Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7

Contents lists available at ScienceDirect

Seminars in Fetal & Neonatal Medicine


journal homepage: www.elsevier.com/locate/siny

The use of phenobarbital and other anti-seizure drugs in newborns


Mohamed El-Dib a, *, Janet S. Soul b
a
Neonatal Neurocritical Care, Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
b
FetaleNeonatal Neurology Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA

a b s t r a c t
Keywords: Neonatal seizures constitute the most frequent presenting neurologic sign encountered in the neonatal
Anticonvulsants intensive care unit. Despite limited efficacy and safety data, phenobarbital continues to be used near-
Neonatal seizures
universally as the first-line anti-seizure drug (ASD) in neonates. The choice of second-line ASDs varies
EEG
Hypoxiceischemic encephalopathy
by provider and institution, and is still not supported by sufficient scientific evidence. In this review, we
discuss the available evidence supporting the efficacy, mechanism of action, potential adverse effects, key
pharmacokinetic characteristics such as interaction with therapeutic hypothermia, logistical issues, and
rationale for use of neonatal ASDs. We describe the widely used neonatal ASDs, namely phenobarbital,
phenytoin, midazolam, and levetiracetam, in addition to potential ASDs, including lidocaine, topiramate,
and bumetanide.
© 2017 Published by Elsevier Ltd.

1. Introduction discussed in more detail below, other controversies persist. The


question of whether continuous electroencephalographic (cEEG)
Neonatal seizures affect 1 to 3.5 per 1000 live-born newborns, monitoring is needed to detect seizures has been settled by
and their most frequent etiologies are hypoxiceischemic enceph- numerous studies showing the high incidence of subclinical sei-
alopathy (HIE), ischemic stroke, and intracranial hemorrhage [1e3]. zures, and the difficulty of determining whether a clinical event is a
Management of neonatal seizures varies widely, due in part to the seizure [5]. A second controversy concerns the treatment of sub-
lack of sufficient efficacy and safety data on available ASDs, in clinical (electrographic-only) seizures. Basic science data show that
addition to evidence suggesting that some ASDs may have delete- electrographic-only seizures contribute to neonatal brain injury
rious effects on brain development. In fact, no ASD has been [6,7]. In human newborns, one small study of neonates randomized
approved by the US Food and Drug Administration for use in to receive ASD for EEG-proven seizures showed lower seizure
neonates. burden and better neurodevelopmental outcome at 18e24 months
In the absence of strong scientific evidence, most providers use than among those treated for only clinical seizures [8]. A third
the same ASDs they have used for decades. Some clinicians employ controversy is the duration of ASD treatment. For many clinicians,
new ASDs in neonates after successful use in older infants and the duration of ASD therapy depends largely on the etiology, but
children. In a recent US retrospective report of 9134 neonates with there is no consensus on ASD treatment duration in newborns with
seizures, the use of phenobarbital over the last decade has slightly acute symptomatic etiologies, whose seizures often subside within
declined but is still used in 96% of neonates with seizures, largely as days of onset [9].
a first-line agent. Whereas the use of phenytoin has significantly
declined, the use of levetiracetam has increased ten-fold [4].
3. Choice of anti-seizure drugs

2. Controversies in the treatment of neonatal seizures Of course, clinicians prefer the most efficacious ASD with the
least adverse effects. Unfortunately, currently available data
Apart from the controversy regarding ASD choice, which is regarding efficacy and safety are inadequate. Studies on neonatal
ASD efficacy need to be examined carefully with specific questions
in mind regarding the methodology and results. What definition of
* Corresponding author. Department of Pediatric Newborn Medicine, Brigham
and Women's Hospital, Harvard Medical School, 75 Francis Street, CWN-418, Bos-
efficacy was used: cessation of seizures or decreased seizure
ton, MA 02115, USA. burden, and, if the latter, what magnitude of seizure reduction?
E-mail address: MEl-Dib@bwh.harvard.edu (M. El-Dib). How long was that effect maintained? Was there a control group in

http://dx.doi.org/10.1016/j.siny.2017.07.008
1744-165X/© 2017 Published by Elsevier Ltd.

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
2 M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7

the study? Was the medication used as first-line therapy or second- nervous system (CNS) depression and potential for respiratory
and third-line therapies? Seizures from acute symptomatic etiol- depression requiring additional support. Moreover, there is evi-
ogies typically have a crescendoedecrescendo pattern [10]. dence from multiple animal studies that phenobarbital, as well as
Therefore, a medication used late in the course of acute seizures other ASDs, induces apoptosis in rodent neurons. This apoptosis
(e.g. as second or third line drug) may be misinterpreted as having involves the cortex, hypothalamus, thalamus, and basal ganglia as
good efficacy, when in fact the seizures are subsiding naturally. well as developing white matter [18e20]. Of note, the average dose
Another caveat in some of the studies reported in literature is the of phenobarbital used in these animal studies was 75 mg/kg, which
use of cEEG monitoring. Without cEEG for detection of seizures at significantly exceeds doses typically used in neonates. In addition,
baseline, as well as determination of drug response, efficacy cannot both phenobarbital and phenytoin have been shown to disrupt the
be determined with certainty as clinical events might not have synaptic maturation of neonatal rat brain and impair behavior [21].
been seizures. Moreover, medications may cause electroclinical Long-term effects in rats exposed to similarly very high loading
uncoupling with persistence of the electrographic component doses of phenobarbital include schizophrenia-like behavioral ab-
without clinical manifestations. To measure efficacy accurately, normalities, and impaired learning, memory, and social interaction
drugs need to be tested early in the course of seizures, compared [22]. It is difficult to determine whether phenobarbital has similar
with a control group, and their effect monitored by cEEG. Finally, deleterious effects on human neonates at typical doses.
drug efficacy ideally should be associated with improved long-term Phenobarbital has been shown to be efficacious, at least
neurodevelopmental outcome and decreased risk of later epilepsy. partially, in controlling seizures. In a randomized, crossover trial
Published data on neonatal ASDs should be interpreted cautiously conducted by Painter et al., when 59 neonates with EEG-confirmed
after considering these factors. neonatal seizures were randomized to receive either phenobarbital
or phenytoin, phenobarbital controlled seizures (80% reduction in
4. Effect of hypothermia severity) in 43% of patients, which increased to 57% when both
phenobarbital and phenytoin were used in combination [15]. In
Therapeutic hypothermia is standard of care in neonatal HIE, the another small trial using EEG diagnosis of seizures, 50% of patients
most frequent cause of neonatal seizures, and needs to be consid- responded (complete cessation or 80% decrease in burden of sei-
ered with regard to its interaction with ASDs. Data indicate that zures) to loading doses of phenobarbital up to 40 mg/kg [16]. In a
hypothermia itself is likely to reduce seizure burden [11,12]. In recent retrospective report, phenobarbital was completely effective
addition, hypothermia may alter ASD pharmacodynamics and in 62.6% and partially effective in 16.5% of neonates to eliminate
pharmacokinetics. Hypothermia mainly affects biotransformation both clinical and electrographic seizures; this report did not include
of drugs via reducing the activity of the hepatic cytochrome P450 newborns with status epilepticus [17]. Animal studies suggested
(CYP450) enzymes, and it can affect enzymatic conjugation. The that use of phenobarbital during therapeutic hypothermia after HIE
effect of hypothermia is exaggerated in cases with hypo- insult could be neuroprotective [50]. However, phenobarbital has
xiceischemic (HI)-induced hepatic and renal dysfunction hindering not been shown to improve neurologic outcome in human studies
drug metabolism and elimination [13,14]. Similar concerns are [51,52]. Additionally, a recent Cochrane review reported insufficient
evident during rewarming, but in the opposite direction. evidence to support use of prophylactic phenobarbital following
Rewarming could be associated with increased metabolism and neonatal encephalopathy [53].
accelerated elimination of drugs leading to significant decrease in Neonates receiving phenobarbital (or any other ASD) are already
drug concentrations [14]. at high risk of developmental delay and behavioral impairment
because of both the primary hypoxiceischemic injury and/or the
5. Widely used anti-seizure drugs effect of repeated seizures on the developing brain. It remains
unknown whether phenobarbital worsens or potentially improves
In this review, we discuss widely used ASDs, namely pheno- long-term developmental outcome, when effective in controlling
barbital, phenytoin, midazolam, and levetiracetam, and other po- neonatal seizures.
tential ASDs for newborns, including lidocaine, topiramate and
bumetanide. For each of these medications, we discuss mechanism 5.2. Phenytoin
of action, rationale of use, potential adverse effects, logistical issues,
and any interaction with hypothermia. A summary of efficacy, Traditionally, phenytoin/fosphenytoin has been the most widely
adverse effects, effect of hypothermia and logistical issues is pre- used second-line ASD for neonatal seizures, with fosphenytoin
sented in Table 1 [15e46]. preferred over phenytoin due to reduced adverse effects such as
local irritation. However, in a recent survey, the use of phenytoin
5.1. Phenobarbital has declined [4]. Adverse effects of phenytoin include arrhythmia,
hypotension, and CNS depression [25]. Large doses of phenytoin of
Phenobarbital has been used as the standard first-line of therapy 50 mg/kg (far exceeding the typical 20 mg/kg loading dose in
for neonatal seizures for decades, and is the drug most frequently humans) was shown to cause similar apoptosis and synaptic
used by providers worldwide [4,47e49]. This is likely attributed to disruption in the developing brain of different animal models as did
available efficacy data, predictable pharmacokinetics with long phenobarbital [20,21].
half-life making it feasible to monitor, and likely most of all, long Hypothermia decreases the activities of both CYP2C9 and
experience in its use. CYP2C19 that metabolize phenytoin. In children with traumatic
Phenobarbital is metabolized by CYP2C19, which may be brain injury, increased phenytoin concentrations extended beyond
depressed by therapeutic hypothermia. Although factors affecting the period of therapeutic hypothermia, warranting careful and
renal or hepatic elimination, that may occur in neonatal encepha- prolonged monitoring of phenytoin concentrations [26]. Although
lopathy, could also affect phenobarbital metabolism, the pharma- the pharmacokinetics of phenytoin during neonatal therapeutic
cokinetics of phenobarbital are not generally altered by hypothermia have not been studied, it is possible that this combi-
hypothermia [23,24], so its dosing may not need adjustment for nation could be associated with prolonged clearance and
hypothermia. bradycardia.
The most frequently adverse effect of phenobarbital is central Available data show that phenytoin is about as effective as

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7 3

Table 1
Comparison of efficacy, adverse effects and logistical issues among widely used anti-seizure drugs in neonates.

Anti-seizure drug Efficacy Adverse effects Effect of hypothermia Logistical issues

Phenobarbital Partial efficacy data. May cause CNS depression and Minimal effect; dose does not Long half-life may be increased
As first-line drug, may control respiratory depression. Animal need to be adjusted [23,24] with hepatic dysfunction
seizures in up to 43e63% of studies report neuronal common with systemic HI
newborns [15e17] apoptosis, altered synaptic insult, therefore dose may need
maturation, long-term to be decreased
behavior, learning, memory and
social interaction [18e22]
Phenytoin Partial efficacy data. Hypotension and CNS Hypothermia might decrease Poor enteral bioavailability (IV
As first-line drug may control depression [25]. clearance and augment use preferred) and non-linear
seizures in up to 45% of Apoptosis and synaptic bradycardic effect [26] pharmacokinetics with variable
newborns [15] disruption in animal models elimination [27]
[20,21]
Levetiracetam Limited retrospective data. Few known adverse effects, Likely not affected by Available in both IV and PO
As second- or third-line drug, including somnolence [29]. hypothermia formulation
may be effective in up to 32% Conflicting animal data but
[28] some showed increased brain
injury and apoptosis [30,31]
Midazolam Very limited clinical data. Possible increased length of No effect with limited data in Administered as continuous IV
Retrospective reports describe stay, mortality, and poor short- neonates [35,36] infusion
0e100% efficacy as second- or term neurological outcome
third-line drug [16,32,33] [34]. Possible apoptosis, similar
to other benzodiazepines [18
e20]
Lidocaine Limited retrospective data. Risk of arrhythmias, Hypothermia decreases Administered as continuous IV
Retrospective reports describe particularly with cardiac clearance so modified dosing is infusion. Should be avoided
efficacy of 20e91% as second- dysfunction, hyperkalemia, and needed [37] with cardiac dysfunction or
or third-line drug [16,33,37,38] phenytoin [39,40] phenytoin administration
Topiramate Very limited human data but Anorexia, metabolic acidosis, Increased half-life and IV formulation not
promising animal data. lethargy, irritability, and concentration [45] commercially available
Case series describe efficacy but cognitive dulling reported in
total n ¼ 15 [41e43] older children [44]
Bumetanide Promising experimental data. Potential adverse effects Unknown IV formulation available, and
Limited human data from one include fluid and electrolyte pharmacokinetic data available
small trial [46], awaiting data imbalance, hemodynamic but effective dose is not known
from a larger RCT instability, nephrocalcinosis,
(NCT00830531) azotemia, and ototoxicity

CNS, central nervous system; IV, intravenous; PO, per oral; RCT, randomized controlled trial.

phenobarbital in treating neonatal seizures, as evidenced by the assumption has not been studied. Adverse effects such as mild
only randomized trial comparing these two ASDs, in which sedation, feeding difficulty, mild apnea and bradycardia, and
phenytoin was effective in 45% of cases (compared to 43% with decreased urine output have been reported in neonates receiving
phenobarbital) [15]. The challenges of using phenytoin are related LEV [29,54]. However, no clinically significant adverse effects have
to poor enteral bioavailability and non-linear pharmacokinetics, been documented [28,57e59].
leading to significant variability in elimination and hence, serum In rodent models, LEV showed mixed results regarding brain
concentrations [27]. injury versus a possible neuroprotective role. A loading dose of
80 mg/kg and maintenance daily dose of 40 mg/kg for a week in rat
models of HI was associated with decreased apoptosis suggestive of
5.3. Levetiracetam
a neuroprotective role [60]. This could be due its effect on
increasing expression of superoxide dismutase and glutathione
Levetiracetam (LEV) use in neonatal seizures has increased ten-
peroxidase [30]. In another HI model, LEV 50 m/kg every12 h for 3
fold over the last decade [4]. The reasons for its increased use are
days increased brain injury significantly when used with normo-
probably related to its the wide safety margin and efficacy reported
thermia, but not under hypothermic conditions [31]. Yet another
in older infants and children, availability of pharmacokinetic data,
murine model showed different dose-related effects where a low
and the availability of both parenteral and enteral formulations
dose of 7 mg/kg/day for one week did not have a neuroprotective
[54]. However, RCT data are lacking to demonstrate its efficacy or
effect, whereas a high dose of 70 mg/kg/day for one week was
safety as an ASD in neonates.
associated with apoptosis, whether used with or without hypo-
The mechanism of action of LEV is not fully known. It likely
thermia [61].
works via its effect on a synaptic vesicle protein expressed
Several small retrospective studies have reported efficacy
throughout the brain (synaptic vesicle glycoprotein 2A, SV2A),
(32e82%) of LEV mainly as a second-line treatment both in human
thereby impeding release of neurotransmitters [55]. In newborns
term and preterm neonates; however, the interpretation of these
with HIE, levetiracetam clearance increases significantly from day
studies is limited by methodological issues [28,57e59]. In small
1e7, as half-life decreases from 18.5 to 9.1 h [54]. LEV differs from
retrospective cohorts of 22 term and 12 preterm neonates, a
other ASDs in that 66% of the drug is excreted unchanged in urine
loading dose of levetiracetam was reported to result in complete
and the rest undergoes enzymatic hydrolysis to inactive metabo-
cessation of seizures in 32% and 36% of patients, respectively, with
lites without involving the hepatic cytochrome P450 [56]. This
increasing seizure reduction over time [58,59]. A larger retrospec-
might make the LEV pharmacokinetics less affected by therapeutic
tive cohort of 32 neonates with HIE who received levetiracetam,
hypothermia or hepatic dysfunction than other ASDs, but this

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
4 M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7

mostly as second line of therapy for clinical or electrographic sei- channels, which inhibits neuronal depolarization, and has been
zures, reported that seizures abated within 72 h in 84% of subjects used largely as an intravenous infusion in newborns [64]. Although
[57]. These data should be interpreted cautiously as most seizures widely used in Europe, there is very limited use of lidocaine in the
were diagnosed clinically, drug response was not monitored by USA [65]. The main concern for its use is the risk of cardiac ar-
cEEG, and the vast majority of these infants were first treated with rhythmias, whose incidence was originally reported to be up to
other ASD medications. Thus it is unclear to what degree the 4.8% [39]. A more recent report showed an incidence <2% and as
seizure cessation in these studies is related to LEV efficacy or nat- low as 0.4% when a reduced-dose regimen was adopted. However,
ural resolution of neonatal seizures, particularly with time there is consensus that lidocaine should be avoided in the presence
advancement. In a retrospective study that used cEEG as a standard of cardiac dysfunction or treatment with phenytoin/fosphenytoin
to determine seizure burden in 23 neonates, LEV was associated to avoid the increased risk of arrhythmias [66].
with complete cessation of seizures in only 32% of the patients Lidocaine is largely metabolized by CYP1A2 and its clearance is
within 24 h, while being used mainly as a second- and third-line related to hepatic blood flow [40]. Since reduced hepatic flow
agent [28]. Thus current evidence suggests that LEV works at best during hypothermia is associated with decreased clearance by 24%,
in around 30% of patients with a slow response. An ongoing phase II a specific dosing regimen has been suggested for this clinical sit-
randomized trial testing levetiracetam in neonates (NCT01720667) uation [37].
will hopefully provide more information on LEV efficacy and safety. Lidocaine has been used as a second- and a third-line ASD in
While awaiting these results, LEV may be considered as a second- multiple, small, single-center studies with reported efficacy
or a third-line ASD to treat neonatal seizures, and there is insuffi- ranging from 20% to 91% [16,33,37,38]. Data have been largely
cient evidence to use it as a neuroprotective agent. derived from retrospective studies, with just two small prospective
cohorts [16,37]. When used to treat persistent seizures in spite of
5.4. Midazolam phenobarbital and midazolam treatment in 22 neonates with HIE
receiving therapeutic hypothermia, it was reported to have 91%
Midazolam is one of the benzodiazepines most widely used in efficacy. In that study, efficacy was defined as 80% decrease in the
treating neonatal seizures. Like phenobarbital, its anti-seizure ef- burden of aEEG electrographic seizures within 4 h of administra-
fect is due to its action at GABA receptors [62]. Midazolam is tion without administration of additional rescue ASD [37]. The
metabolized by CYP3A enzyme; hence hepatic dysfunction in ne- limitations of this study are the use of aEEG instead of cEEG to
onates with HIE may lead to greater concentrations of midazolam detect seizures and the late initiation of lidocaine following
[35]. In addition, renal impairment in these patients may affect phenobarbital and midazolam. Other reports have shown signifi-
elimination of its active metabolite. Although neonatal studies have cantly less efficacy than this study. In a large retrospective cohort,
not documented a significant effect of hypothermia on midazolam only 37 out of the 186 (19.9%) who received lidocaine as a second
pharmacokinetics, these studies were limited by small sample size line after phenobarbital had a good response (cessation of aEEG
[35,36]. seizures for >4 h) [33] Lidocaine will continue to have limited
Potential adverse effects of midazolam include sedation and applicability in the USA because of its modest efficacy data and the
need for mechanical ventilation, particularly for midazolam infu- concerns for cardiac arrhythmias.
sion or repeated doses. The routine use of midazolam for sedation
in the neonatal intensive care unit has been discouraged due to 5.6. Topiramate
concerns for increased length of hospital stay, mortality, and poor
short-term neurological outcome [34]. Although other benzodiaz- Although used widely as an ASD in children and adults, top-
epines are associated with apoptotic degeneration in animal iramate has not been used much in neonates, mainly due to the lack
models, midazolam has not been specifically studied [18e20]. of a parenteral formulation and the paucity of human neonatal data
The efficacy of midazolam as a second- or a third-line therapy [67]. An intravenous formulation of topiramate has been tested in
has been reported to vary widely, from 0% to 100%, with data Phase 1 studies in adults [68], so there is a possibility of testing this
limited by small sample size and other methodological limitations. ASD in newborns in the future. Topiramate has multiple mecha-
One retrospective study reported that all 13 neonates with EEG- nisms of action, including modulation of glutamate activity at the
confirmed seizures, refractory to phenobarbital/phenytoin, a-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) and
responded to midazolam bolus and infusion within 2 h [32]. kainate receptors in addition to the modulation of GABA activity at
However, this effect was not reproducible in other studies, and GABA receptors [69,70]. Its neuroprotective role has been demon-
might have been related to its late administration at a time of strated in numerous animal models for HI brain injury, stroke, re-
natural resolution of seizures. In a larger retrospective cohort, fractory seizures, and periventricular leukomalacia, making it a
midazolam had a good response (seizure cessation for >4 h) in only potentially promising ASD for newborns with symptomatic sei-
12.7% of the 161 neonates who received it as a second line of zures caused by acute brain injury [44,45,71,72].
treatment [33]. It should be noted that aEEG used in this latter The pharmacokinetics of topiramate have been described in
study is not reliable for detection of all (especially brief) seizures, infants and in neonatal animal models [73] and one human
which further limits data on midazolam efficacy [63]. In the only neonatal study [74]. Topiramate is metabolized chiefly by CYP3A4,
small prospective randomized controlled trial of midazolam as a which makes it susceptible to interactions with drugs or therapies
second-line treatment of phenobarbital-non-responsive seizures affecting cytochrome P450, including therapeutic hypothermia.
detected by video-EEG monitoring, none of the six babies treated When pharmacokinetics were studied in 13 term neonates
with midazolam had significant decrease in seizure burden receiving therapeutic hypothermia, there was an increased half-life
(defined as <80%) in 48 h [16]. Based on available data, the use of and concentration of topiramate compared with normothermic
midazolam should be reserved for refractory neonatal seizures, neonates. However, the majority of neonates had concentrations
anticipating need for increased respiratory support. within the therapeutic range without apparent adverse effects [74].
Adverse effects reported in older infants and children include
5.5. Lidocaine anorexia, metabolic acidosis, lethargy, irritability, and cognitive
dulling [75]. Although there are no clinical data, topiramate has
Lidocaine works as an ASD by inhibiting voltage-gated sodium potential for less apoptosis compared with other ASDs [76].

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7 5

The only clinical data available are from several case series 6.1. Practice points
which describe promising efficacy and safety [41e43]. These data
showed cessation of refractory seizures in three out of six patients  Phenobarbital continues to be the most widely used first-line
in two reports [42,43] and all three patients in another small report ASD in neonates, despite only partial efficacy and long-term
[41]. Although promising, the extremely limited data available in safety data.
neonates means that its efficacy and safety remain unknown.  Therapeutic hypothermia may alter effects and metabolism of
ASDs, requiring adjustment of ASD dosing.
5.7. Bumetanide  Newer ASDs are often used to treat neonatal seizures, despite
insufficient data regarding efficacy, safety, and even
Bumetanide, a loop diuretic used for decades in newborns, pharmacokinetics.
blocks a sodium/potassium/chloride (NKCC1) co-transporter that is
highly expressed in immature neurons, thereby altering the chlo-
ride gradient and resulting in GABA-mediated inhibition instead of 6.2. Research directions
depolarization [67].
The potential adverse effects of bumetanide include fluid and  Efficacy and safety of new ASDs should be tested in rigorously
electrolyte imbalance (hyponatremia, hypochloremia, hypo- designed clinical trials.
phosphatemia, and hypocalcemia), nephrocalcinosis, azotemia, and  Short- and long-term safety of all ASDs needs further study in
ototoxicity [67]. These adverse effects have been well documented newborn infants.
by studies of bumetanide as a diuretic. Notably, ototoxicity was
reported in a single clinical trial of bumetanide, although no other Funding sources
significant adverse events were reported [56].
The ability of bumetanide to augment the anti-seizure effect of None.
phenobarbital was demonstrated in in-vitro preparations [77] and
in a rat model of hypoxic neonatal seizures [78]. In addition, Conflict of interest statement
bumetanide was found to augment the neuroprotective effect of
phenobarbital in HIE models with hypothermia [79]. In humans, None declared.
although a beneficial effect was demonstrated in one case report
[80], there is not enough evidence from current published data to References
demonstrate safety or efficacy. An open-label Phase I/II trial was
conducted in Europe for neonates with HIE whose seizures did not [1] Lanska MJ, Lanska DJ. Neonatal seizures in the United States: results of the
respond to a loading dose of phenobarbital. This trial aimed to national hospital discharge survey, 1980e1991. Neuroepidemiology 1996;15:
117e25.
evaluate the efficacy and safety of four doses of bumetanide using [2] Glass HC, Pham TN, Danielsen B, Towner D, Glidden D, Wu YW. Antenatal and
bivariate Bayesian sequential dose-escalation design. The study intrapartum risk factors for seizures in term newborns: a population-based
was stopped prematurely after 14 babies were enrolled due to study, California 1998e2002. J Pediatr 2009;154. 24e28.e1.
[3] Glass HC, Shellhaas RA, Wusthoff CJ, Chang T, Abend NS, Chu CJ, et al.
hearing loss in three out of 11 survivors and reported lack of effi- Contemporary profile of seizures in neonates: a prospective cohort study.
cacy [46], although other authors argued that this small trial did J Pediatr 2016;174. 98e103.e1.
achieve the primary efficacy endpoint with an expected rate of [4] Ahmad KA, Desai SJ, Bennett MM, Ahmad SF, Ng YT, Clark RH, et al. Changing
antiepileptic drug use for seizures in US neonatal intensive care units from
ototoxicity [81]. A larger double-blind RCT with dose-escalation 2005 to 2014. J Perinatol 2016.
design to obtain pharmacokinetic and safety data in newborns [5] Murray DM, Boylan GB, Ali I, Ryan CA, Murphy BP, Connolly S. Defining the
with refractory seizures (NCT00830531) has recently completed gap between electrographic seizure burden, clinical expression and staff
recognition of neonatal seizures. Archs Dis Childh Fetal Neonatal Ed 2008;93:
enrollment and results are anticipated soon. This trial aimed to test
F187e91.
bumetanide early in the course of seizures and used continuous [6] Wasterlain CG. Recurrent seizures in the developing brain are harmful. Epi-
EEG monitoring and a standard therapy control group to determine lepsia 1997;38:728e34.
[7] Stafstrom CE, Chronopoulos A, Thurber S, Thompson JL, Holmes GL. Age-
both drug response and safety. This design and the larger number of
dependent cognitive and behavioral deficits after kainic acid seizures. Epi-
subjects may provide more robust data regarding the safety and lepsia 1993;34:420e32.
utility of this medication for neonatal seizures. [8] Srinivasakumar P, Zempel J, Trivedi S, Wallendorf M, Rao R, Smith B, et al.
Treating EEG seizures in hypoxic ischemic encephalopathy: a randomized
controlled trial. Pediatrics 2015;136:e1302e9.
6. Conclusion [9] Fitzgerald MP, Kessler SK, Abend NS. Early discontinuation of antiseizure
medications in neonates with hypoxiceischemic encephalopathy. Epilepsia
Management of neonatal seizures continues to be highly 2017;58:1047e53.
[10] Painter MJ, Scher MS, Stein AD, Armatti S, Wang Z, Gardiner JC, et al.
controversial and variable, largely because of insufficient data to Phenobarbital compared with phenytoin for the treatment of neonatal sei-
guide drug choice in neonates. Data regarding the efficacy and zures. N Engl J Med 1999;341:485e9.
safety of ASDs are largely driven from retrospective reports using [11] Srinivasakumar P, Zempel J, Wallendorf M, Lawrence R, Inder T, Mathur A.
Therapeutic hypothermia in neonatal hypoxic ischemic encephalopathy:
highly variable methodologies and definitions of efficacy. Apoptosis electrographic seizures and magnetic resonance imaging evidence of injury.
reported in animal models was induced by very high doses of ASD J Pediatr 2013;163:465e70.
and has not been demonstrated in humans. ASD safety in humans [12] Low E, Boylan GB, Mathieson SR, Murray DM, Korotchikova I, Stevenson NJ,
et al. Cooling and seizure burden in term neonates: an observational study.
needs to be evaluated both in the neonatal period as well as in long- Archs Dis Childh Fetal Neonatal Ed 2012;97:F267e72.
term neurodevelopmental studies. In the absence of data from [13] Tortorici MA, Kochanek PM, Poloyac SM. Effects of hypothermia on drug
well-designed clinical trials and prospective studies, ASD choice disposition, metabolism, and response: a focus of hypothermia-mediated al-
terations on the cytochrome P450 enzyme system. Crit Care Med 2007;35:
and dosing will continue to be guided by clinical experience. Given
2196e204.
these limitations, ASDs should be used carefully in conjunction [14] Zhou J, Poloyac SM. The effect of therapeutic hypothermia on drug meta-
with continuous EEG monitoring to guide management, and dis- bolism and response: cellular mechanisms to organ function. Expert Opin
continued when the risk of recurrence of seizures declines. This Drug Metab Toxicol 2011;7:803e16.
[15] Bartha AI, Shen J, Katz KH, Mischel RE, Yap KR, Ivacko JA, et al. Neonatal
review highlights the critical need for better evidence from ongoing seizures: multicenter variability in current treatment practices. Pediatr Neurol
and future trials to guide management of neonatal seizures. 2007;37:85e90.

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
6 M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7

[16] Glass HC, Kan J, Bonifacio SL, Ferriero DM. Neonatal seizures: treatment [46] Strasser K, Lueckemann L, Kluever V, Thavaneetharajah S, Hoeber D, Bendix I,
practices among term and preterm infants. Pediatr Neurol 2012;46:111e5. et al. Dose-dependent effects of levetiracetam after hypoxia and hypothermia
[17] Hellstrom-Westas L, Boylan G, Agren J. Systematic review of neonatal seizure in the neonatal mouse brain. Brain Res 2016;1646:116e24.
management strategies provides guidance on anti-epileptic treatment. Acta [47] Holmes GL, Riviello Jr JJ. Midazolam and pentobarbital for refractory status
Paediatr 2015;104:123e9. epilepticus. Pediatr Neurol 1999;20:259e64.
[18] Van Den Broek MPH, Groenendaal F, Toet MC, Van Straaten HLM, Van [48] Welzing L, Junghaenel S, Weiss V, Roth B, Mueller C, Wiesen MHJ. Disposition
Hasselt JGC, Huitema ADR, et al. Pharmacokinetics and clinical efficacy of of midazolam in asphyxiated neonates receiving therapeutic hypothermia e a
phenobarbital in asphyxiated newborns treated with hypothermia: a ther- pilot study. Klin Padiatr 2013;225:398e404.
mopharmacological approach. Clin Pharmacokinet 2012;51:671e9. [49] Van Den Broek MPH, Van Straaten HLM, Huitema ADR, Egberts T, Toet MC, De
[19] Shellhaas RA, Ng CM, Dillon CH, Barks JDE, Bhatt-Mehta V. Population phar- Vries LS, et al. Anticonvulsant effectiveness and hemodynamic safety of
macokinetics of phenobarbital in infants with neonatal encephalopathy midazolam in full-term infants treated with hypothermia. Neonatology
treated with therapeutic hypothermia. Pediatr Crit Care Med 2013;14: 2015;107:150e6.
194e202. [50] Ng E, Taddio A, Ohlsson A. Intravenous midazolam infusion for sedation of
[20] Bittigau P, Sifringer M, Genz K, Reith E, Pospischil D, Govindarajalu S, et al. infants in the neonatal intensive care unit. Cochrane Database Syst Rev
Antiepileptic drugs and apoptotic neurodegeneration in the developing brain. 2017;1. Cd002052.
Proc Natl Acad Sci U. S. A 2002;99:15089e94. [51] Castro Conde JR, Hernandez Borges AA, Domenech Martinez E, Gonzalez
[21] Forcelli PA, Kim J, Kondratyev A, Gale K. Pattern of antiepileptic drug-induced Campo C, Perera Soler R. Midazolam in neonatal seizures with no response to
cell death in limbic regions of the neonatal rat brain. Epilepsia 2011;52: phenobarbital. Neurology 2005;64:876e9.
e207e11. [52] Weeke LC, Toet MC, Van Rooij LGM, Groenendaal F, Boylan GB, Pressler RM,
[22] Kaushal S, Tamer Z, Opoku F, Forcelli PA. Anticonvulsant drug-induced cell et al. Lidocaine response rate in aEEG-confirmed neonatal seizures: retro-
death in the developing white matter of the rodent brain. Epilepsia 2016;57: spective study of 413 full-term and preterm infants. Epilepsia 2016;57:
727e34. 233e42.
[23] Forcelli PA, Janssen MJ, Vicini S, Gale K. Neonatal exposure to antiepileptic [53] El-Dib M, Chang T, Tsuchida TN, Clancy RR. Amplitude-integrated electroen-
drugs disrupts striatal synaptic development. Ann Neurol 2012;72:363e72. cephalography in neonates. Pediatr Neurol 2009;41:315e26.
[24] Frankel S, Medvedeva N, Gutherz S. Comparison of the long-term behavioral [54] Donovan MD, Griffin BT, Kharoshankaya L, Cryan JF, Boylan GB. Pharmaco-
effects of neonatal exposure to retigabine or phenobarbital in rats. Epilepsy therapy for neonatal seizures: current knowledge and future perspectives.
Behav 2016;57(Pt A):34e40. Drugs 2016;76:647e61.
[25] Painter MJ, Scher MS, Stein AD, Armatti S, Wang Z, Gardiner JC, et al. [55] Vento M, De Vries L, Alberola A, Blennow M, Steggerda S, Greisen G, et al.
Phenobarbital compared with phenytoin for the treatment of neonatal sei- Approach to seizures in the neonatal period: a European perspective. Acta
zures. N Engl J Med 1999;341:485e9. Paediatr Int J Paediatr 2010;99:497e501.
[26] Boylan GB, Rennie JM, Chorley G, Pressler RM, Fox GF, Farrer K, et al. Second- [56] Van Rooij LGM, Toet MC, Rademaker KMA, Groenendaal F, De Vries LS. Cardiac
line anticonvulsant treatment of neonatal seizures: a video-EEG monitoring arrhythmias in neonates receiving lidocaine as anticonvulsive treatment. Eur J
study. Neurology 2004;62:486e8. Pediatr 2004;163:637e41.
[27] Spagnoli C, Seri S, Pavlidis E, Mazzotta S, Pelosi A, Pisani F. Phenobarbital for [57] Weeke LC, Schalkwijk S, Toet MC, Van Rooij LGM, De Vries LS, Van Den
neonatal seizures: response rate and predictors of refractoriness. Neuro- Broek MPH. Lidocaine-associated cardiac events in newborns with seizures:
pediatrics 2016;47:318e26. incidence, symptoms and contributing factors. Neonatology 2015;108:130e6.
[28] Barks JD, Liu YQ, Shangguan Y, Silverstein FS. Phenobarbital augments hy- [58] Van Den Broek MPH, Huitema ADR, Van Hasselt JGC, Groenendaal F, Toet MC,
pothermic neuroprotection. Pediatr Res 2010;67:532e7. Egberts TCG, et al. Lidocaine (lignocaine) dosing regimen based upon a pop-
[29] Meyn Jr DF, Ness J, Ambalavanan N, Carlo WA. Prophylactic phenobarbital and ulation pharmacokinetic model for preterm and term neonates with seizures.
whole-body cooling for neonatal hypoxiceischemic encephalopathy. J Pediatr Clin Pharmacokinet 2011;50:461e9.
2010;157:334e6. [59] Van Den Broek MPH, Rademaker CMA, Van Straaten HLM, Huitema ADR,
[30] Sarkar S, Barks JD, Bapuraj JR, Bhagat I, Dechert RE, Schumacher RE, et al. Does Toet MC, De Vries LS, et al. Anticonvulsant treatment of asphyxiated new-
phenobarbital improve the effectiveness of therapeutic hypothermia in in- borns under hypothermia with lidocaine: efficacy, safety and dosing. Archs
fants with hypoxiceischemic encephalopathy? J Perinatol 2012;32:15e20. Dis Childh Fetal Neonat Ed 2013;98:F341e5.
[31] Young L, Berg M, Soll R. Prophylactic barbiturate use for the prevention of [60] Hellstrom-Westas L, Westgren U, Rosen I, Svenningsen NW. Lidocaine for
morbidity and mortality following perinatal asphyxia. Cochrane Database Syst treatment of severe seizures in newborn infants. I. Clinical effects and cerebral
Rev 2016;5. CD001240. electrical activity monitoring. Acta Paediatr Scand 1988;77:79e84.
[32] Zeller B, Giebe J. Pharmacologic management of neonatal seizures. Neonatal [61] Soul J. Novel medications for neonatal seizures: bumetanide and topiramate.
Netw 2015;34:239e44. J Pediatr Neurol 2009;7:85e93.
[33] Empey PE, De Mendizabal NV, Bell MJ, Bies RR, Anderson KB, Kochanek PM, [62] Clark AM, Kriel RL, Leppik IE, White JR, Henry TR, Brundage RC, et al. Intra-
et al. Therapeutic hypothermia decreases phenytoin elimination in children venous topiramate: safety and pharmacokinetics following a single dose in
with traumatic brain injury. Crit Care Med 2013;41:2379e87. patients with epilepsy or migraines taking oral topiramate. Epilepsia 2013;54:
[34] Bourgeois BF, Dodson WE. Phenytoin elimination in newborns. Neurology 1106e11.
1983;33:173e8. [63] Kudin AP, Debska-Vielhaber G, Vielhaber S, Elger CE, Kunz WS. The mecha-
[35] Sharpe CM, Capparelli EV, Mower A, Farrell MJ, Soldin SJ, Haas RH. A seven- nism of neuroprotection by topiramate in an animal model of epilepsy. Epi-
day study of the pharmacokinetics of intravenous levetiracetam in neonates: lepsia 2004;45:1478e87.
marked changes in pharmacokinetics occur during the first week of life. [64] White HS, Brown SD, Woodhead JH, Skeen GA, Wolf HH. Topiramate enhances
Pediatr Res 2012;72:43e9. GABA-mediated chloride flux and GABA-evoked chloride currents in murine
[36] Mruk AL, Garlitz KL, Leung NR. Levetiracetam in neonatal seizures: a review. brain neurons and increases seizure threshold. Epilepsy Res 1997;28:167e79.
J Pediatr Pharmacol Ther 2015;20:76e89. [65] Glauser TA, Miles MV, Tang P, Clark P, McGee K, Doose DR. Topiramate
[37] Patsalos PN. Clinical pharmacokinetics of levetiracetam. Clin Pharmacokinet pharmacokinetics in infants. Epilepsia 1999;40:788e91.
2004;43:707e24. [66] Filippi L, la Marca G, Fiorini P, Poggi C, Cavallaro G, Malvagia S, et al. Top-
[38] Merhar SL, Schibler KR, Sherwin CM, Meinzen-Derr J, Shi J, Balmakund T, et al. iramate concentrations in neonates treated with prolonged whole body hy-
Pharmacokinetics of levetiracetam in neonates with seizures. J Pediatr pothermia for hypoxic ischemic encephalopathyEpilepsia 2009;50:2355e61.
2011;159. 152e154.e3. [67] Korinthenberg R, Schreiner A. Topiramate in children with West syndrome: A
[39] Venkatesan C, Young S, Schapiro M, Thomas C. Levetiracetam for the treat- retrospective multicenter evaluation of 100 patients. J Child Neurol 2007;22:
ment of seizures in neonatal hypoxic ischemic encephalopathy. J Child Neurol 302e6.
2017;32:210e4. [68] Glier C, Dzietko M, Bittigau P, Jarosz B, Korobowicz E, Ikonomidou C. Thera-
[40] Khan O, Chang E, Cipriani C, Wright C, Crisp E, Kirmani B. Use of intravenous peutic doses of topiramate are not toxic to the developing rat brain. Exp
levetiracetam for management of acute seizures in neonates. Pediatr Neurol Neurol 2004;187:403e9.
2011;44:265e9. [69] Liu Y, Barks JD, Xu G, Silverstein FS. Topiramate extends the therapeutic
[41] Abend NS, Gutierrez-Colina AM, Monk HM, Dlugos DJ, Clancy RR. Levetir- window for hypothermia-mediated neuroprotection after stroke in neonatal
acetam for treatment of neonatal seizures. J Child Neurol 2011;26:465e70. rats. Stroke 2004;35:1460e5.
[42] Khan O, Cipriani C, Wright C, Crisp E, Kirmani B. Role of intravenous levetir- [70] Koh S, Jensen FE. Topiramate blocks perinatal hypoxia-induced seizures in rat
acetam for acute seizure management in preterm neonates. Pediatr Neurol pupsAnn Neurol 2001;50:366e72.
2013;49:340e3. [71] Cha BH, Silveira DC, Liu X, Hu Y, Holmes GL. Effect of topiramate following
[43] Kilicdag H, Daglioglu K, Erdogan S, Guzel A, Sencar L, Polat S, et al. The effect of recurrent and prolonged seizures during early development. Epilepsy Res
levetiracetam on neuronal apoptosis in neonatal rat model of hypoxic 2002;51:217e32.
ischemic brain injury. Early Hum Dev 2013;89:355e60. [72] Follett PL, Deng W, Dai W, Talos DM, Massillon LJ, Rosenberg PA, et al.
[44] Komur M, Okuyaz C, Celik Y, Resitoglu B, Polat A, Balci S, et al. Neuroprotective Glutamate receptor-mediated oligodendrocyte toxicity in periventricular
effect of levetiracetam on hypoxic ischemic brain injury in neonatal rats. leukomalacia: a protective role for topiramate. J Neurosci 2004;24:4412e20.
Childs Nerv Syst 2014;30:1001e9. [73] Riesgo R, Winckler MI, Ohlweiler L, Ranzan J, Becker M, Salvador S, et al.
[45] Griesmaier E, Stock K, Medek K, Stanika RI, Obermair GJ, Posod A, et al. Treatment of refractory neonatal seizures with topiramate. Neuropediatrics
Levetiracetam increases neonatal hypoxiceischemic brain injury under 2012;43:353e6.
normothermic, but not hypothermic conditions. Brain Res 2014;1556:10e8. [74] Kundak AA, Okumus N, Dilli D, Erol S, Zenciroglu A. Topiramate use in the

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008
M. El-Dib, J.S. Soul / Seminars in Fetal & Neonatal Medicine xxx (2017) 1e7 7

neonatal period. Pediatr Neurol 2012;46:410. author reply. enhances phenobarbital efficacy in a rat model of hypoxic neonatal seizures.
[75] Glass HC, Poulin C, Shevell MI. Topiramate for the treatment of neonatal PLoS One 2013;8:e57148.
seizures. Pediatr Neurol 2011;44:439e42. [79] Liu Y, Shangguan Y, Barks JDE, Silverstein FS. Bumetanide augments the
[76] Pressler RM, Boylan GB, Marlow, Blennow M., Chiron C., Cross J.H. N, et al. neuroprotective efficacy of phenobarbital plus hypothermia in a neonatal
Bumetanide for the treatment of seizures in newborn babies with hypoxic hypoxiaeischemia model. Pediatr Res 2012;71:559e65.
ischaemic encephalopathy (NEMO): an open-label, dose finding, and feasi- [80] Kahle KT, Barnett SM, Sassower KC, Staley KJ. Decreased seizure activity in a
bility phase 1/2 trial. Lancet Neurol 2015;14:469e77. human neonate treated with bumetanide, an inhibitor of the Na(þ)-K(þ)-
[77] Dzhala VI, Brumback AC, Staley KJ. Bumetanide enhances phenobarbital effi- 2Cl(e) cotransporter NKCC1. J Child Neurol 2009;24:572e6.
cacy in a neonatal seizure model. Ann Neurol 2008;63:222e35. [81] Thoresen M, Sabir H. Epilepsy: neonatal seizures still lack safe and effective
[78] Cleary RT, Sun H, Huynh T, Manning SM, Li Y, Rotenberg A, et al. Bumetanide treatment. Nat Rev Neurol 2015;11:311e2.

Please cite this article in press as: El-Dib M, Soul JS, The use of phenobarbital and other anti-seizure drugs in newborns, Seminars in Fetal &
Neonatal Medicine (2017), http://dx.doi.org/10.1016/j.siny.2017.07.008

You might also like