733. [DOI] [PubMed] [Google Scholar]
- 16.Andes D, van Ogtrop M. 1999. Characterization and quantitation of the pharmacodynamics of fluconazole in a neutropenic murine disseminated candidiasis infection model. Antimicrob Agents Chemother 43:2116–2120. doi: 10.1128/AAC.43.9.2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Baddley JW, Patel M, Bhavnani SM, Moser SA, Andes DR. 2008. Association of fluconazole pharmacodynamics with mortality in patients with candidemia. Antimicrob Agents Chemother 52:3022–3028. doi: 10.1128/AAC.00116-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Louie A, Drusano GL, Banerjee P, Liu QF, Liu W, Kaw P, Shayegani M, Taber H, Miller MH. 1998. Pharmacodynamics of fluconazole in a murine model of systemic candidiasis. Antimicrob Agents Chemother 42:1105–1109. doi: 10.1128/AAC.42.5.1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pai MP, Turpin RS, Garey KW. 2007. Association of fluconazole area under the concentration-time curve/MIC and dose/MIC ratios with mortality in nonneutropenic patients with candidemia. Antimicrob Agents Chemother 51:35–39. doi: 10.1128/AAC.00474-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rex JH, Pfaller MA, Galgiani JN, Bartlett MS, Espinel-Ingroff A, Ghannoum MA, Lancaster M, Odds FC, Rinaldi MG, Walsh TJ, Barry AL. 1997. 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 24:235–247. doi: 10.1093/clinids/24.2.235. [DOI] [PubMed] [Google Scholar]
- 21.Clancy CJ, Yu VL, Morris AJ, Snydman DR, Nguyen MH. 2005. Fluconazole MIC and the fluconazole dose/MIC ratio correlate with therapeutic response among patients with candidemia. Antimicrob Agents Chemother 49:3171–3177. doi: 10.1128/AAC.49.8.3171-3177.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Clancy CJ, Staley B, Nguyen MH. 2006. In vitro susceptibility of breakthrough Candida bloodstream isolates correlates with daily and cumulative doses of fluconazole. Antimicrob Agents Chemother 50:3496–3498. doi: 10.1128/AAC.00741-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Clinical and Laboratory Standards Institute. 2017. Performance standards for antifungal susceptibility testing of yeasts, 1st ed, CLSI supplement M60, vol 37 Clinical and Laboratory Standards Institute, Wayne, PA. [Google Scholar]
- 24.Pappas PG, Rex JH, Sobel JD, Filler SG, Dismukes WE, Walsh TJ, Edwards JE, Infectious Diseases Society of America. 2004. Guidelines for treatment of candidiasis. Clin Infect Dis 38:161–189. doi: 10.1086/380796. [DOI] [PubMed] [Google Scholar]
- 25.Turner K, Manzoni P, Benjamin DK, Cohen-Wolkowiez M, Smith PB, Laughon MM. 2012. Fluconazole pharmacokinetics and safety in premature infants. Curr Med Chem 19:4617–4620. doi: 10.2174/092986712803306367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Autmizguine J, Guptill JT, Cohen-Wolkowiez M, Benjamin DK Jr, Capparelli EV. 2014. Pharmacokinetics and pharmacodynamics of antifungals in children: clinical implications. Drugs 74:891–909. doi: 10.1007/s40265-014-0227-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.MacMillan ML, Goodman JL, DeFor TE, Weisdorf DJ. 2002. Fluconazole to prevent yeast infections in bone marrow transplantation patients: a randomized trial of high versus reduced dose, and determination of the value of maintenance therapy. Am J Med 112:369–379. doi: 10.1016/S0002-9343(01)01127-5. [DOI] [PubMed] [Google Scholar]
- 28.Safdar A, van Rhee F, Henslee-Downey JP, Singhal S, Mehta J. 2001. Candida glabrata and Candida krusei fungemia after high-risk allogeneic marrow transplantation: no adverse effect of low-dose fluconazole prophylaxis on incidence and outcome. Bone Marrow Transplant 28:873–878. doi: 10.1038/sj.bmt.1703252. [DOI] [PubMed] [Google Scholar]
- 29.Isaacs D. 2008. Fungal prophylaxis in very low birth weight neonates: nystatin, fluconazole or nothing? Curr Opin Infect Dis 21:246–250. doi: 10.1097/QCO.0b013e3282f8adab. [DOI] [PubMed] [Google Scholar]
- 30.Kicklighter SD, Springer SC, Cox T, Hulsey TC, Turner RB. 2001. Fluconazole for prophylaxis against candidal rectal colonization in the very low birth weight infant. Pediatrics 107:293–298. doi: 10.1542/peds.107.2.293. [DOI] [PubMed] [Google Scholar]
- 31.Lee J, Kim HS, Shin SH, Choi CW, Kim EK, Choi EH, Kim BI, Choi JH. 2016. Efficacy and safety of fluconazole prophylaxis in extremely low birth weight infants: multicenter pre-post cohort study. BMC Pediatr 16:67. doi: 10.1186/s12887-016-0605-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Allegaert K, Anderson BJ, Cossey V, Holford NH. 2006. Limited predictability of amikacin clearance in extreme premature neonates at birth. Br J Clin Pharmacol 61:39–48. doi: 10.1111/j.1365-2125.2005.02530.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Holford N, Heo YA, Anderson B. 2013. A pharmacokinetic standard for babies and adults. J Pharm Sci 102:2941–2952. doi: 10.1002/jps.23574. [DOI] [PubMed] [Google Scholar]
- 34.Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. 2003. Developmental pharmacology—drug disposition, action, and therapy in infants and children. N Engl J Med 349:1157–1167. doi: 10.1056/NEJMra035092. [DOI] [PubMed] [Google Scholar]
- 35.Rhodin MM, Anderson BJ, Peters AM, Coulthard MG, Wilkins B, Cole M, Chatelut E, Grubb A, Veal GJ, Keir MJ, Holford NH. 2009. Human renal function maturation: a quantitative description using weight and postmenstrual age. Pediatr Nephrol 24:67–76. doi: 10.1007/s00467-008-0997-5. [DOI] [PubMed] [Google Scholar]
- 36.Holford NH. 1996. A size standard for pharmacokinetics. Clin Pharmacokinet 30:329–332. doi: 10.2165/00003088-199630050-00001. [DOI] [PubMed] [Google Scholar]
- 37.Anderson B. 2004. Disentangling PK-PD in neonates. Arch Dis Child Fetal Neonatal Ed 89:F3–F4. doi: 10.1136/fn.89.1.F3-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Peters RH. 1986. The ecological implications of body size, vol 2 Cambridge University Press, Cambridge, United Kingdom. [Google Scholar]
- 39.Wade KC, Wu D, Kaufman DA, Ward RM, Benjamin DK Jr, Sullivan JE, Ramey N, Jayaraman B, Hoppu K, Adamson PC, Gastonguay MR, Barrett JS, National Institute of Child Health and Development Pediatric Pharmacology Research Unit Network. 2008. Population pharmacokinetics of fluconazole in young infants. Antimicrob Agents Chemother 52:4043–4049. doi: 10.1128/AAC.00569-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Schmidt IM, Chellakooty M, Boisen KA, Damgaard IN, Mau Kai C, Olgaard K, Main KM. 2005. Impaired kidney growth in low-birth-weight children: distinct effects of maturity and weight for gestational age. Kidney Int 68:731–740. doi: 10.1111/j.1523-1755.2005.00451.x. [DOI] [PubMed] [Google Scholar]
- 41.Staples A, LeBlond R, Watkins S, Wong C, Brandt J. 2010. Validation of the revised Schwartz estimating equation in a predominantly non-CKD population. Pediatr Nephrol 25:2321–2326. doi: 10.1007/s00467-010-1598-7. [DOI] [PubMed] [Google Scholar]
- 42.Gross AS, McLachlan AJ, Minns I, Beal JB, Tett SE. 2001. 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 51:547–555. doi: 10.1046/j.1365-2125.2001.01390.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Debruyne D, Ryckelynck JP. 1993. Clinical pharmacokinetics of fluconazole. Clin Pharmacokinet 24:10–27. doi: 10.2165/00003088-199324010-00002. [DOI] [PubMed] [Google Scholar]
- 44.Arant BS., Jr 1978. Developmental patterns of renal functional maturation compared in the human neonate. J Pediatr 92:705–712. doi: 10.1016/S0022-3476(78)80133-4. [DOI] [PubMed] [Google Scholar]
- 45.Bitsori M. 2012. The development of renal function, p 9–20. In Sakellaris S. (ed), Essentials in pediatric urology. Research Signpost, Kerala, India. [Google Scholar]
- 46.Mooij MG, Schwarz UI, de Koning BA, Leeder JS, Gaedigk R, Samsom JN, Spaans E, van Goudoever JB, Tibboel D, Kim RB, de Wildt SN. 2014. Ontogeny of human hepatic and intestinal transporter gene expression during childhood: age matters. Drug Metab Dispos 42:1268–1274. doi: 10.1124/dmd.114.056929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Johnson PJ. 2011. Neonatal pharmacology—pharmacokinetics. Neonatal Netw 30:54–61. doi: 10.1891/0730-0832.30.1.54. [DOI] [PubMed] [Google Scholar]
- 48.Reiter PD. 2002. Neonatal pharmacology and pharmacokinetics. NeoReviews 3:c229–c235. [Google Scholar]
- 49.Momper JD, Capparelli EV, Wade KC, Kantak A, Dhanireddy R, Cummings JJ, Nedrelow JH, Hudak ML, Mundakel GT, Natarajan G, Gao J, Laughon M, Smith PB, Benjamin DK Jr. 2016. Population pharmacokinetics of fluconazole in premature infants with birth weights less than 750 grams. Antimicrob Agents Chemother 60:5539–5545. doi: 10.1128/AAC.00963-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schwartz GJ, Brion LP, Spitzer A. 1987. The use of plasma creatinine concentration for estimating glomerular filtration rate in infants, children, and adolescents. Pediatr Clin North Am 34:571–590. doi: 10.1016/S0031-3955(16)36251-4. [DOI] [PubMed] [Google Scholar]
- 51.Bariciak E, Yasin A, Harrold J, Walker M, Lepage N, Filler G. 2011. Preliminary reference intervals for cystatin C and beta-trace protein in preterm and term neonates. Clin Biochem 44:1156–1159. doi: 10.1016/j.clinbiochem.2011.06.987. [DOI] [PubMed] [Google Scholar]
- 52.Aghai ZH, Mudduluru M, Nakhla TA, Amendolia B, Longo D, Kemble N, Kaki S, Sutsko R, Saslow JG, Stahl GE. 2006. Fluconazole prophylaxis in extremely low birth weight infants: association with cholestasis. J Perinatol 26:550–555. doi: 10.1038/sj.jp.7211570. [DOI] [PubMed] [Google Scholar]
- 53.Costa S, Barone G, Vento G, Tirone C, Perelli S, Romagnoli C. 2014. Adrenocortical insufficiency is not a problem in preterm infants treated with antifungal prophylaxis with fluconazole. Acta Paediatr 103:e234–e237. doi: 10.1111/apa.12613. [DOI] [PubMed] [Google Scholar]
- 54.Kaufman DA, Cuff AL, Wamstad JB, Boyle R, Gurka MJ, Grossman LB, Patrick P. 2011. Fluconazole prophylaxis in extremely low birth weight infants and neurodevelopmental outcomes and quality of life at 8 to 10 years of age. J Pediatr 158:759.e1–765.e1. doi: 10.1016/j.jpeds.2010.11.002. [DOI] [PubMed] [Google Scholar]
- 55.World Medical Association. 2013. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA 310:2191–2194. doi: 10.1001/jama.2013.281053. [DOI] [PubMed] [Google Scholar]
- 56.Dixon JR., Jr 1998. The International Conference on Harmonization good clinical practice guideline. Qual Assur 6:65–74. doi: 10.1080/105294199277860. [DOI] [PubMed] [Google Scholar]
- 57.Beal SL, Sheiner LB, Boeckmann AJ, Bauer RJ. 2009. NONMEM user’s guides. Icon Development Solutions, Ellicott City, MD. [Google Scholar]
- 58.Lindbom L, Ribbing J, Jonsson EN. 2004. Perl-speaks-NONMEM (PsN)—a Perl module for NONMEM related programming. Comput Methods Programs Biomed 75:85–94. doi: 10.1016/j.cmpb.2003.11.003. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




