Skip to main content
Erschienen in: BMC Pediatrics 1/2019

Open Access 01.12.2019 | Research article

Optimizing gentamicin conventional and extended interval dosing in neonates using Monte Carlo simulation – a retrospective study

verfasst von: Monique Bergenwall, Sandra A. N. Walker, Marion Elligsen, Dolores C. Iaboni, Carla Findlater, Winnie Seto, Eugene Ng

Erschienen in: BMC Pediatrics | Ausgabe 1/2019

Abstract

Background

Although aminoglycosides are routinely used in neonates, controversy exists regarding empiric dosing regimens. The objectives were to determine gentamicin pharmacokinetics in neonates, and develop initial mg/kg dosing recommendations that optimized target peak and trough concentration attainment for conventional and extended-interval dosing (EID) regimens.

Methods

Patient demographics and steady-state gentamicin concentration data were retrospectively collected for 60 neonates with no renal impairment admitted to a level III neonatal intensive care unit. Mean pharmacokinetics were calculated and multiple linear regression was performed to determine significant covariates of clearance (L/h) and volume of distribution (L). Classification and regression tree (CART) analysis identified breakpoints for significant covariates. Monte Carlo Simulation (MCS) was used to determine optimal dosing recommendations for each CART-identified sub-group.

Results

Gentamicin clearance and volume of distribution were significantly associated with weight at gentamicin initiation. CART-identified breakpoints for weight at gentamicin initiation were: ≤ 850 g, 851-1200 g, and > 1200 g. MCS identified that a conventional dose of gentamicin 3.5 mg/kg given every 48 h or an EID of 8-9 mg/kg administered every 72 h in neonates weighing ≤ 850 g, and every 24 and 48 h, respectively, in neonates weighing 851-1200 g, provided the best probability of attaining conventional (peak: 5-10 mg/L and trough: ≤ 2 mg/L) and EID targets (peak:12-20 mg/L, trough:≤ 0.5 mg/L). Insufficient sample size in the > 1200 g neonatal group precluded further investigation of this weight category.

Conclusions

This study provides initial gentamicin dosing recommendations that optimize target attainment for conventional and EID regimens in neonates weighing ≤ 1200 g. Prospective validation and empiric dose optimization for neonates > 1200 g is needed.
Hinweise
Monique Bergenwall and Sandra A. N. Walker contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ANOVA
Analysis of variance
BUN
Blood urea nitrogen
BW
Birth weight
CART
Classification and regression tree
CGA
Corrected gestational age
CI
Confidence interval
Cl
Clearance
EID
Extended-interval dosing
GA
Gestational age
GNB
Gram negative bacteria
ke
Elimination rate constant
MCS
Monte Carlo Simulation
MIC
Minimum inhibitory concentration
MLR
Multiple linear regression
NICU
Neonatal intensive care unit
PK
Pharmacokinetic
PNA
Post-natal age
sCr
Serum creatinine
SD
Standard deviation
SGA
Small for gestational age
SHSC
Sunnybrook Health Sciences Centre
t1/2
Half-life
TDM
Therapeutic drug monitoring
Vd
Volume of distribution

Background

Although aminoglycosides are routinely used in neonates, controversy exists regarding recommended empiric dosing to optimize target attainment with either conventional dosing (peak: 5-10 mg/L and troughs ≤2 mg/L) or extended-interval dosing (higher peak and undetectable trough) [1, 2]. In adult and older pediatric populations, EID regimens targeting peak concentrations of ≥ 20 mg/L are routinely recommended based on data suggesting that aminoglycoside activity is optimized with peak: minimum inhibitory concentration (MIC) ratios of 8–10:1 [35]. For these patient populations, EID has consistently demonstrated equal efficacy, and equal or reduced toxicity versus conventional dosing [1, 610].
While data exist to support the use of EID in neonates [6, 1126], consensus is lacking regarding optimal EID target concentrations that optimize efficacy and minimize toxicity in this patient population. Peak concentrations investigated in neonates vary from 4 to 20 mg/L [1526], and typically remain below 12 mg/L, with no clear rationale. Furthermore, infants born at a gestational age (GA) ≤ 28 weeks, along with those with a birth weight (BW) of ≤1500 g, are underrepresented in EID studies. These infants constitute approximately 20% of all neonates admitted to Canadian neonatal intensive care units (NICUs), and 50% of those admitted to Level III NICUs [27]. Since aminoglycoside pharmacokinetic (PK) parameters in neonates may be influenced by weight [15, 1820, 28, 29], gestational age [15, 28, 29] and postnatal age [19, 28, 29], further research is required in this unique population in order to optimize target attainment and thereby, maximize the probability of efficacy of the antibiotic while minimizing the risk of nephrotoxicity.
The objectives of this study were to determine the pharmacokinetics of gentamicin in neonates with no clinical evidence of renal impairment in a Level III NICU, identify significant covariates of gentamicin PK parameters in neonates, and develop practical initial dosing recommendations with the highest probability of attaining target peak and trough serum concentrations currently accepted in clinical practice for both conventional dosing (trough < 2 mg/L and peak 5–10) and EID (trough < 0.5 mg/L and peak 8-20 mg/L, 12-20 mg/L, 15-20 mg/L and > 20 mg/L) of gentamicin.

Methods

This retrospective study was conducted in the level III NICU at Sunnybrook Health Sciences Centre (SHSC) in Toronto, Ontario, Canada. SHSC is a 1325-bed tertiary care teaching hospital, with 48 NICU beds [30].

Patient eligibility

Neonates admitted to the NICU from March 12th, 2010-November 26th, 2013 who were prescribed gentamicin to treat a documented or presumed infection and received > 48 h of gentamicin were identified from a hospital electronic database [31]. Patients with at least one set of steady state gentamicin serum concentrations (trough and peak concentrations obtained at the earliest before and after the third dose of a given dosing regimen, respectively) with documentation of gentamicin administration and serum sampling times were included.
Neonates were excluded if they developed acute renal failure (urine output < 1 mL/kg/hr. or serum creatinine [sCr] > 100 μmol/L) before or during gentamicin therapy, had an increase in sCr > 25% from baseline during treatment, or had a calculated gentamicin half-life > two standard deviations (SDs) from the mean half-life observed in the study population following data analysis, without the availability of an additional set of serum concentrations to confirm the accuracy of this calculated half-life.

Gentamicin dosing and sampling procedure

At the time of this study, neonatal SHSC conventional gentamicin dosing recommendations aimed to target a peak and trough serum concentration of 5–10 mg/L and ≤ 2 mg/L, respectively. (Appendix 1).

Gentamicin pharmacokinetics

The PK profile of gentamicin in neonates has been previously described using one [14, 15, 1719, 21, 24], two [20, 25, 29] and three [28] compartment models. Once gentamicin distribution is complete, it follows first order elimination [15, 20, 28]. Therefore, a one compartment model is appropriate to evaluate the post-distribution pharmacokinetics of gentamicin. Gentamicin concentrations were analyzed using first order PK principles to calculate extrapolated gentamicin trough and peak, elimination rate constant (ke), half-life (t1/2), volume of distribution (Vd), clearance (Cl), initial estimated dose (mg/kg, rounded to nearest 0.5 mg) and dosing interval for conventional (trough ≤ 2 mg/L and peak 5-10 mg/L) and EID (trough ≤ 0.5 mg/L and peak 8-20 mg/L, 12-20 mg/L, 15-20 mg/L and > 20 mg/L) using an infusion time of 1 h. (Appendix 2) When multiple sets of gentamicin serum concentrations were obtained from the same patient, each set was evaluated independently for inclusion, and if eligible, was included as a separate sample for the PK analysis along with the corresponding post-natal age (PNA) and corrected GA (CGA) at time of gentamicin initiation; weight closest to gentamicin initiation; and weight within 24 h of gentamicin levels.

Microbiological cultures

Data for all positive bacterial isolates along with the culture source were extracted from the hospital electronic data base and patient charts.

Statistical analysis

Descriptive statistics were used for patient characteristics and microbiological results (number, percent, mean, SD and range). Since PK parameters display a lognormal distribution, the geometric mean, 95% confidence interval (CI) and range were reported for ke, t1/2, Vd, and Cl.
The data consisted of 60 neonates, of which only 4 had a second set of data with gentamicin levels. This sample size, along with the limited number of repeated measures, was insufficient to run a robust hierarchical model. To circumvent this problem, only data from the first set of gentamicin levels were included for the analyses. Clinical parameters that would have been known prior to the initiation of gentamicin, were not calculated using other parameters input into the regression analysis and were parameters with values available for > 80% of the gentamicin levels (GA at birth; CGA at gentamicin initiation; PNA at gentamicin initiation; gender; BW; weight at gentamicin initiation; Apgar score at one and 5 min of age; blood urea nitrogen [BUN] closest to gentamicin initiation, sCr closest to gentamicin initiation, 24 h urine output [ml/hr], and albumin closest to gentamicin initiation; use of concomitant nephrotoxins [indomethacin, ibuprofen, furosemide, amphotericin B, vancomycin]; and small-for-gestational age [SGA; i.e neonates with a birth weight below the 10th percentile for neonates of the same GA] status) were input in the regression analysis. Variables that were significant (p <  0.05) with bivariate analysis and had a tolerance statistic of ≥0.4 when assessed for multicollinearity were included in a multivariable linear regression (MLR) model to identify those that remained significant using a p <  0.05. Analyses were run using SAS Version 9.4 (SAS Institute, Cary, NC, USA).
A Classification and Regression Tree (CART) analysis (CART1 Professional Extended Edition, Salford Systems, San Diego, California) was used to identify whether practical breakpoints existed for statistically significant MLR-identified covariates of gentamicin Cl (L/h) and Vd (L). The initial CART analyses input all statistically significant variables identified in the MLR analyses for Vd (L) and/or Cl (L/h). CART analyses for Cl and Vd were pruned to the simplest tree, utilizing forced splits to identify clinically practical breakpoints, with the lowest relative error. Forced splits were selected as practical rounded breakpoints derived from the CART identified breakpoint and which had equal or lower relative error than the CART identified breakpoint. The optimal CART model was that which allowed for the fewest sub-groups and had the lowest relative error. CART-identified breakpoints for covariates of gentamicin Vd and/or Cl were used to create patient sub-groups. Mean pharmacokinetic data were calculated for each identified sub-group and the sub-groups were compared to verify the existence of a significant difference in pharmacokinetic parameters (ke [h− 1], Vd [L/kg], and Cl [L/h/kg]) to confirm the validity of the CART-identified breakpoints. An analysis of variance (ANOVA) with Tukey-Kramer Multiple Comparisons Test for data that passed the test for normality or a Kruskal-Wallis Test with Dunn’s Multiple Comparison Test for data that did not pass the test for normality (if > 2 sub-groups were identified) or unpaired t-test (if 2 sub-groups were identified) were used to compare the sub-groups (GraphPad InStat version 3.05, 32-bit for Win95/NT; GraphPad Software Inc., La Jolla, California) for differences in pharmacokinetic parameters with a p <  0.05 considered statistically significant.
Mean PK data of each sub-group were used to explore initial dosing recommendations using first order PK equations for a suggested dose and interval based on inputs for the desired peak and trough concentrations with an infusion time of 1 h. The exploratory gentamicin dose and intervals were subsequently evaluated using Monte Carlo simulation (Oracle Crystal Ball, version 11.1.2.4.000, 32-bit for Windows, Redwood City, California) (MCS). The mean and SDs for ke, Vd, and weight for each determined patient subgroup were input with one million iterations to determine the probability of attaining target steady state peak gentamicin concentrations of 5–10 mg/L, 8–12 mg/L, 8–15 mg/L, 8–20 mg/L, 12–20 mg/L, 15–20 mg/L and > 20 mg/L, as well as target trough concentrations of ≤2 mg/L and ≤ 0.5 mg/L with any given dosing simulation. For the purpose of the MCSs, ke and Vd were assigned a lognormal distribution; weight was assumed to have a triangular distribution and was truncated at the value corresponding to the CART analysis breakpoint for weight for the given sub-group. The upper and lower limits for weight selection were truncated at 4 kg and 0.3 kg, respectively, to reflect values above and below which would be improbable for surviving neonates (< 0.3 kg) and would be greater than 2 SDs from the mean of any sub-group weight category. As part of each MCS, an assessment of the probability of attaining a Peak:MIC ratio of ≥8 was completed. The MIC was assumed to have a normal distribution truncated at a minimum of 0.5 mg/L and maximum of 8 mg/L (Clinical and Laboratory Standards Institute breakpoint for intermediate susceptibility of Enterobactereaceae to gentamicin [32]) with a mean MIC90 of 2 mg/L and SD of 1 mg/L, resembling the current MIC distribution for E coli in Canadian pediatric patients [33].

Results

Demographics

Of a total of 99 patients for whom there was documentation of therapeutic drug monitoring (TDM), 60 patients were eligible for study inclusion to complete the pharmacokinetic analysis (Fig. 1 and Table 1). Patients with a rise in sCr of > 25% during gentamicin therapy were excluded and represent patients who developed nephrotoxicity while on gentamicin (8/99 patients (8%)); recognizing that nephrotoxicity may have been multifactorial and no assumptions can be made about causation associated with gentamicin in this retrospective study (Fig. 1).
Table 1
Patient Characteristics
Patient Demographics Based on Number of Patients = 60
Mean ± Standard Deviation (Range)
Number (%)
Gender (Female)
 
24 (40)
Gestational Age at Birth (Weeks)
27 ± 3 (23–36)
 
Birth Weight (g)
990 ± 482 (488–2740)
 
Small for Gestational Age Status (Yes)
 
1 (2)
Place of Birth (Outborn)
 
15 (25)
Apgar Score at 1 minutea
5 (1–9)
 
Apgar Score at 5 minutea
7 (2–9)
 
Neonatal Intensive Care Unit Survival
 
56 (93)
Gentamicin Treatment (Based on Number of Sets of Gentamicin Levels = 64)
 Post-Natal Age at Gentamicin Initiation (Days)
10 ± 12 (1–46)
 
 Corrected Gestational Age at Gentamicin Initiation (Weeks)
28 ± 3 (24–36)
 
 Weight at Gentamicin Initiation(g)
1059 ± 496 (488–2789)
 
 Gentamicin Dose (mg/kg/dose)
3.0 ± 0.7 (2–5.6)
 
 Gentamicin Dosing Interval (Hours)a
24 (12–36)
 
 Duration of Gentamicin Therapy (Days)
7 ± 2 (2–13)
 
Indication for Antibiotic Therapyb (Based on Number of Patients = 60)
 Culture Negative Sepsis
 
30 (50)
 Necrotizing enterocolitis Septic Ileus
 
10 (17)
 Respiratory Tract Infection
 
6 (10)
 Empiric Treatment (<  5 days)
 
5 (8)
 Confirmed Sepsis
 
4 (7)
 Urinary Tract Infection
 
4 (7)
 Meningitis
 
2 (3)
 Skin and Soft Tissue Infection
 
1 (2)
 Other
 
1 (2)
Laboratory Parameters (Closest to and BEFORE Gentamicin Start Date, unless otherwise noted)
(Based on Number of Sets of Gentamicin Levels = 64)
 Serum creatinine (μmol/L)
57 ± 21 (24–93)
 
 Maximum serum creatinine during Gentamicin (μmol/L)
64 + 23 (19–100)
 
 Blood Urea Nitrogen (mmol/L)
8 ± 5 (2–24)
 
 Maximum blood urea nitrogen during Gentamicin (mmol/L)
11 ± 5 (3–26)
 
 24-h urine output (ml/kg/hr)
4 ± 1 (1–7)
 
 Lowest 24-h urine output during Gentamicin (ml/kg/hr)
3 ± 2 (1–19)
 
 Trough gentamicin concentration (mg/L)c
1.1 ± 0.6 (0.2–3.9)
 
 Peak gentamicin concentration (mg/L)c
7.1 ± 2.2 (3.7–17.1)
 
Nephrotoxins & Ototoxinsd (Based on Number of Sets of Gentamicin Levels = 64)
Concomitant Nephrotoxins during a course of gentamicin
 
43 (67)
Concomitant Ototoxins during a course of gentamicin
 
44 (69)
 Vancomycine,f
 
25 (39)
 Indomethacine,f
 
21 (33)
 Furosemidee,f
 
3 (5)
 Amphotericin Be
 
3 (5)
 Ibuprofene,f
 
1 (2)
 Erythromycinf
 
2 (3)
Prior course of Gentamicin
 
30 (47)
aMedian reported since apgar scores are ordinal data and standard dosing gentamicin intervals were used (e.g. every 12, 24, or 36 h), therefore, gentamicin dosing interval data are ordinal
bThree patients with 2 sets of gentamicin levels had a different diagnosis for each set of gentamicin levels. Therefore, the sum (%) of total indications is greater than 60 (100%) (i.e. 63 (105%))
cExtrapolated concentration using first order pharmacokinetics
dEach course of gentamicin may have had greater than one nephrotoxin or ototoxin, therefore, sum of individual nephrotoxins and ototoxins is greater than the total number of courses of gentamicin with a concomitant nephrotoxin or ototoxin
eNephrotoxin
fOtotoxin
Forty-five of the 60 neonates (75%) included in this study were born at ≤28 weeks gestation. The mean (± standard deviation (SD), range) GA of neonates at birth and CGA at gentamicin initiation were 27 (± 3, 23–36) weeks and 28 (± 3, 24–36) weeks, respectively. Thirty-nine patients (65%) had a BW of < 1000 g (defined as extremely low BW [34]) and 55 patients (92%) had a BW of < 1500 g (defined as very low BW [34]). In this cohort, gentamicin was most commonly used for the treatment of culture negative sepsis (30/60; 50%). Forty-four percent (16/36) of all bacterial isolates were gram-negative bacteria (GNB), most commonly Escherichia coli (7/36; 19%) and Klebsiella spp (5/36; 14%) (Table 2).
Table 2
Bacterial Isolates Cultured at Time of Gentamicin Initiation
Number of gentamicin treatment courses
64 (4 patients had 2 separate gentamicin treatment courses)
Number of gentamicin treatment courses with a positive culture (%)
24 (37.5)
Number of gentamicin treatment courses that were Polymicrobial (2 or more bacterial isolates) (%)
7 (10.9)
 
Total Number of Isolates n = 36 (%)a
Source of Culture
Blood
Cerebrospinal Fluid
Endotracheal Tube
Urine
Eye
Skin
Escherichia coli
7 (19)
4 (11)
1 (3)
1 (3)
1 (3)
0
0
Klebsiella species
5 (14)
1 (3)
0
2 (6)
2 (6)
0
0
Pseudomonas species
2 (6)
0
0
1 (3)
0
1 (3)
0
Enterobacter species
1 (3)
0
0
0
0
0
1 (3)
Raoultella species
1 (3)
0
0
0
0
0
1 (3)
Gram-positive organismsb
15 (42)
7 (19)
1 (3)
1 (3)
3 (9)
1 (3)
2 (6)
Otherc
5 (14)
0
0
5 (14)
0
0
0
a All percentages are determined from total isolates (n = 36)
b Total of 15 g positive organisms include coagulase-negative Staphylococcus (12); Enterococcus species (1); Staphylococcus aureus (1) Group B Streptococcus (1)
cTotal of 5 ‘Other’ organisms include Mycoplasma spp (1) and Ureaplasma urealyticum (4)

Bivariate and multivariable analyses

Significant predictors (p < 0.05) of gentamicin Vd (L) and Cl (L/h) from the bivariate screen and multivariable model are detailed in Table 3. The only covariate that remained significant following MLR for Vd (L) was weight at gentamicin initiation (P < 0.0001). Covariates that remained significant following MLR for Cl (L/h) were PNA at gentamicin initiation (p = 0.0001), gender (p = 0.0447), and weight at gentamicin initiation (p < 0.0001).
Table 3
Bivariate and Multivariable Analysis
Parametera
Clearance (L/h)
Volume of Distribution (L)
Bivariate p-value
Multivariable
p-value
Bivariate p-value
Multivariable p-value
Post-natal age (Days) at gentamicin initiation
< 0.0001
0.0001
0.0037
0.0563
Gender
0.0311
0.0447
0.0667
Weight at gentamicin initiation (g)
< 0.0001
< 0.0001
< 0.0001
<  0.0001
Blood urea nitrogen at baseline (mmol/L)
< 0.0001
0.5855
< 0.0001
0.6643
Serum creatinine at baseline (μmol/L)
< 0.0001
0.0569
0.0011
0.4553
Concomitant nephrotoxins
0.0154
0.9332
0.0279
0.8204
aBaseline values needed to be reported within 14 days prior to the initiation of gentamicin; if unavailable, first value taken during course of gentamicin was used as a surrogate
Bold data indicates statistically significant p-values for a given parameter with either bivariate or multivariable analysis

CART analysis

The optimal CART analyses for Vd(L) and Cl(L/h) produced breakpoints based on the patients’ weight at gentamicin initiation, with a forced split at ≤ 850 g, > 850 g – 1200 g, and > 1200 g. These breakpoints provided the simplest trees with the lowest relative error (Relative Error for Vd tree = 0.347; Relative Error for Cl tree = 0.344). CART identified trees and breakpoints for other parameters in the MLR regression equations (PNA and gender) did not exist.
The mean ke and Cl (L/h/kg) for neonates ≤ 850 g were significantly different from the other weight breakpoints (Table 4). Mean pharmacokinetic parameters for neonates weighing 851 - 1200 g versus > 1200 g were not statistically different (p > 0.05) (Table 4). The small number of participants (n = 13, with 15 gentamicin levels), limited weight range (1210-2789 g; mean 1744 g) and wide confidence intervals of the mean calculated pharmacokinetic parameters in the > 1200 g weight sub-group caused concern regarding the robustness of any dosing recommendations derived for this weight sub-category. As a result, the > 1200 g weight sub-category of neonates was excluded from further analyses. The significant difference in both ke and Cl (L/h/kg) between the ≤850 g and 851-1200 g sub-groups (Table 4), and absence of CART identified trees and breakpoints for the other MLR equation covariates (PNA and gender) supports the use of the simple weight range breakpoints of ≤850 g and 851-1200 g as the sub-groups for practical and convenient empiric gentamicin dosing calculations in neonates.
Table 4
Mean pharmacokinetic parameters
 
≤  850 g
851–1200 g
>  1200 g
p-valuea
n = 25 gentamicin levels in 25 patientsb
n = 24 gentamicin levels in 23 patientsb
n = 15 gentamicin levels in 13 patients
Overall p-value
≤ 850 g vs 851 − 1200 g
≤ 850 g vs > 1200 g
851 − 1200 g vs > 1200 g
Mean
95% Confidence Interval
Range
Mean
95% Confidence Interval
Range
Mean
95% Confidence Interval
Range
Elimination rate constant (h−1)
0.06415
0.05762–0.07068
0.0456–0.1139
0.09087
0.08447–0.09728
0.0652–0.1327
0.09734
0.08519–0.10948
0.05693–0.14332
< 0.0001
< 0.001
< 0.001
> 0.05
Half-life (h)
10.8
9.8–11.8
6.1–15.2
7.6
7.1–8.2
5.2–10.6
7.1
6.0–8.2
4.8–12.2
Volume of distribution (L)
0.36
0.33–0. 39
0.22–0.51
0.51
0.46–0.57
0.33–0.87
0.88
0.76–1.00
0.53–1.23
   
Volume of distribution (L/kg)
0.55
0.50–0.60
0.35–0.83
0.50
0.46–0.54
0.38–0.76
0.52
0.43–0.61
0.26–0.96
0.2471
Clearance (L/h)
0.023
0.021–0.025
0.016–0.038
0.047
0.041–0.053
0.028–0.086
0.086
0.070–0.101
0.040–0.137
Clearance (L/h/kg)
0.035
0.032–0.038
0.026–0.056
0.045
0.041–0.049
0.032–0.067
0.050
0.043–0.058
0.028–0.081
< 0.0001
< 0.01
< 0.001
> 0.05
Multiple Comparison Test for data that did not pass the test for normality
aANOVA with Tukey-Kramer Multiple Comparisons Test for data that passed the test for normality or a Kruskal-Wallis Test with Dunn’s Multiple Comparison Test for data that did not pass the test for normality
bOne patient contributed 1 set of gentamicin levels to weight categories ≤ 850 g and 851-1200 g
Bold data indicates statistically significant p-values for a given parameter with either bivariate or multivariable analysis

Monte Carlo simulation

MCS of weight-based dosing regimens were performed for neonates weighing ≤ 850 g (Table 5) and those weighing between 851 and 1200 g (Table 6). The MCS-identified optimal practical dosing regimens for conventional peaks (5–10 mg/L) and troughs (≤ 2 mg/L) were: 3.5 mg/kg given iv q48h in neonates weighing ≤ 850 g (probability of target peak and trough attainment of 86 and 100%, respectively) and q24h in neonates weighing 851 – 1200 g (probability of target peak and trough attainment of 91 and 97%, respectively). The MCS-identified optimal practical dosing regimens to produce higher peak concentrations of 12–20 mg/L and undetectable trough concentrations (≤ 0.5 mg/L) were: 8-9 mg/kg dose given iv q72h in neonates weighing ≤ 850 g (probability of target peak and trough attainment of > 73 and > 85%, respectively) and given q48h in neonates between 851 and 1200 g (probability of target peak and trough attainment of > 75 and > 84%, respectively).
Table 5
Monte Carlo Simulation Results for Neonates Weighing ≤ 850 g
Dosing Regimen
Target Peak Serum Concentration (mg/L)
Target Trough Serum Concentration (mg/L)
Peak:Minimum Inhibitory Concentration Ratio
Dose (mg/kg)
Dosing Interval (h)
5–10
12–20
15–20
≥ 20
≤ 2
≤ 0.5
≥ 8
2.5a
24
68.76%
0.12%
0.01%
0.00%
81.16%
5.18%
4.21%
3.5a
24
80.45%
4.61%
0.51%
0.01%
58.24%
1.68%
11.26%
3.0
48
69.05%
0.02%
0.00%
0.00%
99.89%
78.65%
3.26%
3.5b
48
86.31%
0.27%
0.00%
0.00%
99.74%
71.89%
6.35%
4.0
48
87.81%
1.53%
0.07%
0.00%
99.45%
65.51%
9.30%
4.5
48
78.22%
5.12%
0.40%
0.00%
99.01%
59.83%
12.54%
5.0
48
62.65%
12.44%
1.53%
0.02%
98.42%
53.62%
16.18%
5.5
48
45.62%
23.47%
4.10%
0.12%
97.69%
49.95%
20.08%
6.0
48
30.54%
36.89%
8.66%
0.42%
96.76%
45.74%
24.21%
6.5
48
18.92%
50.59%
15.39%
1.13%
95.72%
41.94%
28.74%
7.0
48
11.15%
62.11%
23.60%
2.61%
94.51%
38.77%
33.22%
7.5
48
6.10%
70.60%
32.27%
5.19%
93.14%
35.73%
37.96%
8.0
48
3.16%
74.94%
39.79%
9.10%
91.71%
32.96%
42.66%
8.0c
72
4.62%
73.37%
35.31%
5.94%
99.84%
87.65%
39.55%
8.5c
72
2.41%
76.38%
42.25%
10.04%
99.79%
86.31%
43.92%
9.0c
72
1.10%
75.99%
47.24%
15.56%
99.73%
84.88%
48.44%
9.5
72
0.45%
72.46%
49.45%
22.35%
99.65%
83.46%
52.81%
10
72
0.10%
66.97%
49.27%
30.06%
99.55%
82.07%
56.95%
aDosing regimens recommended at Sunnybrook at time of study: ≤ 27 weeks corrected gestational age (CGA): 2.5 mg/kg q24h; 28–32 weeks CGA: 3.5 mg/kg q24h; 33–34 wks CGA: 4.5 mg/kg q24h
bRecommended dosing to target gentamicin concentrations: Peak 5-10 mg/L and Trough < 2 mg/L
cRecommended dosing to target gentamicin concentrations: Peak 12-20 mg/L and Trough ≤ 0.5 mg/L
Table 6
Monte Carlo Simulation Results for Neonates Weighing Between 851 and 1200 g
Dosing Regimen
Target Peak Serum Concentration (mg/L)
Target Trough Serum Concentration (mg/L)
Peak: Minimum Inhibitory Concentration Ratio
Dose (mg/kg)
Dosing Interval (h)
5–10
12–20
15–20
≥ 20
≤ 2
≤ 0.5
≥ 8
2.5a
24
66.21%
0.00%
0.00%
0.00%
99.66%
27.12%
3.27%
3.5a
24
91.17%
0.83%
0.02%
0.00%
96.57%
9.94%
9.39%
2.5
24
66.21%
0.00%
0.00%
0.00%
99.66%
27.12%
3.27%
3.0
24
90.28%
0.06%
0.00%
0.00%
98.68%
16.37%
6.08%
3.5b
24
91.17%
0.83%
0.02%
0.00%
96.57%
9.94%
9.39%
4.0
24
77.21%
4.42%
0.21%
0.00%
93.13%
6.17%
13.23%
4.5
24
55.29%
13.88%
1.23%
0.01%
88.39%
4.01%
17.50%
4.5
36
72.05%
5.81%
0.27%
0.00%
99.96%
69.44%
14.27%
5.0
36
50.61%
15.89%
1.38%
0.00%
99.90%
62.85%
18.27%
5.5
36
30.74%
31.43%
4.51%
0.05%
99.80%
56.50%
22.73%
6.0
36
16.46%
49.27%
10.87%
0.28%
99.66%
50.67%
27.44%
6.5
36
7.96%
65.32%
20.63%
0.96%
99.42%
45.56%
32.25%
7.0
36
3.50%
76.80%
32.38%
2.61%
99.08%
40.71%
37.26%
7.5
36
1.43%
82.50%
43.50%
5.85%
98.62%
36.70%
42.38%
8.0
36
0.56%
82.76%
52.04%
11.15%
98.06%
32.80%
47.42%
8.5
36
0.21%
78.31%
56.09%
18.62%
97.40%
29.56%
52.36%
9.0
36
0.07%
70.46%
55.41%
28.09%
94.61%
21.62%
66.33%
7.5
48
14.52%
81.31%
39.39%
4.12%
99.99%
89.88%
40.26%
8.0c
48
7.95%
83.70%
49.22%
8.35%
99.99%
88.06%
45.22%
8.5c
48
4.07%
81.27%
55.18%
14.72%
99.98%
86.11%
50.20%
9.0c
48
2.00%
74.96%
56.71%
23.04%
99.96%
84.10%
54.91%
9.5
48
0.94%
66.18%
54.11%
32.88%
99.95%
82.15%
59.48%
10
48
0.43%
56.10%
48.53%
43.50%
99.93%
80.12%
63.87%
aDosing regimens recommended at Sunnybrook at time of study: ≤ 27 weeks corrected gestational age (CGA): 2.5 mg/kg q24h; 28–32 weeks CGA: 3.5 mg/kg q24h; 33–34 wks CGA: 4.5 mg/kg q24h
bRecommended dosing to target gentamicin concentrations: Peak 5-10 mg/L and Trough < 2 mg/L
cRecommended dosing to target gentamicin concentrations: Peak 12-20 mg/L and Trough ≤ 0.5 mg/L

Discussion

This retrospective pharmacokinetic study evaluated hospitalized neonates with normal renal function, and a median CGA at gentamicin initiation of < 28 weeks. Seventy-five percent of those included were born at ≤ 28 weeks gestation and 92% had a BW of < 1500 g. Gentamicin Cl (L/h) and Vd (L) were significantly associated with weight at gentamicin initiation (≤ 850 g, 851-1200 g, and > 1200 g). Since no significant difference in pharmacokinetics existed for neonates weighing > 1200 g versus 851-1200 g, due to inadequate sample size in the largest weight category, we did not explore the > 1200 g sub-group further. No CART identified trees with breakpoints for the other MLR equation covariates (PNA and gender) existed. Based on the absence of CART identified trees and breakpoints for PNA and gender and the identification of a significant difference in both ke and Cl (L/h/kg) between the ≤850 g and 851-1200 g sub-groups, the use of the simple weight range breakpoints of ≤850 g and 851-1200 g as the sub-groups for practical and convenient empiric gentamicin dosing calculations in neonates is rational. Dosing of 3.5 mg/kg/dose administered every 48 h for neonates weighing ≤ 850 g, and every 24 h for neonates weighing 851-1200 g provided the best probability of attaining conventional targets (peak:5-10 mg/L, trough:≤ 2 mg/L). Dosing of 8-9 mg/kg/dose administered every 72 h in neonates weighing ≤ 850 g and every 48 h in neonates weighing 851-1200 g provided the best probability of attaining EID targets (peak:12-20 mg/L, trough:≤ 0.5 mg/L).
The strengths of our study include the determination of gentamicin pharmacokinetics in a large sample of premature and low-birth weight neonates for whom data are currently lacking; the identification of significant covariates for Vd and Cl with determination of practical weight breakpoints; the utilization of MCS with 1 million iterations to develop simple initial gentamicin dosing nomograms for both conventional and EID for low-birth weight neonates with an excellent probability of target peak and trough attainment; and the provision of tables itemizing probabilities of target attainment (including Peak:MIC ratio) for a range of potential dosing options enabling institutional selection of initial dosing guidelines based on their GNB susceptibility patterns and desired target serum concentrations. In addition, our rigorous study design which limited the inclusion of gentamicin levels to those with a confirmed time for dose administration and serum sampling increases the validity of our results.
The weaknesses of our study include its retrospective design and associated risk of unrecognized confounders; the inability to generalize our results to neonates > 1200 g and SGA infants; and the risk of incomplete gentamicin distribution at time of sampling for peak concentrations. However, since our mean pharmacokinetic parameters were comparable to those reported in other studies [15], our sampling practice is unlikely to have affected the validity of our results.
Similar to other pharmacokinetic studies, our multivariable analysis indicated that the Vd of gentamicin in neonates is associated with body weight [15, 1820, 28, 29]. Pharmacokinetic studies have identified that extracellular fluid volume correlates closely with bodyweight [35].
Our multivariable analysis indicated that gentamicin clearance in neonates is associated with PNA, as well as bodyweight, and gender. The correlation between PNA and gentamicin elimination has been previously reported in the literature [19, 28, 29], and is explained by the maturation of renal function in neonates. Since glomerulogenesis proceeds until 32–34 weeks gestation, preterm neonates are expected to have a reduced rate of glomerular filtration compared to their mature counterparts [36]. In the first 48–72 h of life there is a marked increase in glomerular filtration rate of full term newborns to rates of 8–20 ml/min, compared with increases in preterm neonates of only 2–3 ml/min [35, 37]. The half-life of elimination of gentamicin is therefore expected to decrease with increasing PNA because it is renally eliminated [37], as evidenced in our study. In addition, bodyweight likely serves as a surrogate marker for physiological maturity. Therefore, it is expected that the half-life of elimination of gentamicin decreases as body weight increases. This relationship was demonstrated in our study, as well as in previously published literature [15, 1820, 29].
CART analysis confirmed breakpoints for weight at gentamicin initiation for both Vd and Cl and demonstrated that neonates had altered Vd (L) and Cl (L/h) based on these weight breakpoints. This allowed the use of the CART derived weight breakpoints (≤850 g and 851-1200 g) to divide our data into homogenous patient sub-groups for practical empiric gentamicin dosing recommendations and provides a new and convenient nomogram for gentamicin dosing (either conventional or EID) with a MCS demonstrated high probability of target attainment. The mean gentamicin Vd (0.55 L/kg and 0.50 L/kg for neonates weighing ≤ 850 g and 851-1200 g, respectively) and Cl (0.035 L/h/kg and 0.045 L/h/kg, for neonates weighing ≤ 850 g and 851-1200 g, respectively) identified in this study are comparable to those reported in a study of infants born at less than 28 weeks gestation (Vd = 0.50 L/kg and Cl = 0.032 L/h/kg) [15].
Our study confirms previous reports [2, 27, 38] that GNB, particularly E coli, are emerging as the leading cause of systemic infections in neonates. Recent microbiological reports of E coli isolates from Canadian pediatric patients report a mean MIC90 of 2 mg/L for gentamicin [33]. Therefore, to meet the PK/PD target of a peak: MIC ratio between 8 and 10, peak gentamicin concentrations should range from 16 to 20 mg/L. A single published study approximates these recommendations by targeting a peak concentration of 15–20 mg/L in neonates [19]. In this study, initial doses of 10 mg/kg administered at 36 h intervals were used in term newborns and 12 mg/kg doses administered every 48 h were used in premature neonates (GA 31–38 weeks) [19]. Our MCS derived initial EID recommendations for gentamicin of 8-9 mg/kg/dose administered every 72 h in neonates weighing ≤ 850 g and every 48 h in neonates weighing 851-1200 g has > 73% probability of attaining a peak between 12 and 20 mg/L and > 84% probability of attaining a trough of ≤ 0.5 mg/L. Our work is further supported by results from a recent study concluding that a prolonged dosing interval for gentamicin ranging from 36 to 72 h was appropriate for neonates weighing less than 1000 g [25]. However, our results provide a new easy to use gentamicin dosing nomogram for both conventional and EID gentamicin with a MCS demonstrated high probability of target attainment, which has not previously been completed for neonates. In all cases the weight based initial dosing recommendations derived in our study provided a better probability of target attainment than the CGA-based gentamicin dosing regimens used at our institution at the time of this study conduct. In 2014 our centre changed its gentamicin dosing practice to adopt the weight based nomogram developed in this study; where EID is now predominantly used for NICU babies. We have received positive feedback about the simplicity, safety and efficacy of the nomogram from our NICU physicians and pharmacists. Plans are underway to evaluate the safety, efficacy and health care personnel workload of the weight based nomograms for conventional and EID using a pragmatic study design.
Although the study by Lanao et al [19] was published in 2004, higher peak concentration targets have not been routinely adopted by clinicians. Therefore, we chose to report the probabilities of achieving a range of peak gentamicin concentrations with various dosing regimens because GNB MICs, along with desired target peak concentrations, may vary among hospitals. Our MCS dosing tables may assist clinicians in choosing a gentamicin dosing regimen that would be optimal based on their institutional MIC patterns for relevant GNB, such as E. coli.

Conclusions

The study contributes new data based gentamicin dosing guidelines for both initial conventional and EID in neonates ≤ 1200 g, a patient population under-represented in neonatal studies and for whom limited data exists for gentamicin dosing. Our results provide clinicians with practical and simple initial dosing recommendations based on weight at time of gentamicin initiation with a high probability of target peak and trough attainment. Confirmatory gentamicin levels (peak and trough with third dose for conventional therapy and a peak and 8–12 h post level with the first dose of EID) are recommended to further refine dosing. If more prolonged therapy is needed, then repeat levels are recommended to identify changes in the neonate’s gentamicin pharmacokinetics with PNA and weight. The gentamicin levels that were targeted in this study reflect accepted safe and effective levels for gentamicin in neonates [1, 2, 6, 1126]. However, due to the retrospective design of our study, a prospective pharmacokinetic clinical study in neonates ≤ 1200 g is needed to confirm the efficacy and safety of the gentamicin EID nomogram recommendations.

Acknowledgements

Dr. Alex Kiss, Research Design and Biostatistics, Sunnybrook Health Sciences Centre for his assistance with the statistical analysis.
This study was approved by the Sunnybrook Health Sciences Centre (SHSC) Research Ethics Board on February 3, 2014 (Project Identification Number 038–2014), and was conducted in accordance with the Declaration of Helsinki and national and institutional standards.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Appendix 1

Table 7
Sunnybrook Health Sciences Centre’s Dosing Recommendations for Gentamicin in Neonates at Time of Study (2013)
Corrected Gestational Age [Weeks]
Dose [mg/kg]
Dosing Interval [Hours]
Administration Technique
Serum Concentration Sampling
< 28
2.5
24
1 h infusion
Immediately following end of infusion
28–32
3.5
24
1 h infusion
Immediately following end of infusion
33–34
4
24
1 h infusion
Immediately following end of infusion
35–36
2
12
Intravenous bolus
30 min following bolus
>  36
2.5
12
Intravenous bolus
30 min following bolus

Appendix 2

First Order Pharmacokinetic Equations:
1.
\( \mathrm{k}=\frac{\hbox{-} \left(\ln\ {\mathrm{C}}^{\mathrm{obs}.\min}\hbox{-} \ln\ {\mathrm{C}}^{\mathrm{obs}.\max}\right)}{\mathrm{t}\mathrm{min}-\mathrm{tmax}\kern0.75em };{\mathrm{t}}_{1/2}=0.693/\mathrm{k} \)
 
2.
\( {\mathrm{C}}^{\operatorname{ext}.\max }=\frac{{\mathrm{C}}^{\mathrm{obs}.\max }}{{\mathrm{e}}^{\hbox{-} \mathrm{ktmax}}} \)
 
3.
\( {\mathrm{C}}^{\operatorname{ext}.\min }={\mathrm{C}}^{\operatorname{ext}.\max}\mathrm{x}\ {\mathrm{e}}^{\hbox{-} \mathrm{k}\left(\mathrm{t}\hbox{-} \mathrm{t}\hbox{'}\right)} \)
 
4.
\( \mathrm{V}=\frac{\mathrm{Dose}\ \mathrm{x}\ \left[1\hbox{-} {\mathrm{e}}^{\hbox{-} \mathrm{kt}\hbox{'}}\right]}{\mathrm{kt}\hbox{'}\kern0.5em \left[{\mathrm{C}}^{\operatorname{ext}.\max}\hbox{-} {\mathrm{C}}^{\operatorname{ext}.\min}\mathrm{x}\ {\mathrm{e}}^{\hbox{-} \mathrm{kt}\hbox{'}}\right]} \)
 
5.
Suggested Dosing Interval:
 
$$ \mathrm{t}=\frac{\hbox{-} 1/\mathrm{k}\ \ln \left({\mathrm{C}}^{\mathrm{des}.\mathrm{minss}}\right)+\mathrm{t}\hbox{'}}{\left({\mathrm{C}}^{\mathrm{des}.\mathrm{maxss}}\right)} $$
Round to 4, 6, 8, 12, 16, 18, 24 etc.
6.
Suggested Maintenance Dose:
 
$$ \mathrm{Dose}={\mathrm{kVC}}^{\mathrm{des}.\mathrm{maxss}}\mathrm{x}\frac{\left(1\hbox{-} {\mathrm{e}}^{\hbox{-} \mathrm{kt}}\right)\kern0.5em \mathrm{x}\ \mathrm{t}\hbox{'}}{\left(1\hbox{-} {\mathrm{e}}^{\hbox{-} \mathrm{kt}\hbox{'}}\right)} $$
7.
Predicted Peak
 
$$ {\mathrm{C}}^{\mathrm{pred}.\mathrm{maxss}}=\frac{\mathrm{Dose}\ \mathrm{x}\ \left(1\hbox{-} {\mathrm{e}}^{\hbox{-} \mathrm{kt}\hbox{'}}\right)}{\mathrm{t}\hbox{'}\mathrm{kV}\ \mathrm{x}\ \left(1\hbox{-} {\mathrm{e}}^{\hbox{-} \mathrm{kt}}\right)} $$
8.
Predicted Trough
 
$$ {\mathrm{C}}^{\mathrm{pred}.\mathrm{minss}}={\mathrm{C}}^{\mathrm{pred}.\mathrm{maxss}}\left({\mathrm{e}}^{\hbox{-} \mathrm{k}\left(\mathrm{t}\hbox{-} \mathrm{t}\hbox{'}\right)}\right) $$
Key:
  • k= elimination rate constant (h-1) ; t1/2 = half-life (h)
  • Cobs.min & Cobs.max = observed minimum and maximum concentrations
  • tmin & tmax = time post infusion (in hours) of observed minimum and maximum concentrations
  • Cext.min & Cext.max = extrapolated or actual minimum and maximum concentrations
  • V=volume of distribution (L); t = dosing interval in hours ; t' = infusion time in hours
  • Cdes.minss & Cdes.maxss = desired minimum and maximum concentrations at steady state
  • Cpred.minss & Cpred.maxss = predicted minimum and maximum concentrations at steady state
Literatur
6.
Zurück zum Zitat Miron D. Once daily dosing of gentamicin in infants and children. Pediatr Infect Dis J. 2001;20(12):1169–73.CrossRefPubMed Miron D. Once daily dosing of gentamicin in infants and children. Pediatr Infect Dis J. 2001;20(12):1169–73.CrossRefPubMed
13.
Zurück zum Zitat Skopnik H, Heimann G. Once daily aminoglycoside dosing in full term neonates. Pediatr Infect Dis J. 1995;14(1):71–2.CrossRefPubMed Skopnik H, Heimann G. Once daily aminoglycoside dosing in full term neonates. Pediatr Infect Dis J. 1995;14(1):71–2.CrossRefPubMed
17.
Zurück zum Zitat Lundergan FS, Glasscock GF, Kim EH, Cohen RS. Once-daily gentamicin dosing in newborn infants. Pediatrics. 1999;103(6):1228–34.CrossRefPubMed Lundergan FS, Glasscock GF, Kim EH, Cohen RS. Once-daily gentamicin dosing in newborn infants. Pediatrics. 1999;103(6):1228–34.CrossRefPubMed
21.
Zurück zum Zitat Vervelde ML, Rademaker CM, Krediet TG, Fleer A, van Asten P, van Dijk A. Population pharmacokinetics of gentamicin in preterm neonates: evaluation of a once-daily dosage regimen. Ther Drug Monit. 1999;21(5):514–9.CrossRefPubMed Vervelde ML, Rademaker CM, Krediet TG, Fleer A, van Asten P, van Dijk A. Population pharmacokinetics of gentamicin in preterm neonates: evaluation of a once-daily dosage regimen. Ther Drug Monit. 1999;21(5):514–9.CrossRefPubMed
26.
Zurück zum Zitat Alshaikh B, Dersch-Mills D, Taylor R, Akierman AR, Yusuf K. Extended interval dosing of gentamicin in premature neonates ≤ 28-week gestation. Acta Paediatr. 2012;101:1134–9.CrossRefPubMed Alshaikh B, Dersch-Mills D, Taylor R, Akierman AR, Yusuf K. Extended interval dosing of gentamicin in premature neonates ≤ 28-week gestation. Acta Paediatr. 2012;101:1134–9.CrossRefPubMed
31.
Zurück zum Zitat Elligsen M, Walker SAN, Simor A, Daneman N. Prospective audit and feedback of antimicrobial stewardship in critical care: program implementation, experience, and challenges. Can J Hosp Pharm. 2012;65(1):31–6.PubMedPubMedCentral Elligsen M, Walker SAN, Simor A, Daneman N. Prospective audit and feedback of antimicrobial stewardship in critical care: program implementation, experience, and challenges. Can J Hosp Pharm. 2012;65(1):31–6.PubMedPubMedCentral
32.
Zurück zum Zitat CLSI. Performance standards for antimicrobial susceptibility testing; 25th informational supplement. CLSI document M100-S25. Wayne: Clinical and laboratory Standards Institute; 2015. CLSI. Performance standards for antimicrobial susceptibility testing; 25th informational supplement. CLSI document M100-S25. Wayne: Clinical and laboratory Standards Institute; 2015.
33.
Zurück zum Zitat Adam HJ, Baxter MR, Davidson RJ, Rubinstein E, Fanella S, Karlowsky JA, Lagace-Wiens PRS, Hoban DJ, Zhanel GG. Comparison of pathogens and their antimicrobial resistance patterns in pediatric, adult and elderly patients in Canadian hospitals. J Antimicrob Chemother. 2013;68(Suppl 1):i31–7. https://doi.org/10.1093/jac/dkt024.CrossRefPubMed Adam HJ, Baxter MR, Davidson RJ, Rubinstein E, Fanella S, Karlowsky JA, Lagace-Wiens PRS, Hoban DJ, Zhanel GG. Comparison of pathogens and their antimicrobial resistance patterns in pediatric, adult and elderly patients in Canadian hospitals. J Antimicrob Chemother. 2013;68(Suppl 1):i31–7. https://​doi.​org/​10.​1093/​jac/​dkt024.CrossRefPubMed
34.
Zurück zum Zitat World Health Organization. International statistical classification of diseases and related health problems, tenth revision, 2nd ed. World Health Organization; 2004. World Health Organization. International statistical classification of diseases and related health problems, tenth revision, 2nd ed. World Health Organization; 2004.
38.
Zurück zum Zitat Stoll BJ, Hansen N, Fanaroff AA, Wright LL, Carlo WA, Ehrenkranz RA, Lemons JA, Donovan EF, Stark AR, Tyson JE, Oh W, Bauer CR, Korones SB, Shankaran S, Laptook AR, Stevenson DK, Papile L, Poole WK. Changes in pathogens causing early-onset sepsis in very-low-birth-weight infants. N Engl J Med. 2002;347(4):240–7. https://doi.org/10.1056/NEJMoa012657.CrossRefPubMed Stoll BJ, Hansen N, Fanaroff AA, Wright LL, Carlo WA, Ehrenkranz RA, Lemons JA, Donovan EF, Stark AR, Tyson JE, Oh W, Bauer CR, Korones SB, Shankaran S, Laptook AR, Stevenson DK, Papile L, Poole WK. Changes in pathogens causing early-onset sepsis in very-low-birth-weight infants. N Engl J Med. 2002;347(4):240–7. https://​doi.​org/​10.​1056/​NEJMoa012657.CrossRefPubMed
Metadaten
Titel
Optimizing gentamicin conventional and extended interval dosing in neonates using Monte Carlo simulation – a retrospective study
verfasst von
Monique Bergenwall
Sandra A. N. Walker
Marion Elligsen
Dolores C. Iaboni
Carla Findlater
Winnie Seto
Eugene Ng
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2019
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-019-1676-3

Weitere Artikel der Ausgabe 1/2019

BMC Pediatrics 1/2019 Zur Ausgabe

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.