Sei sulla pagina 1di 10

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/258528854

High-dose gentamicin in newborn infants: Is it


safe?

Article in European Journal of Pediatrics November 2013


DOI: 10.1007/s00431-013-2194-1 Source: PubMed

CITATIONS READS

9 479

4 authors, including:

Johannes N van den Anker Claus Klingenberg


Children's National Medical Center University Hospital of North Norway
445 PUBLICATIONS 8,217 CITATIONS 102 PUBLICATIONS 1,047 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

INVESTMENT View project

All content following this page was uploaded by Claus Klingenberg on 04 April 2017.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
High-dose gentamicin in newborn infants:
is it safe?

Jon Widding Fjalstad, Einar Laukli, John


N.van den Anker & Claus Klingenberg

European Journal of Pediatrics

ISSN 0340-6199

Eur J Pediatr
DOI 10.1007/s00431-013-2194-1

1 23
Your article is protected by copyright and
all rights are held exclusively by Springer-
Verlag Berlin Heidelberg. This e-offprint is
for personal use only and shall not be self-
archived in electronic repositories. If you wish
to self-archive your article, please use the
accepted manuscript version for posting on
your own website. You may further deposit
the accepted manuscript version in any
repository, provided it is only made publicly
available 12 months after official publication
or later and provided acknowledgement is
given to the original source of publication
and a link is inserted to the published article
on Springer's website. The link must be
accompanied by the following text: "The final
publication is available at link.springer.com.

1 23
Author's personal copy
Eur J Pediatr
DOI 10.1007/s00431-013-2194-1

ORIGINAL ARTICLE

High-dose gentamicin in newborn infants: is it safe?


Jon Widding Fjalstad & Einar Laukli &
John N. van den Anker & Claus Klingenberg

Received: 4 September 2013 / Accepted: 21 October 2013


# Springer-Verlag Berlin Heidelberg 2013

Abstract Dosing regimens often recommend lower gentami- of life 0.8 (0.6)mg/L. In 31 (6 %) episodes, TPC was 2.0 mg/
cin doses in neonates (35 mg/kg) than in older children L, predominantly among term infants with renal impairment.
(7 mg/kg or more) despite the higher volume of distribution Thirty-eight patients failed the neonatal hearing screening, but
in neonates. We studied an extended-interval high-dose (6 mg/ only four of these 38 had permanent hearing loss. All four had
kg) gentamicin regimen in a single tertiary neonatal unit from a TPC <2.0 mg/L. Conclusions: This extended-interval high-
20042012. During the first week of life, dosing interval was dose gentamicin regimen was associated with low numbers of
24 h for term infants, 36 h for preterm infants with gestational elevated TPCs, low numbers of prescription errors and no
age (GA) 2936 weeks and 48 h for preterm infants with GA evidence for ototoxicity.
<29 weeks. After the first week of life, dosing interval was
24 h if corrected age (GA + postnatal age) 29 weeks and 36 h Keywords Gentamicin . High-dose . Trough plasma
if corrected age <29 weeks. Outcome measures were trough concentration . Ototoxicity . Prescription errors . Neonate
plasma concentration (TPC), ototoxicity and prescription er-
rors. In 546 treatment episodes, TPC was measured prior to
the third gentamicin dose. There were 37 episodes (6.7 %) of Introduction
prescription errors, mainly a too long dosing interval. We
included 509 treatment episodes (440 infants) in the final Gentamicin, in combination with a beta-lactam antibiotic, is
analysis. Mean (standard deviation) gentamicin TPC during widely used for treatment of neonatal sepsis [19, 36]. The
the first week of life was 1.1 (0.5)mg/L and after the first week bactericidal effect of gentamicin is concentration-dependent.
A high ratio between the peak plasma concentration and the
minimum inhibitory concentration of the infecting microor-
J. W. Fjalstad : C. Klingenberg ganism is therefore important for therapeutic response [18,
Paediatric Research Group, Faculty of Health Sciences,
28]. In contrast, the potential ototoxicity and nephrotoxicity is
University of Troms, Troms, Norway
thought to be associated with high trough plasma concentra-
E. Laukli tions (TPCs) and a prolonged duration of therapy [3], but
Department of Ear, Nose and Throat, University Hospital of North many neonatal studies fail to find proof for this association
Norway, Troms, Norway
[6, 8, 16]. It is commonly suggested for neonates that TPC
J. N. van den Anker should be less than 2 mg/L to minimise potential toxic effects
Division of Paediatric Clinical Pharmacology, Childrens National [24, 25, 33].
Medical Center, Washington, DC, USA Gentamicin is distributed in the extracellular water com-
partment. Neonates, and particularly those born prematurely,
J. N. van den Anker
Intensive Care and Department of Paediatric Surgery, Erasmus have a proportionally larger volume of distribution than older
MC-Sophia Childrens Hospital, Rotterdam, The Netherlands children and adults [2, 15, 30]. A higher dose per kilogram is
therefore needed to achieve the same peak concentrations in a
C. Klingenberg (*)
preterm neonate as compared with an older infant or child.
Department of Paediatrics, University Hospital of North Norway,
9038 Troms, Norway Gentamicin clearance correlates with functional maturation of
e-mail: claus.klingenberg@unn.no the glomerular filtration rate (GFR). The nephrogenesis is
Author's personal copy
Eur J Pediatr

completed by 3435 weeks of gestation, and preterm neonates patients, identified from the hospital laboratory database, were
have a lower GFR than term neonates [5, 23]. Even in term neonates up to 50 weeks of corrected age (GA + PNA) that
neonates, the GFR is low at birth but increases rapidly during had received gentamicin treatment for suspected or proven
the first week of life with increased renal blood flow and a infection and had one or more TPCs measured from January
decrease in renal vascular resistance [5, 21]. 2004 through May 2012. The regional ethical committee
Based on these important pharmacokineticpharmacody- approved the study.
namic factors, extended-interval gentamicin dosing regimens
for neonates have been developed and are widely used today Gentamicin dosing regimen and monitoring
[4, 25]. However, many of these dosing regimens do not
differentiate between dosing during the first week of life and We administered 6 mg/kg gentamicin as a 30-min infusion,
later in the neonatal period. Few studies report ideal adherence irrespective of GA and PNA. The dosing intervals during the
to target TPCs [20]. The British National Formulary for Chil- first week of life were 24 h if GA was 37 weeks (group A),
dren recommends gentamicin 45 mg/kg every 24 or 36 h for 36 h if GA was 2936 weeks (group B) and 48 h if GA was
neonates [24]. However, high TPC (>2 mg/L) is frequently <29 weeks (group C). After the first week of life, dosing
seen in moderate preterm infants and among infants with intervals were 24 h if corrected age (GA + PNA) was
severe perinatal asphyxia when using this dosing regimen 29 weeks (group D) and 36 h if corrected age was <29 weeks
[27]. The Neofax suggests a more complex dosing regimen (group E). TPCs were obtained right before the third dose.
with different dosages (45 mg/kg) and intervals depending During the study period, two different immunoassays with a
on maturation [35]. Complicated dosing regimens may pose a lower limit of detection <0.3 mg/L were used to analyse
risk for prescription errors, which again increases the risk of gentamicin plasma concentration (2004-09: GENT2, Roche,
toxicity [34]. It is also a paradox that most dosing regimens Mannheim, Germany, 20102012: CEDIA Gentamicin II
recommend a lower gentamicin dose per kilogram in neonates Assay, Microgenics, Passau, Germany). An internal validation
(35 mg/kg) than in older children (7 mg/kg) despite the fact showed a good correlation between both methods.
that neonates have a larger volume of distribution [15].
We previously validated a netilmicin dosing regimen giv- Data collection
ing 6 mg/kg every 24 or 36 h, depending on gestational age
(GA) and postnatal age (PNA) [12]. We found mean peak Two of the authors (JF and CK) reviewed the medical records
levels >910 mg/L, but too many potential toxic TPC for of all patients. We registered background data (sex, age,
moderate preterm infants [12]. In 2004, we changed our weight, diagnoses and complications incl. acute renal failure)
standard aminoglycoside from netilmicin to gentamicin and and gentamicin TPC. We took extra care to assess medical
developed a new high-dose extended-interval regimen. We staff prescription and evaluated whether dosing (milligrams
continued using the same aminoglycoside dose (6 mg/kg) as per kilogram) and dosing intervals were in line with dosing
pharmacokinetic parameters for netilmicin and gentamicin are protocol. We evaluated nursing staff administration, and for
similar [35], but we increased the dosing interval for the more this study, we defined that a dose given >3 h earlier or later
preterm infants to 48 h. The three major aims for this study than scheduled was an administration error.
were, based on the 8-year experience with this dosing regi-
men, (1) to evaluate gentamicin TPCs to assess pharmacoki- Audiology assessment
netic safety, (2) to assess the potential ototoxicity and (3) to
evaluate the number of prescription errors. Before discharge, all infants were screened for ototoxicity by a
transient-evoked otoacoustic emission (OAE) test (Madsen,
AccuScreen, GN Otometrics, Denmark). Prior to 2007, a risk-
Material and methods based screening approach was used, including all neonates
treated with gentamicin. Since January 2007, OAE has been
Study design and patients implemented as a universal screening test of all newborn
infants. Patients who failed OAE screening had an automatic
This is a retrospective single-centre cohort study. The neonatal auditory brainstem response test as first follow-up test. Further
intensive care unit at the University Hospital of Northern follow-up was then individualised in the audiology unit. We
Norway in Troms is the only tertiary neonatal unit in the carefully reviewed data on hearing for all patients referred for
two most northern counties in Norway covering a population follow-up. An experienced audiologist (EL) reassessed all
of 230,000 with around 3,000 births per year. All infants cases with possible persistent hearing problems. To ensure
<34 weeks gestation and all infants receiving mechanical that no patients with severe ototoxicity were missed, the
ventilation in the catchment area are treated in this unit, so audiologist also identified all children who went on to have
for these infants, our data are population-based. Eligible hearing aids or cochlear implants and were born during the
Author's personal copy
Eur J Pediatr

audit period. Furthermore, all patient at risk for neurological audiology unit. Seventeen had normal hearing on the first
sequelae (GA <32 weeks/birth weight <1,500 g and severe follow-up. Another 17 patients were diagnosed with middle
perinatal asphyxia) were seen at regular intervals in the out- ear problems but did not have any confirmed sensorineural
patient clinic up to 2 years of age, and sensory impairment was hearing loss later. Four patients who failed their OAE test were
recorded. suspected to have permanent hearing loss, and one additional
patient who passed the OAE test later received a cochlear
Statistics implant. Two of these five patients probably have small uni-
lateral hearing losses, two received hearing aids and one
Data were analysed using IBM-SPSS (version 20.0) statistical received a cochlear implant (Table 3). Only 1 out of 31
software. Gentamicin TPCs <0.3 mg/L were assigned a value patients with a TPC 2.0 mg/L suffered a permanent hearing
of 0.2 mg/L. Descriptive results are expressed as mean (stan- loss. This patient, diagnosed with a congenital cytomegalovi-
dard deviation (SD)) or median (interquartile range (IQR)), as rus (CMV) infection, was the one who initially passed the
appropriate. A p value<0.05 was considered significant. OAE test but later developed severe hearing loss and received
a cochlear implant.
Thirty-one of 37 treatment episodes with medical staff
Results prescription errors involved ordering a 12-h too long interval.
Mean (SD) TPC among these was 0.6 (0.4)mg/L, and none
There were 2,247 infants admitted to the neonatal unit during had a TPC 2.0 mg/L. Six treatment episodes were prescribed
the 8-year study period, and 315 infants were discharged with with too short intervals (12 h). Mean (SD) among these was
a diagnosis of culture-positive (n =55) or culture-negative 1.5 (0.9)mg/L, and in two episodes, TPC was 2.0 mg/L
(n =260) neonatal sepsis. We identified 546 treatment epi- (33 %). Due to obviously lower/higher TPCs in episodes with
sodes from 457 neonates who had one or more gentamicin wrong dosing intervals compared to those with correct dosing
TPC registered. There were 37 episodes (37/546; 6.7 %) of interval, we found it correct to exclude these 37 episodes in the
incorrect medical staff prescription, which were excluded final analyses.
from final analyses on TPC and ototoxicity. We included a We identified 81/509 (16 %) episodes with nursing staff
total of 509 treatment episodes (three doses gentamicin) errors regarding timing of administration. Gentamicin was
belonging to 440 patients; 395 patients received one, 36 administered too late in 59 episodes (mean (SD) TPC, 0.9
patients received two and 9 patients received 3 separate treat- (0.4)mg/L) and too early in 22 episodes (mean (SD) TPC, 1.0
ment episodes. For the whole study population, the mean (SD) (0.5)mg/L). The TPCs in these 81 episodes were within the
GA was 36.4 (5.3)weeks and the mean (SD) birth weight was range and not statistically different than the other 428 episodes
2,739 (1,326)gram. There were 85 (19 %) patients with a very (mean (SD) TPC, 1.0 (0.6)mg/L). We decided to include these
low birth weight (<1,500 g) and 61 patients (14 %) with GA 81 episodes in the final analyses as we considered they reflect
<29 weeks. normal deviations in a busy neonatal unit.
Table 1 shows population and outcome data among the
different treatment groups. Patients in groups D and E were
predominantly preterm infants with suspected or proven epi- Discussion
sodes of late onset sepsis. Many of these patients had already
received gentamicin treatment during the first week of life. We This clinical study is to our knowledge the largest study ever
decided to include these patients when describing population to analyse an extended-interval gentamicin dosing regimen in
data of infants treated after first week of life clearly aware of neonates over a wide range of GA, including a large number
the fact that 33 of 118 patients in groups D and E were already of patients after the first week of life. All infants included
included in groups AC. received at least two doses of gentamicin. We found that 6 %
The mean (SD) gentamicin TPC for all treatment episodes of all treatment episodes had a TPC 2 mg/l, a proportion
during the first week of life was 1.1 (0.5)mg/L and after first similar or lower than most comparable studies [11, 14, 20, 27].
week of life 0.8 (0.6)mg/L. Table 2 and Fig. 1 show pharma- Two studies report even lower rates of potential toxic TPCs
cokinetic data on all 509 treatment episodes, divided by the [10, 29]. Hansen and co-workers administered gentamicin
five treatment groups. We observed a potential toxic TPC every 24 h and gave a lower dose to preterm infants <35 weeks
(2.0 mg/L) in 31/509 (6.1 %) treatment episodes. Of these, (3 mg/kg) than to term and near-term infants (4 mg/kg) [9]. In
22 were observed in group A and predominantly in children their study, no patients had a TPC >2.0 mg/L, but >20 % of the
with perinatal asphyxia (n =13) or renal failure of other rea- preterm infants had peak levels <6 mg/L. The Christchurch
sons including congenital renal malformations (n =4). dosing protocol [4, 29] has complex dosing equations based
Thirty-eight of 440 patients (8.6 %) failed the OAE screen- on birth weight, leading to higher dose (milligrams per kilo-
ing before discharge and were referred for follow-up in the gram) and longer intervals (up to 60 h) for infants with the
Author's personal copy
Eur J Pediatr

Table 1 Gentamicin treatment groups, population data and audiology assessment

Group A Group B Group C Group D Group E


PNA 07 days PNA 07 days PNA 07 days PNA >7 days PNA >7 days
GA >36 weeks GA 2936 weeks GA <29 weeks CA 29 weeks CA <29 weeks

Dosing intervals Every 24 h Every 36 h Every 48 h Every 24 h Every 36 h


Treatment episodes (n) 250 61 48 138 12
First treatment in this treatment group 247 60 48 107 11
Patients (n) 247 60 48 83 2
GA (weeks) 39.9 (1.3) 32.6 (2.3) 26.0 (1.5) 33.9 (5.7) 24.1 (1.0)
CA (weeks) 38.0 (5.6) 26.5 (1.2)
BW (g) 3668 (630) 2087 (751) 790 (196) 2403 (1301) 661 (140)
Failed OAE 7 (3 %) 8 (13 %) 11 (23 %) 12 (15 %)
Confirmed hearing impairment 1 (0 %) 2 (3 %) 0 (0 %) 2 (3 %)

Values are presented as mean (SD)


GA gestational age, PNA postnatal age, CA corrected age (PNA + GA), BW birth weight, OAE otoacoustic emission test

lowest body weight [4, 29]. In their evaluation of more than 1, the literature for an association between high peak levels and
000 TPCs, they reported high peak levels and low TPCs, but toxicity in neonates [6, 16, 26]. Based on our previous results
87 % of all patients had only received one dose of gentamicin using the same dose (milligrams per kilogram) for netilmicin
[4, 29]. [12] and other studies using gentamicin 45 mg/kg [10, 11,
Impaired renal function and high plasma creatinine values 20, 27], we would expect that the majority of peak levels with
are well-known risk factors for high aminoglycoside TPC [12, our dosing regimen are >10 mg/L.
14]. Accordingly, we found that most term infants in the first Newborn infants treated with aminoglycosides are at risk
week of life with a TPC >2 mg/L had perinatal asphyxia and of developing hearing impairment. However, there are many
renal impairment. When renal failure is likely, it may be other potential risk factors for hearing impairment including
advisable to either check TPC before the second dose of perinatal asphyxia, CMV infections, intracranial complica-
gentamicin, to routinely increase dosing intervals to 36 h or tions, prematurity and treatment with loop diuretics [7, 13,
to use a different empiric antibiotic until renal function is 26]. A combination of more than one risk factor is often found
clarified. In our unit, we routinely use cefotaxime for empiric in children who later develop hearing impairment. In one
treatment of infants with severe perinatal asphyxia, in partic- recent study, gentamicin did not seem to induce any ototoxic-
ular infants undergoing hypothermia who already are at high ity, and in fact, a protective effect against ototoxicity was
risk for later hearing impairment [12]. proposed [33]. OAE is considered an effective screening test
Our dosing regimen has a higher gentamicin dosage (mil- for detecting aminoglycoside-induced cochlear ototoxicity,
ligrams per kilogram) than what is commonly recommended but positive predictive value is low (67 %) [31]. In our study,
for neonates. Higher peak levels most likely optimise the 38 (8.6 %) infants failed the OAE test. Only 4 out of 38
efficacy of gentamicin treatment. There is little support in patients who failed the OAE tests were later diagnosed having

Table 2 Gentamicin trough plasma concentrations (TPCs) from 509 treatment episodes

Group A Group B Group C Group D Group E


PNA 07 days PNA 07 days PNA 07 days PNA >7 days PNA >7 days
GA >36 weeks GA 2936 weeks GA <29 weeks CA 29 weeks CA <29 weeks

Treatment episodes (n) 250 61 48 138 12


TPC, mean (SD) 1.2 (0.6) 0.9 (0.4) 0.9 (0.5) 0.8 (0.6) 1.1 (0.7)
TPC, median (IQR) 1.1 (0.81.4) 0.9 (0.61.1) 0.9 (0.61.3) 0.6 (0.40.9) 0.9 (0.61.7)
TPC 2.0 mg/L 22 (9 %) 1 (2 %) 1 (2 %) 5 (4 %) 2 (17 %)
TPC 1.5 mg/L 60 (24 %) 5 (8 %) 6 (13 %) 13 (9 %) 3 (25 %)
TPC <0.3 mg/La 5 (2 %) 0 1 (2 %) 10 (7 %) 0

GA gestational age, PNA postnatal age, CA corrected age (PNA + GA), TPC trough plasma concentration
a
TPCs below the lower limit of detection (<0.3 mg/L) were assigned a value of 0.2 mg/L
Author's personal copy
Eur J Pediatr

Fig. 1 Gentamicin trough plasma


concentrations (TPCs) for each
treatment group. Box plots show
median values (solid bar),
interquartile range (margins of
box) and 5 and 95 percentile
(whiskers). Created using IBM-
SPSS (version 20.0)

permanent hearing impairment, and none of these four had Improvements in education of medical staff may reduce such
TPC >2 mg/L. The only child who had a TPC >2 mg/L and errors.
acquired a hearing impairment passed the OAE test but grad- Our study has limitations. First, a lack of data on peak
ually evolved hearing impairment due to a congenital CMV gentamicin levels diminishes our ability to fully assess the
infection. The low rate of hearing impairment among our pharmacokinetic efficacy of our dosage regimen. However,
high-risk intensive care infants, and in particular among pa- we felt it unethical to continue measuring peak levels in this
tients with potential toxic TPCs, is a strong indication that high-dose regimen after having evaluated this dose in our
gentamicin treatment is safe. Long-term follow-up studies previous study. Therapeutic drug monitoring increases the
with detailed hearing evaluation are still needed to confirm patient's pain and may cause clinically important blood loss.
this. Furthermore, 75 % of the cost of gentamicin therapy is due to
Gentamicin is one of the drugs most commonly associated therapeutic drug monitoring [32]. For these reasons, it is
with prescription errors in the paediatric setting [22]. Simpler important that a dosing regimen achieve the targeted peak
dosing protocols are associated with less prescription errors and trough concentrations without requiring additional dosage
[34]. In our study, 93 % of all treatment episodes were cor- adjustment. A second limitation is that the retrospective nature
rectly prescribed. Among the cases were we detected prescrip- of our study makes it difficult to fully assess all levels of
tion errors, almost 2/3 were made in preterm infants after the ototoxicity. Infants with severe hearing impairment are iden-
first week of life, leading to a too large dosing interval and less tified, but we may have missed less severe ototoxicity in the
potential toxicity. It is likely that medical staff only considered neonates who were born towards the end of our study with less
the low GA and failed to recognise and assess the PNA. than 21 months of observation. To overcome these problems,

Table 3 Characteristics of five patients with confirmed hearing impairment in this study cohort

Patient TPC gentamicin (mg/L) GA Asphyxia Otofacial malformation CMV infection Neuro-development Hearing intervention

1 2.5 38 Yes No Yes Delayed CI


2 0.5 38 No No No Delayed HA
3 1.8 40 No Yesa No Delayed HA
4 0.8 31 Maybe No No Normal None
5 0.3 35 No No No Normal None

TPC through plasma concentration, GA gestational age, CMV cytomegalovirus, HA hearing aids, CI cochlear implant
a
Chromosomal abnormality
Author's personal copy
Eur J Pediatr

we are planning a prospective long-term follow-up with a associated with congenital cytomegalovirus infection. Rev Med
Virol 17:355363
complete audiological assessment. Finally, we did not perform
8. Finitzo-Hieber T, McCracken GH Jr, Roeser RJ, Allen DA, Chrane
serial creatinine measurement or analysed urinary biomarkers DF, Morrow J (1979) Ototoxicity in neonates treated with gentamicin
[17] for detailed assessment of potential gentamicin nephro- and kanamycin: results of a four-year controlled follow-up study.
toxicity. Gentamicin nephrotoxicity, however, is challenging Pediatrics 63:443450
9. Glover ML, Shaffer CL, Rubino CM, Cuthrell C, Schoening S, Cole
to assess in the first week of life when plasma creatinine
E, Potter D, Ransom JL, Gal P (2001) A multicenter evaluation of
values are unstable and influenced by renal maturity and gentamicin therapy in the neonatal intensive care unit.
changes in systemic circulation of sick neonates [1]. Further- Pharmacotherapy 21:710
more, it seems that in neonates aminoglycosides rarely induce 10. Hansen A, Forbes P, Arnold A, O'Rourke E (2003) Once-daily
gentamicin dosing for the preterm and term newborn: proposal
clinically relevant renal failure in a normal course of treatment
for a simple regimen that achieves target levels. J Perinatol
when TPC is in a safe range [9, 25]. In contrast, when infants 23:635639
have high TPCs, gentamicin is often discontinued, as these 11. Hoff DS, Wilcox RA, Tollefson LM, Lipnik PG, Commers AR, Liu
infants usually already have an impaired renal function and M (2009) Pharmacokinetic outcomes of a simplified, weight-based,
extended-interval gentamicin dosing protocol in critically ill neo-
one does not want to further exaggerate this with gentamicin.
nates. Pharmacotherapy 29:12971305
12. Klingenberg C, Smabrekke L, Lier T, Flaegstad T (2004) Validation
of a simplified netilmicin dosage regimen in infants. Scand J Infect
Dis 36:474479
Conclusions 13. Lieu JE, Ratnaraj F, Ead B (2013) Evaluating a prediction model for
infant hearing loss. Laryngoscope. doi:10.1002/lary.24033
This simplified gentamicin high-dose (6 mg/kg) regimen was 14. Liu X, Borooah M, Stone J, Chakkarapani E, Thoresen M
associated with a low number of elevated TPCs (>2 mg/L) and (2009) Serum gentamicin concentrations in encephalopathic in-
fants are not affected by therapeutic hypothermia. Pediatrics 124:
a low number of prescription errors, and we found no evi- 310315
dence for ototoxicity. 15. Marsot A, Boulamery A, Bruguerolle B, Simon N (2012) Population
pharmacokinetic analysis during the first 2 years of life: an overview.
Clin Pharmacokinet 51:787798
Acknowledgments JWF receives financial support from the University 16. McCormack JP, Jewesson PJ (1992) A critical reevaluation of the
of Troms as a medical research student. "therapeutic range" of aminoglycosides. Clin Infect Dis 14:320339
17. McWilliam SJ, Antoine DJ, Sabbisetti V, Turner MA, Farragher T,
Conflict of interest None. Bonventre JV, Park BK, Smyth RL, Pirmohamed M (2012)
Mechanism-based urinary biomarkers to identify the potential for
Ethics approval The study was approved by the Regional Ethics aminoglycoside-induced nephrotoxicity in premature neonates: a
Committee of North Norway. proof-of-concept study. PloS ONE 7:e43809
18. Moore RD, Lietman PS, Smith CR (1987) Clinical response to
aminoglycoside therapy: importance of the ratio of peak concentra-
tion to minimal inhibitory concentration. J Infect Dis 155:9399
References 19. Muller-Pebody B, Johnson AP, Heath PT, Gilbert RE,
Henderson KL, Sharland M (2011) Empirical treatment of
neonatal sepsis: are the current guidelines adequate? Arch
1. Allegaert K, Kuppens M, Mekahli D, Levtchenko E, Vanstapel F, Dis Child 96:F4F8
Vanhole C, van den Anker JN (2012) Creatinine reference values in 20. Nestaas E, Bangstad HJ, Sandvik L, Wathne KO (2005) Aminoglycoside
ELBW infants: impact of quantification by Jaffe or enzymatic meth- extended interval dosing in neonates is safe and effective: a meta-
od. J Matern Fetal Neonatal Med 25:16781681 analysis. Arch Dis Child 90:F294F300
2. Allegaert K, Langhendries JP, van den Anker JN (2013) Educational 21. Nielsen EI, Sandstrm M, Honor PH, Ewald U, Friberg LE (2009)
paper: do we need neonatal clinical pharmacologists? Eur J Pediatr Developmental pharmacokinetics of gentamicin in preterm and term
172:429435 neonates. Clin Pharmacokinet 48:253263
3. Begg EJ, Barclay ML (1995) Aminoglycosides50 years on. Br J 22. O'Meara M, Lyons E (2013) An audit of prescribing errors in neo-
Clin Pharmacol 39:597603 nates and paediatrics. Arch Dis Child 98:e1
4. Begg EJ, Vella-Brincat JW, Robertshawe B, McMurtrie MJ, 23. Pacifici GM (2009) Clinical pharmacokinetics of aminoglycosides in
Kirkpatrick CM, Darlow B (2009) Eight years' experience of an the neonate: a review. Eur J Clin Pharmacol 65:4194274
extended-interval dosing protocol for gentamicin in neonates. J 24. Paediatric Formulary Committee - BNF for children (65th 2013).
Antimicrob Chemother 63:10431049 London: Brit Med J Pharmaceutical Press, and RCPCH Publications Ltd
5. De Cock RF, Allegaert K, Schreuder MF, Sherwin CM, de Hoog M, 25. Rao SC, Srinivasjois R, Hagan R, Ahmed M (2011) One dose per day
van den Anker JN, Danhof M, Knibbe CA (2012) Maturation of the compared to multiple doses per day of gentamicin for treatment of
glomerular filtration rate in neonates, as reflected by amikacin clear- suspected or proven sepsis in neonates. Cochrane Database Syst Rev
ance. Clin Pharmacokinet 51:105117 1, CD005091
6. de Hoog M, van Zanten BA, Hop WC, Overbosch E, Weisglas- 26. Robertson CM, Tyebkhan JM, Peliowski A, Etches PC, Cheung PY
Kuperus N, van den Anker JN (2003) Newborn hearing screening: (2006) Ototoxic drugs and sensorineural hearing loss following se-
tobramycin and vancomycin are not risk factors for hearing loss. J vere neonatal respiratory failure. Acta Paediatr 95:214223
Pediatr 142:4146 27. Serane TV, Zengeya S, Penford G, Cooke J, Khanna G, McGregor-
7. Dollard SC, Grosse SD, Ross DS (2007) New estimates of the Colman E (2009) Once daily dose gentamicin in neonatesis our
prevalence of neurological and sensory sequelae and mortality dosing correct? Acta Paediatr 98:11001105
Author's personal copy
Eur J Pediatr

28. Skopnik H, Wallraf R, Nies B, Troster K, Heimann G (1992) neonates 34 weeks' gestation: cost-effectiveness analyses.
Pharmacokinetics and antibacterial activity of daily gentamicin. Pediatrics 103:594598
Arch Dis Child 67:5761 33. Vella-Brincat JW, Begg EJ, Robertshawe BJ, Lynn AM, Borrie TL,
29. Stickland MD, Kirkpatrick CM, Begg EJ, Duffull SB, Oddie SJ, Darlow BA (2011) Are gentamicin and/or vancomycin associated
Darlow BA (2001) An extended interval dosing method for genta- with ototoxicity in the neonate? A retrospective audit. Neonatology
micin in neonates. J Antimicrob Chemother 48:887893 100:186193
30. Stolk LM, Degraeuwe PL, Nieman FH, de Wolf MC, de Boer A 34. Wong E, Taylor Z, Thompson J, Tuthill D (2009) A simplified
(2002) Population pharmacokinetics and relationship between demo- gentamicin dosing chart is quicker and more accurate for nurse
graphic and clinical variables and pharmacokinetics of gentamicin in verification than the BNFc. Arch Dis Child 94:542545
neonates. Ther Drug Monit 24:527531 35. Young TE, Magnum B (2011) NeoFax. A manual of drugs used in
31. Thompson DC, McPhillips H, Davis RL, Lieu TL, Homer CJ, neonatal care. Acorn Publishing, USA: Raleig, North Carolina, pp
Helfand M (2001) Universal newborn hearing screening: summary 4243
of evidence. JAMA-J Am Med Assoc 286:20002010 36. Zingg W, Pfister R, Posfay-Barbe KM, Huttner B, Touveneau S,
32. Thureen PJ, Reiter PD, Gresores A, Stolpman NM, Kawato K, Pittet D (2011) Secular trends in antibiotic use among neonates:
Hall DM (1999) Once- versus twice-daily gentamicin dosing in 20012008. Pediatr Infect Dis J 30:365370

View publication stats

Potrebbero piacerti anche