Clinical Management of Seizures in Newborns: Diagnosis and Treatment

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Pediatr Drugs (2013) 15:9–18

DOI 10.1007/s40272-012-0005-1

THERAPY IN PRACTICE

Clinical Management of Seizures in Newborns


Diagnosis and Treatment

Linda G. M. van Rooij • Marcel P. H. van den Broek •

Carin M. A. Rademaker • Linda S. de Vries

Published online: 19 January 2013


Ó Springer International Publishing Switzerland 2013

Abstract Neonatal seizures can be classified as tonic, seizures as second- or third-line treatment. Although data
clonic, myoclonic, and subtle. A clinical diagnosis is not are scarce, some AEDs with a wide acceptance in adult and
easy as seizures are usually subtle in neonates. In the pediatric neurology practice are being used to treat neo-
majority of newborn infants seizures are subclinical. On the natal seizures (i.e. second-generation AEDs). These drugs
other hand, not all abnormal movements identified by cli- are chemically different from all first-generation AEDs and
nicians as clinical seizures are accompanied by electroen- they have an effect on other pathways so they provide new
cephalographic seizure discharges in the EEG. Precise pharmacological targets for controlling seizures in new-
incidence is difficult to delineate and depends on study borns. Levetiracetam, topiramate, felbamate, bumetanide,
population and criteria used for diagnosis of seizures. lamotrigine and vigabatrin are examples of these second-
Controversy exists as to whether neonatal seizures them- generation AEDs.
selves cause damage to the developing brain, or if the There is an urgent need for prospective, randomized,
damage is primarily due to the underlying cause of the controlled trials to assess the efficacy and safety of these
seizures. As a result of this controversy there is ongoing second-generation AEDs in neonates.
discussion whether all seizures (both clinical and subclin- The aim of this review is to provide an overview of the
ical) should be treated. In addition, when (sub)clinical current knowledge of diagnosis, the effect on brain injury,
seizures are treated, there is no consensus about the most and the treatment of neonatal seizures.
appropriate treatment for neonatal seizures and how to
assess the efficacy of treatment.
Current therapeutic options to treat neonatal seizures 1 Introduction
(i.e. primarily first-generation antiepileptic drugs [AEDs])
are relatively ineffective. In practice, phenobarbital still Seizures occur more often during the neonatal period than
remains the drug of first choice for EEG confirmed or at any other time of life [1]. The most common etiology of
suspected seizures. Benzodiazepines are also used in neonatal seizures is hypoxic-ischemic encephalopathy
(phenobarbital) refractory cases. Several (small) studies (HIE) due to perinatal asphyxia. In the population-based
indicate that lidocaine is an effective drug for refractory study by Ronen et al., 42 % of neonatal seizures were seen
following HIE [2]. Other important causes include intra-
cranial hemorrhage and stroke, infections of the central
L. G. M. van Rooij (&)  L. S. de Vries nervous system, congenital malformations, inborn errors of
Department of Neonatology, KE 04.123.1, Wilhelmina metabolism, transient metabolic disturbances, maternal
Children’s Hospital, University Medical Centre Utrecht,
PO Box 85090, 3508 AB Utrecht, The Netherlands drug abuse or rare neonatal epilepsy syndromes (benign
e-mail: l.g.m.vanrooij@umcutrecht.nl familial neonatal-infantile seizures or fifth-day seizures) [1,
2]. When seizures persist in spite of administration of
M. P. H. van den Broek  C. M. A. Rademaker different antiepileptic drugs (AEDs) one should always
Department of Clinical Pharmacy, Wilhelmina Children’s
Hospital, University Medical Centre Utrecht, Utrecht, consider pyridoxine-dependent epilepsy or pyridoxine
The Netherlands phosphate oxidase deficiency as possible causes, and a
10 L. G. M. van Rooij et al.

therapeutic trial with oral pyridoxal 50 -phosphate in addi- other hand, Mizrahi and Kellaway described in their study,
tion to pyridoxine needs to be undertaken [3, 4]. over 20 years ago, that around two-thirds of clinical seizure
Precise incidence is difficult to delineate and depends on events were not accompanied by seizure patterns in the
the study population and criteria used for diagnosis of EEG [9]. Other studies confirmed this poor correlation
seizures. In studies from the US, an incidence of 0.15–3.5 between the clinical and electrographic manifestations of
per 1000 live births has been reported [2, 5, 6]. Clinical neonatal seizures. Another problem that is common in the
recognition of seizures in newborns is not always newborn with neonatal seizure is ‘electroclinical dissocia-
straightforward because of a highly variable clinical tion’ or ‘uncoupling’, which means that clinical seizures
expression [1]. Since the use of prolonged (video-) EEG decrease or disappear while electroencephalographic sei-
registration it is known that a substantial part of electro- zure patterns continue or even increase [7, 8, 10, 16]. Only
graphic neonatal seizure patterns are not accompanied by the EEG can confirm the epileptic nature of neurologically
clinical signs, especially following treatment with AEDs suspected movements and is considered essential for the
[7–10]. evaluation and confirmation of neonatal seizures. It is
Controversy exists as to whether neonatal seizures known that electrographic neonatal seizure patterns are
themselves cause damage to the developing brain, or if the strikingly different from seizure patterns in older infants
damage is primarily due to the underlying cause of the and adults. Typically, they start focally and evolve in
seizures. amplitude, duration, and waveform morphology. The focal
As a result of this controversy there is ongoing discus- onset of neonatal seizures is regardless of whether the brain
sion whether all seizures (both clinical and subclinical) injury is diffuse or more localized. They commonly extend
should be treated. When (sub)clinical seizures are treated, from their place of origin, recruiting nearby areas and even
there is no consensus about the most appropriate treatment remote locations of the contralateral hemisphere [17–19].
for neonatal seizures and how to assess the efficacy of Patrizi et al. reported that in full-term infants focal onset of
treatment. seizures is more common and that in preterm infants sei-
In this review, an overview is given of the current zures start with a more regional onset [21]. They also
knowledge of the diagnosis, the effect on brain injury, and showed that there is no clear relationship between etiology
the treatment of neonatal seizures with the most frequently and location of onset or morphology of the discharges [21].
used AEDs (phenobarbital, midazolam, lorazepam, clona- A recent study by Shellhaas and Clancy reported that 78 %
zepam, phenytoin, and lidocaine) in clinical practice, and of individual seizures appear in the C3–C4 channel [22].
some relatively new upcoming AEDs (levetiracetam, Nowadays, the most commonly used diagnostic criteria for
topiramate, felbamate, bumetanide, lamotrigine, and viga- electrographic seizure patterns in the neonatal EEG are ‘‘a
batrin) [11–13]. sudden, repetitive, evolving and stereotyped ictal pattern
with a clear beginning, middle and ending and a minimum
duration of 5–10 seconds’’. However, shorter durations of
2 Diagnosis only a few seconds of epileptiform patterns have also been
described to be associated with an adverse outcome [23–
Neonatal seizures can be classified as tonic, clonic, myo- 27]. In a recent study, Murray et al. found that with detailed
clonic, and subtle [14]. A clinical diagnosis is not easy as review of continuous video-EEG monitoring, only 34 % of
seizures tend to be subtle in neonates, including blinking, electrographic seizures had evident clinical signs [28].
eye deviation, chewing, or apnea. In some cases clinical They also reported that only 27 % of the seizures which
recognition is not even possible; for example, when the had clinical features were recognized by medical and
infant is given drugs for muscle paralysis while being nursing staff and that there was an overdiagnosis of clinical
ventilated [1]. On the other hand, not all abnormal move- seizures of 73 %. Thus, the requisite to diagnose and
ments identified by clinicians as clinical seizures are evaluate treatment of neonatal seizures will be a continuous
accompanied by electroencephalographic seizure dis- video-EEG registration. Prolonged video-EEG recordings
charges in the EEG. These movements have been described are available, but this is not applicable in the clinical set-
by Mizrahi and Kellaway as ‘motor automatisms’ of ting in most hospitals and, therefore, at present are mainly
uncertain origin [9]. They may reflect ‘brainstem release used for research applications. Prolonged video-EEG reg-
phenomena’ or may be seizure discharges in deep cerebral istration requires regular attendance of technicians and a
structures, which are not transmitted to surface EEG trained clinical neurophysiologist for correct interpretation
electrodes because of the immature synaptogenesis and of the neonatal EEG. Hence there is a need for alternative
cortical projections [9, 15]. In view of this clinical com- methods of continuous brain monitoring for daily care in
plexity it is argued that the diagnosis of neonatal seizures the neonatal intensive care units (NICUs). With the use of
should not be based on clinical observation alone. On the amplitude integrated EEG (aEEG), a simplified and
Clinical Management of Seizures in Newborns 11

practical tool was introduced in many NICUs. The aEEG recent studies suggest an adverse effect of both clinical and
method was first applied in the original Cerebral Function subclinical neonatal seizures on neurodevelopmental out-
Monitor, developed by Maynard et al. [29]. The aEEG come. Neonatal seizures have been reported to predispose
records a single-channel EEG from two parietal electrodes to later problems with regard to cognition, behavior and
(corresponding to P3–P4 according to the international development of postneonatal epilepsy [44, 45]. In a first,
10–20 system for electrode placement). The EEG signal is multicenter, randomized, controlled trial, full-term infants
filtered (suppresses activity below 2 Hz and above 15 Hz), with moderate to severe HIE and subclinical seizures were
rectified, smoothed, and time-compressed. The signal is assigned randomly to either treating clinical seizures and
displayed on a semi-logarithmic scale at slow speed (6 cm/h) subclinical seizure patterns (group A) or to blinding the
at the cot side [30, 31]. Published papers on aEEG in neo- aEEG registration and only treat clinical seizures (group
nates include both clinical and experimental studies [32]. A B). Although the groups were small there was a trend for a
recent meta-analysis reported that aEEG is suitable for very shorter median duration of seizure patterns in group A
early prediction of outcome after perinatal asphyxia [33]. compared with group B. Another important observation in
The presence of seizures and the effect of treatment in infants this study was that a significant correlation was found
at risk can also be monitored by aEEG [34–37]. As a result of between the duration of seizure patterns and the severity of
the widespread use of this method, new digital aEEG brain injury in the blinded group, as well as in the whole
equipment has been developed, which allows us to study group [46].
more channels with simultaneous display of the original In recent years, hypothermia has become a standard
EEG signal. Several automatic seizure detection algorithms treatment for term infants with moderate to severe HIE, to
have also been developed to improve seizure detection using improve outcome. However, there are limited data
aEEG [37–39]. regarding seizure occurrence during therapeutic hypother-
However, there is concern about the accuracy of aEEG mia. Wusthoff et al. reported an incidence of 65 %, in
in seizure detection. A study by Rennie et al. reported poor which 46 % of the infants had subclinical seizures [47].
sensitivity and specificity for diagnosis of seizures with They also found that the range of time to seizure onset was
aEEG [40]. They used inexperienced observers and the wide. A recent study by Glass et al. reported an incidence
aEEG traces were recorded at different speeds and there of 30 %, and several AEDs were required to control the
was no access to the raw EEG. Others focused on patient seizures [48].
identification, rather than individual seizure pattern rec-
ognition, and showed that about 80 % of patients with
seizures were correctly detected with aEEG [35, 41]. As a 3 Treatment
result of the technique used in aEEG, it is clear that focal,
brief, or low amplitude seizures are difficult to detect [22, As a consequence of the growing body of evidence that
35, 41]. Shellhaas et al. recently reported that aEEG alone neonatal seizures per se contribute to adverse neurodevel-
has significant limitations in the detection and quantifica- opmental outcome, clinicians are more focused on treat-
tion of neonatal seizures [42]. They found a mean seizure ment of neonatal seizures. The question, however, still
detection rate of 40.3 % among 125 recordings with sei- remains how aggressively neonatal seizures should be
zures. A limitation of their study was the very short dura- treated. Unfortunately, data from randomized controlled
tion of the aEEG recording, and the observers once again trials to support the choice of anticonvulsants are limited,
had no access to the raw EEG, which is nowadays obsolete and no definite recommendations based on available data
and not realistic as everybody does have access to digital can be made [12]. However, European NICUs have a
equipment. In a recent study, Shah et al. showed that the similar therapeutic approach for the first-, second- and
2-channel aEEG in combination with the raw EEG signal third-line anticonvulsants [13]. First-generation, older
detected 78 % of electrical seizure patterns [37]. Therefore, AEDs are still the drugs of first (and second/third) choice
users of aEEG must be aware that not all seizures will be because of extensive clinical experience, despite their
detected, but aEEG is a sensitive method to identify an limited clinical effectiveness.
important proportion of neonatal electrographic seizure
patterns. Further development of seizure detection algo- 3.1 First-Generation Antiepileptic Drugs (AEDs)
rithms is likely to contribute to increased accuracy of sei-
zure detection [38]. This accuracy of seizure detection is of Traditional mechanisms of AEDs comprise an inhibitory
great importance as more evidence has become available effect on the glutamatergic pathway, enhanced c-amino-
from animal studies that seizures may impair brain devel- butyric acid (GABA) inhibition, or an influence on neu-
opment and lead to long-term deficits in learning, memory, ronal ion homeostasis. However, in the developing brain,
and behavior [43]. Although data in humans are scarce, neuronal apoptosis can be triggered by blockade of
12 L. G. M. van Rooij et al.

N-methyl-D-aspartate (NMDA) receptors or (excessive) neonates (e.g. 140 hours in asphyxiated neonates) [60],
activation of GABAA-receptors. First-generation AEDs, rapid maintenance doses (2.5–5 mg/kg/day) are not needed
such as phenobarbital and benzodiazepines, exert their in these patients after administration of a sufficient loading
effect by stimulation of GABA. Bittigau et al. showed that dose. Dosing under neonatal hypothermia does not require
these AEDs may cause apoptotic neurodegeneration in the dose adjustment for phenobarbital [115].
developing rat brain at plasma concentrations relevant for Therapeutic drug monitoring (TDM; therapeutic win-
seizure control in humans [49–51]. dow 20–40 mg/L) is recommended in case of seizures and
to prevent drug accumulation or ineffectiveness during
3.1.1 Phenobarbital maintenance dosages since the half-life has wide ranges in
newborns. The effectiveness was found to plateau at a
In practice, phenobarbital still remains the drug of first serum concentration of 40 mg/L and a further increase
choice for confirmed or suspected seizures [52–54]. The to [50 mg/L induced sedation and compromised neuro-
choice for phenobarbital can be explained by its good logic assessment [56].
safety profile, although there is evidence that phenobarbital
itself may impair neurodevelopmental outcome and 3.1.2 Benzodiazepines
increase neuronal apoptosis. Glier and colleagues observed
that neuronal apoptosis increased with phenobarbital doses Benzodiazepines (particularly midazolam, clonazepam,
of 40 mg/kg and more in rats [55]. Nevertheless, there are and lorazepam [11, 13]) are also used in (phenobarbital)
limited studies that demonstrate good efficacy of pheno- refractory cases, with an efficacy reported from 0 to 100 %
barbital. Seizure response rates after loading doses of [61, 62], with two studies reporting responses of 67 % and
15–20 mg/kg are reported to range from 33 to 40 %. With 80 %, respectively [63, 64]. Midazolam, one of the most
rapid sequential loading doses, up to a total dose of 40 mg/ lipophilic benzodiazepines, readily crosses the blood-brain
kg, the responsiveness could be improved to 77 % [56–58]. barrier and has a very rapid onset of action. Less lipophilic
Responsiveness under hypothermia might even be higher benzodiazepines, such as lorazepam, have a less rapid
than under normothermia [115]. onset but do have a favourable less-rapid redistribution
It has been suggested that early posthypoxia-ischemia from the central nervous system that reduces the risk of
administration of phenobarbital may augment the neuro- symptoms of withdrawal after dose cessation.
protective efficacy of therapeutic hypothermia in newborns Midazolam is short-acting and has a more rapid onset of
as well as in a neonatal rodent model [59, 115]. action. A disadvantage can be its metabolic pathway because
Painter et al. reported the effect of phenobarbital com- pharmacologically active (glucuronidated) metabolites are
pared with phenytoin. In 43 % of the phenobarbital-treated formed. Drug-drug interactions or renal impairment could
group, seizures were controlled compared with 45 % in the cause undesired accumulation of these metabolites. In con-
phenytoin-treated group. Addition of the other drug, when trast, the metabolites of clonazepam and lorazepam are
seizures were not controlled with the first drug, did not pharmacologically inactive. Benzodiazepine dosing regi-
significantly improve seizure control [16]. mens are displayed in Table 1.
Although phenytoin (or its less alkaline prodrug fos- Although no therapeutic window has been established
phenytoin) is not inferior to phenobarbital, it has some for seizure control or adverse effects, TDM can be applied
clinically relevant disadvantages such as a dose-dependent in clinical practice to monitor undesired drug accumulation
rate of elimination, which makes dose adjustment com- during continuous infusion.
plicated (i.e. non-linear pharmacokinetics), risk of
increased central nervous system toxicity (due to increased
free drug fraction) in case of renal impairment and hypo-
albuminemia, risk of hypotension and cardiac arrhythmias Table 1 Benzodiazepine dosing regimens
if administered too fast, and risk of adverse local reactions Benzodiazepine Loading dose Maintenance dose
at the injection site of the high alkalinity of the parenteral
Midazolam (IV) [65] 0.05 mg/kg in 0.15–0.5 mg/kg/ha
formulation. Moreover, fosphenytoin is not available in 10 min
some countries in Southern Europe [13]. Lorazepam (IV) [11] 0.05–0.1 mg/kg None, loading dose may be
Phenobarbital can be administered as an intravenous in 5 min repeated if necessary
loading dose of 20 mg/kg, followed by a 10–20 mg/kg if Clonazepam (IV) [66] 0.1 mg/kg 0.01 mg/kg, 3–5 dosages
required. The loading dose can be divided in two equal bolus
dosages when the neonate is not ventilated, but this will IV intravenous
postpone the time window until a therapeutic concentration a
Increase dosing with steps of 0.05 mg/kg/h until a dosage of 0.3 mg/kg/
will be reached. Since phenobarbital has a long half-life in h, with a maximum of 0.5 mg/kg/h
Clinical Management of Seizures in Newborns 13

3.1.3 Lidocaine seizures [74]. Efficacy and safety of these second-genera-


tion drugs should be established in the neonatal population.
Although not widely used, several (small) studies indicate Second-generation AEDs are assumed to have no or less
that lidocaine is an effective drug for refractory seizures as pro-apoptotic properties at therapeutic doses. For leveti-
second- or third-line treatment. The response rate varies racetam and topiramate, this was observed in an in vivo rat
from 70 to 92 % during normothermia and hypothermia model [55, 75].
[62–64, 67–70, 116]. The physicochemical properties of
lidocaine enable passage of the blood-brain barrier, and in 3.2.1 Levetiracetam
the central nervous system it acts as a depressant. Adverse
cardiac effects, such as ventricular tachycardia or fibrilla- Despite limited experience and data on its safety and effi-
tion, should be closely monitored but are not frequently cacy in newborns, levetiracetam seems to be an effective
observed [70–72]. A renewed bodyweight-based dosing and well-tolerated adjunctive antiepileptic agent for seizure
regimen allows lower cumulative dosages which will fur- control in neonates and infants [20, 76]. Levetiracetam
ther reduce the risk of the occurrence of adverse cardiac binds to the vesicular protein synaptic vesicle protein 2A
effects (SV2A) via vesicular entry in neurons, and reduces pre-
During therapeutic hypothermia, lidocaine dosing synaptic neurotransmitter release [77].
should be reduced. Lowered body temperature may alter Controversy exists as to whether levetiracetam exerts its
pharmacokinetic and pharmacodynamic profiles with, in effect via (indirect) modulation of GABAAergic or via a
case of lidocaine, increased risk of (cardiac) toxicity due to non-GABAergic pathway [78, 79]. Potentiation of the
decreased lidocaine clearance [73]. To account for effect of the benzodiazepine diazepam has been observed
deceased clearance under hypothermia, a dosing regimen by Mazarati and colleagues [80].
has been developed based on population pharmacokinetics; In older children and adults it has shown to exert broad-
an initial bolus loading dose of 2 mg/kg is given followed spectrum action since various seizure types have shown a
by consecutive continuous infusions of 6 mg/kg/h (for 3.5 response [76]. The rapid onset of action, presence of an
hour), 3 mg/kg/h (for 12 hours), 1.5 mg/kg/h (for 12 hours) intravenous formulation and good tolerability of a possible
for patients with bodyweight 2.0–2.5 kg, or for patients loading dose do make levetiracetam a suitable therapeutic
with bodyweights 2.5–4.5 kg an initial bolus loading dose option for treatment of status epilepticus. Since it is elim-
of 2 mg/kg is given followed by consecutive continuous inated by the kidneys, dosages should be reduced in case of
infusions of 7 mg/kg/h (for 3.5 hours), 3.5 mg/kg/h (for impaired renal function. There are no known clinically
12 hours), 1.75 mg/kg/h (for 12 hours), before stopping relevant drug-drug interactions with levetiracetam.
[116]. With this regimen, no cardiac arrhythmias have been Khan et al. observed that 86 % of neonates treated with
observed in the hypothermic infants. levetiracetam intravenously for seizures with different
TDM is advised in case of (suspected) cardiac toxicity types and aetiologies had complete seizure cessation at 1
or persisting seizures. Based on observations and extensive hour [20]. In this study, 20 out of 22 neonates (91 %)
clinical expertise, the suspected threshold for cardio- and received a 50 mg/kg loading dose. The other two received
neurotoxic adverse effects in neonates is [9 mg/L. a dose of 10 and 20 mg/kg, respectively. Overall, 86 % of
Lidocaine should not be given to patients with con- all patients received levetiracetam because of seizures that
genital heart disease or to patients who have already were refractory to one or more first-generation AEDs
received phenytoin. (phenobarbital, fosphenytoin, lorazepam, and midazolam).
Although one patient exhibited increased irritability, no
3.2 Second-Generation AEDs immediate adverse effects were evident during or after
infusions [20].
Unfortunately, (a combination of) first-generation AEDs As well as in clinical observations, (neuronal) safety
are not always effective enough for seizure control in also has been confirmed in an in vivo animal model. In P8
certain neonates. Second-generation AEDs, such as lev- rats, doses of up to 100 mg/kg of levetiracetam were found
etiracetam, topiramate, felbamate, bumetanide, and lamo- not to be more neurotoxic (in terms of inducing neuronal
trigine are chemically different from all first-generation apoptosis) than their controls, as observed by Manthey et al
AEDs and have an effect on other pathways. These drugs [75]. Levetiracetam can be dosed intravenously as a load-
provide promising new pharmacological targets for con- ing dose (20–50 mg/kg) followed by an initial maintenance
trolling seizures in newborns. Although data are scarce, dose of 5 mg/kg, which may be increased if required. With
these drugs, with a wide acceptance in adult and pediatric a median half-life of about 9 hours in term neonates, a
neurology practice, are already being used to treat neonatal twice-daily dosing schedule seems to be advisable [81].
14 L. G. M. van Rooij et al.

TDM could become a future option. In adults, a thera- timely fashion in infants with HIE [92]. A limitation of this
peutic range of 5–25 mg/L (or even up to 46 mg/L) is used, finding is that it is based on a study with only four indi-
although there is some evidence that the clinical effect may viduals. In addition, in animal models no linear relationship
not be related to the concentration in plasma. TDM could between oral drug dosages and drug peak concentrations
be advised in case of ineffectiveness, impaired renal have been observed, which may be due to incomplete
function or (suspected) toxicity. absorption of the drug at the relatively high dosages [93,
94]. This disproportional absorption phenomenon has not
3.2.2 Topiramate been described in humans.
Potential dose-independent fatal adverse effects (aplas-
Topiramate is a second-generation AED that enhances tic anemia and acute liver failure) can be caused by fel-
GABA activity, inhibits kainate-mediated conductance at bamate and limit its use. These risks for toxicity should be
glutamate receptors, and modifies Na?- and Ca2?-depen- evaluated before starting treatment [95]. Moreover, ade-
dent action potentials [82]. It appears to be an AED with quate dosages for neonatal seizure control have not been
neuroprotective properties in animal models, and was published.
found to improve cognitive function [83, 84]. Peak blood levels of 70–120 mg/L are associated with
In a small, retrospective, cohort study in neonates, neuroprotection against selective hypoxic-ischemic neuro-
administration of topiramate resulted in ‘apparent reduc- nal necrosis. Based on the relative lack of clinical experi-
tion or no further seizures’ in four out of six neonates with ence with felbamate and on possible disproportional
seizures refractory to phenobarbital despite plasma con- absorption, felbamate administration to neonates should be
centrations of 34–42 mg/L. However, a randomized, con- avoided or at least restricted. If felbamate is administered,
trolled trial suggested oral topiramate (in doses up to TDM-guided dosing is recommended. In addition, blood
25 mg/kg/day) may not be effective as add-on AED for chemistry should be closely monitored.
infants between 1 month and 2 years of age [85, 86].
In a questionnaire, adverse effects were recognized 3.2.4 Bumetanide
more frequently in topiramate-treated infants than in lev-
etiracetam-treated neonates. The main reported adverse In cases of persistent refractory seizures, the loop-
effects included metabolic acidosis and irritability or diuretic bumetanide may occasionally be used in clinical
feeding problems [74]. practice in the near future. It inhibits the Na(?)-K(?)-
Currently, there is limited data about dosing and efficacy 2Cl(-) co-transporters NKCC1 and KCC2 but has a
of topiramate in neonates. There is some published expe- 500-fold greater affinity for NKCC1 than for KCC2 [96].
rience with topiramate dosing during therapeutic hypo- It is registered as a diuretic because of its inhibition of
thermia [87, 88]. Because topiramate dosages for neonatal NKCC1 in the thick ascending loop of Henle in neph-
seizures are not yet established, TDM-guided dosing is rons, but in neonatal rat pups this inhibition of NKCC1
recommended. A therapeutic range for the antiepileptic in cortical neurons resulted in a depression of epilepti-
effect of topiramate, established in adults, is 5–20 mg/L form activity. The drug seems to pass the blood-brain
[89]. A disadvantage of topiramate is that a parenteral barrier and the antiepileptic effect was observed in
formulation is not yet available since the onset of effect dosages that have been tested in human neonates for
depends on the speed of oral drug absorption. diuresis studies [97–99]. Bumetanide dosages for neo-
natal seizure control are currently under investigation in
3.2.3 Felbamate a prospective multicentre study.
NKCC1 inhibition causes an elevation of intracellular
Felbamate, which is chemically unrelated to topiramate, chloride. The effectiveness of NKCC1 inhibition is sup-
inhibits the NMDA receptor and potentiates GABAergic ported by the hypothesis that GABA is an excitatory neu-
action by interacting with the strychnine insensitive glycine rotransmitter in immature cortical neurons [98, 99]. In
recognition site of the NMDA receptor [90]. In an in vivo mature neurons of adults, KCC2 is more expressed than
animal model of incomplete cerebral ischemia and hypoxia NKCC1, but in immature neurons of neonates NKCC1 is
in 7-day-old rat pups, felbamate showed a neuroprotective more expressed than KCC2. Therefore, in immature neo-
effect [91]. natal neurons the baseline intracellular chloride concen-
Since felbamate is only available as oral preparations, tration is higher than in mature adult neurons. Activation of
rapid absorption and distribution is required in order to GABA in immature neurons results in a chloride efflux
obtain a fast and adequate drug level in the brain. Laroia causing synaptic excitation [100].
et al. concluded that due to slow absorption, oral felbamate The strength of GABAergic neurotransmission depends
is unsuitable to reach adequate neuroprotective levels in a on the intraneuronal chloride concentration. NKCC1 is
Clinical Management of Seizures in Newborns 15

responsible for a chloride (Cl-) flux into the cell (influx), 3.2.6 Vigabatrin
and KCC2 is responsible for a flux out of the cell (efflux).
High intracellular chloride concentrations result in mem- Vigabatrin is a selective enzyme-activated irreversible
brane hyperpolarization, and low intracellular chloride inhibitor of GABA transaminase [107]. It is given as a
concentrations result in membrane depolarization, which racemic mixture, of which only the S(?) isomer has
causes excitation [98, 100]. pharmacological activity [108]. Vigabatrin is only avail-
Hypothetically, low intracellular chloride concentra- able as an oral preparation. It is indicated in infants
tions in immature neurons can be the cause of their (1 month to 2 years) as monotherapy for infantile spasms.
relatively low effectiveness in neonatal convulsions since In the literature, vigabatrin has been shown to be effective
barbiturates and benzodiazepines activate GABAergic in resistant neonatal seizures in two patients [109].
neurotransmission, [98]. Bumetanide has been found to In contrast to the other mentioned AEDs, vigabatrin is
augment phenobarbital effectiveness (seizure frequency predominantly eliminated by the kidneys in its unaltered
and duration decreased) in hippocampal rat slices [101]. form. In patients with renal impairment the dose should be
decreased. No direct relationship between plasma concen-
3.2.5 Lamotrigine tration and effect have been established for vigabatrin,
implying that dose adjustments should be based more on
Lamotrigine is an AED that acts at voltage-dependent clinical response than on plasma drug concentration [110].
sodium channels, stabilizing the neuronal membrane and However, TDM-guided dosing can be recommended in
inhibiting the release of glutamate and aspartate [102]. patients with renal impairment.
Lamotrigine is only available as an oral preparation. Peripheral vision loss is an adverse effect that has been
In children, lamotrigine has been demonstrated to be reported as altered visual acuity, color discrimination, or
effective as adjunctive therapy for the generalized seizures contrast sensitivity [111]. In a large, multinational analysis,
associated with Lennox-Gastaut syndrome, adjunctive the prevalence of vigabatrin-induced vision loss was
therapy of partial seizures, and monotherapy of absence approximately 15 % in children [112]. A study performed by
seizures [103]. Evidence of clinical application of lamo- Jammoul et al. indicates that vigabatrin generates a taurine
trigine in neonates and its efficacy in these patients is very deficiency responsible for its retinal phototoxicity [111].
limited. In one published case report, the introduction of Other adverse effects recorded in children (drowsiness,
lamotrigine (4.4 mg/kg as a single daily dose) as add-on constipation, fatigue and apathy) are generally transient,
therapy was followed by rapid and sustained control of being observed during the dose-modification phase and
seizures in a neonate [104]. disappearing either spontaneously or on reduction of the
Lamotrigine is well tolerated in children. Results from vigabatrin dose [113].
placebo-controlled, add-on trials showed that 85 % of To assess this risk of vision loss, the European Medi-
lamotrigine recipients experienced an adverse event com- cines Agency has recommended visual field testing at
pared with 83 % of placebo recipients. Skin rash associated baseline and at 6 months. However, in neonates visual field
with hospitalization and the discontinuation of the study testing is not possible; visual field testing in infants is
drug was reported more frequently by lamotrigine recipi- possible from 3 months of age [114]. Based on the lack of
ents than by placebo recipients, and more frequently by evidence and the risk of vision loss, which cannot be tested
children than by adults. The incidence of rash may be in neonates, vigabatrin should not be used as an option to
reduced with proper initial dosing and dose escalation treat neonatal convulsions.
[105].
Although clear relationships between lamotrigine
plasma concentration and pharmacologic response have not 4 Conclusion
been demonstrated, TDM-guided dosing would be recom-
mendable because of the limited experience in neonates. Neonatal seizures are a common problem in the neonatal
For (adult) patients with refractory seizures, Morris and period. Although progress has been made in the diagnosis
colleagues advise a therapeutic window of 3–14 mg/L and treatment of neonatal seizures, there are still questions
[106]. that remain unanswered. Antiepileptic drugs that are cur-
Due to necessary dose escalation steps, lamotrigine is rently used to treat neonatal seizures are relatively inef-
not indicated for the rapid treatment of neonatal seizures. fective, but the effectiveness appears to improve under
Based on the relative lack of clinical experience, lamotri- hypothermia therapy. Although data are scarce, some
gine administration to neonates should be avoided or at AEDs with a wide acceptance in adult and pediatric neu-
least restricted. If lamotrigine is administered, TDM-gui- rology practice are being used to treat neonatal seizures.
ded dosing is recommended. There is an urgent need for prospective, randomized,
16 L. G. M. van Rooij et al.

controlled trials to determine the efficacy and safety of 21. Patrizi S, Holmes GL, Orzalesi M, et al. Neonatal seizures:
these second-generation AEDs in neonates. characteristics of EEG ictal activity in preterm and fullterm
infants. Brain Dev. 2003;25(6):427–37.
22. Shellhaas RA, Clancy RR. Characterization of neonatal seizures
Acknowledgements Linda van Rooij and Marcel van den Broek by conventional EEG and single-channel EEG. Clin Neuro-
contributed equally to this article. No sources of funding were used to physiol. 2007;118(10):2156–61.
assist in the preparation of this review. The authors have no conflicts 23. Clancy RR, Legido A. The exact ictal and interictal duration of
of interest that are directly relevant to the content of this review. electroencephalographic neonatal seizures. Epilepsia. 1987;
28(5):537–41.
24. McBride MC, Laroia N, Guillet R. Electrographic seizures in
References neonates correlate with poor neurodevelopmental outcome.
Neurology. 2000;55(4):506–13.
1. Volpe JJ. Neurology of the newborn. 5th ed. Philadelphia: 25. Oliveira AJ, Nunes ML, Haertel LM, et al. Duration of rhythmic
Saunders Elsevier; 2008. EEG patterns in neonates: new evidence for clinical and prog-
2. Ronen GM, Penney S, Andrews W. The epidemiology of clin- nostic significance of brief rhythmic discharges. Clin Neuro-
ical neonatal seizures in Newfoundland: a population-based physiol. 2000;111(9):1646–53.
study. J Pediatr. 1999;134(1):71–5. 26. Scher MS, Hamid MY, Steppe DA, et al. Ictal and interictal
3. Schmitt B, Baumgartner M, Mills PB, et al. Seizures and par- electrographic seizure durations in preterm and term neonates.
oxysmal events: symptoms pointing to the diagnosis of pyri- Epilepsia. 1993;34(2):284–8.
doxine-dependent epilepsy and pyridoxine phosphate oxidase 27. Shewmon DA. What is a neonatal seizure? Problems in defini-
deficiency. Dev Med Child Neurol. 2010;52(7):e133–42. tion and quantification for investigative and clinical purposes.
4. Hoffmann GF, Schmitt B, Windfuhr M, et al. Pyridoxal 50 - J Clin Neurophysiol. 1990;7(3):315–68.
phosphate may be curative in early-onset epileptic encephalop- 28. Murray DM, Boylan GB, Ali I, et al. Defining the gap between
athy. J Inherit Metab Dis. 2007;30(1):96–9. electrographic seizure burden, clinical expression, and staff
5. Lanska MJ, Lanska DJ, Baumann RJ, et al. A population-based recognition of neonatal seizures. Arch Dis Child Fetal Neonatal
study of neonatal seizures in Fayette County, Kentucky. Neu- Ed 2008;93(3):F187–91.
rology. 1995;45(4):724–32. 29. Maynard D, Prior PF, Scott DF. Device for continuous moni-
6. Saliba RM, Annegers JF, Waller DK, et al. Incidence of neo- toring of cerebral activity in resuscitated patients. BMJ.
natal seizures in Harris County, Texas, 1992–1994. Am J Epi- 1969;4(682):545–6.
demiol. 1999;150(7):763–9. 30. Hellstrom-Westas L, Rosen I. Continuous brain-function moni-
7. Boylan GB, Rennie JM, Pressler RM, et al. Phenobarbitone, toring: state of the art in clinical practice. Semin Fetal Neonatal
neonatal seizures, and video-EEG. Arch Dis Child Fetal Neo- Med. 2006;11(6):503–11.
natal Ed. 2002;86(3):F165–70. 31. Toet MC, van Rooij LG, de Vries LS. The use of amplitude
8. Clancy RR, Legido A, Lewis D. Occult neonatal seizures. integrated electroencephalography for assessing neonatal neu-
Epilepsia. 1988;29(3):256–61. rologic injury. Clin Perinatol. 2008;35(4):665–78.
9. Mizrahi EM, Kellaway P. Characterization and classification of 32. Hellstrom-Westas L, Rosen I. Amplitude-integrated electroen-
neonatal seizures. Neurology. 1987;37(12):1837–44. cephalogram in newborn infants for clinical and research pur-
10. Scher MS, Alvin J, Gaus L, et al. Uncoupling of EEG-clinical poses. Acta Paediatr. 2002;91(10):1028–30.
neonatal seizures after antiepileptic drug use. Pediatr Neurol. 33. Spitzmiller RE, Phillips T, Meinzen-Derr J, et al. Amplitude-
2003;28(4):277–80. integrated EEG is useful in predicting neurodevelopmental
11. Glass HC, Sullivan JE. Neonatal seizures. Curr Treat Options outcome in full-term infants with hypoxic-ischemic encepha-
Neurol. 2009;11(6):405–13. lopathy: a meta-analysis. J Child Neurol. 2007;22(9):1069–78.
12. Booth D, Evans DJ. Anticonvulsants for neonates with seizures. 34. Hellstrom-Westas L, Rosen I, Swenningsen NW. Silent seizures
Cochrane Database Syst Rev 2004; (4): CD004218. in sick infants in early life: diagnosis by continuous cerebral
13. Vento M, de Vries LS, Alberola A, et al. Approach to seizures in function monitoring. Acta Paediatr Scand. 1985;74(5):741–8.
the neonatal period: a European perspective. Acta Paediatr. 35. Toet MC, van der Meij W, de Vries LS, et al. Comparison
2010;99(4):497–501. between simultaneously recorded amplitude integrated electro-
14. Volpe JJ. Neonatal seizures: current concepts and revised clas- encephalogram (cerebral function monitor) and standard elec-
sification. Pediatrics. 1989;84(3):422–8. troencephalogram in neonates. Pediatrics. 2002;109(5):772–9.
15. Danner R, Shewmon DA, Sherman MP. Seizures in an ate- 36. Shany E, Khvatskin S, Golan A, et al. Amplitude-integrated
lencephalic infant. Is the cortex essential for neonatal seizures? electroencephalography: a tool for monitoring silent seizures in
Arch Neurol. 1985;42(10):1014–6. neonates. Pediatr Neurol. 2006;34(3):194–9.
16. Painter MJ, Scher MS, Stein AD, et al. Phenobarbital compared 37. Shah DK, Mackay MT, Lavery S, et al. Accuracy of bedside
with phenytoin for the treatment of neonatal seizures. N Engl J electroencephalographic monitoring in comparison with simul-
Med. 1999;341(7):485–9. taneous continuous conventional electroencephalography for
17. Clancy RR. Prolonged electroencephalogram monitoring for sei- seizure detection in term infants. Pediatrics. 2008;121(6):
zures and their treatment. Clin Perinatol. 2006;33(3):649–65. vi. 1146–54.
18. Lombroso CT, Holmes G. Value of the EEG in neonatal sei- 38. Navakatikyan MA, Colditz PB, Burke CJ, et al. Seizure detec-
zures. J Epilepsy. 1993;1(6):39–70. tion algorithm for neonates based on wave-sequence analysis.
19. Clancy RR. The contribution of EEG to the understanding of Clin Neurophysiol. 2006;117(6):1190–203.
neonatal seizures. Epilepsia. 1996;37(Suppl. 1):S52–9. 39. Lavery S, Shah DK, Hunt RW, et al. Single versus bihemi-
20. Khan O, Chang E, Cipriani C, et al. Use of intravenous leveti- spheric amplitude-integrated electroencephalography in relation
racetam for management of acute seizures in neonates. Pediatr to cerebral injury and outcome in the term encephalopathic
Neurol. 2011;44(4):265–9. infant. J Paediatr Child Health. 2008;44(5):285–90.
Clinical Management of Seizures in Newborns 17

40. Rennie JM, Chorley G, Boylan GB, et al. Non-expert use of the 63. Shany E, Benzaqen O, Watemberg N. Comparison of continu-
cerebral function monitor for neonatal seizure detection. Arch ous drip of midazolam or lidocaine in the treatment of intrac-
Dis Child Fetal Neonatal Ed. 2004;89(1):F37–40. table neonatal seizures. J Child Neurol. 2007;22(3):255–9.
41. Hellstrom-Westas L. Comparison between tape-recorded and 64. Yamamoto H, Aihara M, Niijima S, et al. Treatments with
amplitude-integrated EEG monitoring in sick newborn infants. midazolam and lidocaine for status epilepticus in neonates.
Acta Paediatr. 1992;81(10):812–9. Brain Dev. 2007;29(9):559–64.
42. Shellhaas RA, Soaita AI, Clancy RR. Sensitivity of amplitude- 65. van Leuven K, Groenendaal F, Toet MC, et al. Midazolam and
integrated electroencephalography for neonatal seizure detec- amplitude-integrated EEG in asphyxiated full-term neonates.
tion. Pediatrics. 2007;120(4):770–7. Acta Paediatr. 2004;93(9):1221–7.
43. Silverstein FS, Jensen FE. Neonatal seizures. Ann Neurol. 66. Andre M, Boutroy MJ, Dubruc C, et al. Clonazepam pharma-
2007;62(2):112–20. cokinetics and therapeutic efficacy in neonatal seizures. Eur J
44. Levene M. The clinical conundrum of neonatal seizures. Arch Clin Pharmacol. 1986;30(5):585–9.
Dis Child Fetal Neonatal Ed. 2002;86(2):F75–7. 67. Hellstrom-Westas L, Westgren U, Rosen I, et al. Lidocaine for
45. Miller SP, Weiss J, Barnwell A, et al. Seizure-associated brain treatment of severe seizures in newborn infants: I. Clinical
injury in term newborns with perinatal asphyxia. Neurology. effects and cerebral electrical activity monitoring. Acta Paediatr
2002;58(4):542–8. Scand. 1988;77(1):79–84.
46. van Rooij LG, Toet MC, van Huffelen AC, et al. Effect of 68. Rey E, Radvanyi-Bouvet MF, Bodiou C, et al. Intravenous
treatment of subclinical neonatal seizures detected with aEEG: lidocaine in the treatment of convulsions in the neonatal per-
randomized, controlled trial. Pediatrics. 2010;125(2):e358–66. iod: monitoring plasma levels. Ther Drug Monit. 1990;12(4):
47. Wusthoff CJ, Dlugos DJ, Gutierrez-Colina A, et al. Electrographic 316–20.
seizures during therapeutic hypothermia for neonatal hypoxic- 69. Radvanyi-Bouvet MF, Torricelli A, Rey E, et al. Effects of
ischemic encephalopathy. J Child Neurol. 2011;26(6):724–8. lidocaine on seizures in the neonatal period: some electroclinical
48. Glass HC, Nash KB, Bonifacio SL, et al. Seizures and magnetic aspects. In: Wasterlain C, Vert P, editors. Neonatal seizures.
resonance imaging-detected brain injury in newborns cooled for New York: Raven Press; 1990. p. 277–85.
hypoxic-ischemic encephalopathy. J Pediatr. 2011;159(5): 70. Malingre MM, van Rooij LG, Rademaker CM, et al. Develop-
731–5. ment of an optimal lidocaine infusion strategy for neonatal
49. Bittigau P, Sifringer M, Genz K, et al. Antiepileptic drugs and seizures. Eur J Pediatr. 2006;165(9):598–604.
apoptotic neurodegeneration in the developing brain. Proc Natl 71. van den Broek MP, Huitema AD, van Hasselt JG, et al. Lido-
Acad Sci USA. 2002;99(23):15089–94. caine (lignocaine) dosing regimen based upon a population
50. Bittigau P, Sifringer M, Ikonomidou C. Antiepileptic drugs and pharmacokinetic model for preterm and term neonates with
apoptosis in the developing brain. Ann N Y Acad Sci. 2003;993: seizures. Clin Pharmacokinet. 2011;50(7):461–9.
103–14. 72. van Rooij LG, Toet MC, Rademaker KM, et al. Cardiac
51. Ikonomidou C, Bittigau P, Koch C, et al. Neurotransmitters and arrhythmias in neonates receiving lidocaine as anticonvulsive
apoptosis in the developing brain. Biochem Pharmacol. treatment. Eur J Pediatr. 2004;163(11):637–41.
2001;62(4):401–5. 73. van den Broek MP, Groenendaal F, Egberts AC, et al. Effects of
52. Clancy RR. The newborn drug development initiative work- hypothermia on pharmacokinetics and pharmacodynamics: a
shop: summary proceedings from the neurology group on neo- systematic review of preclinical and clinical studies. Clin
natal seizures. Clin Ther. 2006;28(9):1342–52. Pharmacokinet. 2010;49(5):277–94.
53. Bartha AI, Shen J, Katz KH, et al. Neonatal seizures: multi- 74. Silverstein FS, Ferriero DM. Off-label use of antiepileptic drugs
center variability in current treatment practices. Pediatr Neurol. for the treatment of neonatal seizures. Pediatr Neurol. 2008;
2007;37(2):85–90. 39(2):77–9.
54. Carmo KB, Barr P. Drug treatment of neonatal seizures by 75. Manthey D, Asimiadou S, Stefovska V, et al. Sulthiame but not
neonatologists and paediatric neurologists. J Paediatr Child levetiracetam exerts neurotoxic effect in the developing rat
Health. 2005;41(7):313–6. brain. Exp Neurol. 2005;193(2):497–503.
55. Glier C, Dzietko M, Bittigau P, et al. Therapeutic doses of to- 76. Krief P, Li K, Maytal J. Efficacy of levetiracetam in children
piramate are not toxic to the developing rat brain. Exp Neurol. with epilepsy younger than 2 years of age. J Child Neurol.
2004;187(2):403–9. 2008;23(5):582–4.
56. Gilman JT, Gal P, Duchowny MS, et al. Rapid sequential phe- 77. Meehan AL, Yang X, McAdams BD, et al. A new mechanism
nobarbital treatment of neonatal seizures. Pediatrics. for antiepileptic drug action: vesicular entry may mediate the
1989;83(5):674–8. effects of levetiracetam. J Neurophysiol. 2011;106(3):1227–39.
57. Van Orman CB, Darwish HZ. Efficacy of phenobarbital in 78. Kuzniecky R, Pan J, Burns A, et al. Levetiracetam has no acute
neonatal seizures. Can J Neurol Sci. 1985;12(2):95–9. effects on brain gamma-aminobutyric acid levels. Epilepsy
58. Painter MJ, Pippenger C, MacDonald H, et al. Phenobarbital and Behav. 2008;12(2):242–4.
diphenylhydantoin levels in neonates with seizures. J Pediatr. 79. Poulain P, Margineanu DG. Levetiracetam opposes the action of
1978;92(2):315–9. GABAA antagonists in hypothalamic neurones. Neuropharma-
59. Barks JD, Liu YQ, Shangguan Y, et al. Phenobarbital augments cology. 2002;42(3):346–52.
hypothermic neuroprotection. Pediatr Res. 2010;67(5):532–7. 80. Mazarati AM, Baldwin R, Klitgaard H, et al. Anticonvulsant
60. Grasela TH Jr, Donn SM. Neonatal population pharmacokinetics effects of levetiracetam and levetiracetam–diazepam combina-
of phenobarbital derived from routine clinical data. Dev Phar- tions in experimental status epilepticus. Epilepsy Res. 2004;
macol Ther. 1985;8(6):374–83. 58(2–3):167–74.
61. Castro C Jr, Hernandez Borges AA, Domenech ME, et al. 81. Merhar SL, Schibler KR, Sherwin CM, et al. Pharmacokinetics
Midazolam in neonatal seizures with no response to phenobar- of levetiracetam in neonates with seizures. J Pediatr.
bital. Neurology. 2005;64(5):876–9. 2011;159(1):152–4.
62. Boylan GB, Rennie JM, Chorley G, et al. Second-line anticon- 82. Angehagen M, Ben-Menachem E, Ronnback L, et al. Novel
vulsant treatment of neonatal seizures: a video-EEG monitoring mechanisms of action of three antiepileptic drugs, vigabatrin,
study. Neurology. 2004;62(3):486–8. tiagabine, and topiramate. Neurochem Res. 2003;28(2):333–40.
18 L. G. M. van Rooij et al.

83. Schubert S, Brandl U, Brodhun M, et al. Neuroprotective effects 100. Pond BB, Berglund K, Kuner T, et al. The chloride transporter
of topiramate after hypoxia-ischemia in newborn piglets. Brain Na(?)-K(?)-Cl- cotransporter isoform-1 contributes to intra-
Res. 2005;1058(1–2):129–36. cellular chloride increases after in vitro ischemia. J Neurosci.
84. Zhao Q, Hu Y, Holmes GL. Effect of topiramate on cognitive 2006;26(5):1396–406.
function and activity level following neonatal seizures. Epilepsy 101. Dzhala VI, Brumback AC, Staley KJ. Bumetanide enhances
Behav. 2005;6(4):529–36. phenobarbital efficacy in a neonatal seizure model. Ann Neurol.
85. Glass HC, Poulin C, Shevell MI. Topiramate for the treatment of 2008;63(2):222–35.
neonatal seizures. Pediatr Neurol. 2011;44(6):439–42. 102. Choi H, Morrell MJ. Review of lamotrigine and its clinical
86. Novotny E, Renfroe B, Yardi N, et al. Randomized trial of applications in epilepsy. Expert Opin Pharmacother. 2003;4(2):
adjunctive topiramate therapy in infants with refractory partial 243–51.
seizures. Neurology. 2010;74(9):714–20. 103. Messenheimer J. Efficacy and safety of lamotrigine in pediatric
87. Filippi L, la Marca G, Fiorini P, et al. Topiramate concentrations patients. J Child Neurol. 2002;17(Suppl. 2):2S34–42.
in neonates treated with prolonged whole body hypothermia for 104. Barr PA, Buettiker VE, Antony JH. Efficacy of lamotrigine in
hypoxic ischemic encephalopathy. Epilepsia. 2009;50(11): refractory neonatal seizures. Pediatr Neurol. 1999;20(2):161–3.
2355–61. 105. Messenheimer JA, Giorgi L, Risner ME. The tolerability of
88. Filippi L, Poggi C, la Marca G, et al. Oral topiramate in neonates lamotrigine in children. Drug Saf. 2000;22(4):303–12.
with hypoxic ischemic encephalopathy treated with hypother- 106. Morris RG, Black AB, Harris AL, et al. Lamotrigine and ther-
mia: a safety study. J Pediatr. 2010;157(3):361–6. apeutic drug monitoring: retrospective survey following the
89. Patsalos PN, Berry DJ, Bourgeois BF, et al. Antiepileptic introduction of a routine service. Br J Clin Pharmacol.
drugs—best practice guidelines for therapeutic drug monitoring: 1998;46(6):547–51.
a position paper by the subcommission on therapeutic drug 107. Metcalf BW. Inhibitors of GABA metabolism. Biochem Phar-
monitoring. ILAE Commission on Therapeutic Strategies. Epi- macol. 1979;28(11):1705–12.
lepsia. 2008;49(7):1239–76. 108. Haegele KD, Schechter PJ. Kinetics of the enantiomers of vig-
90. Rho JM, Donevan SD, Rogawski MA. Mechanism of action of abatrin after an oral dose of the racemate or the active S-enan-
the anticonvulsant felbamate: opposing effects on N-methyl-D- tiomer. Clin Pharmacol Ther. 1986;40(5):581–6.
aspartate and gamma-aminobutyric acidA receptors. Ann Neu- 109. Baxter PS, Gardner-Medwin D, Barwick DD, et al. Vigabatrin
rol. 1994;35(2):229–34. monotherapy in resistant neonatal seizures. Seizure. 1995;4(1):
91. Wasterlain CG, Adams LM, Hattori H, et al. Felbamate reduces 57–9.
hypoxic-ischemic brain damage in vivo. Eur J Pharmacol. 110. Vauzelle-Kervroedan F, Rey E, Pons G, et al. Pharmacokinetics
1992;212(2–3):275–8. of the individual enantiomers of vigabatrin in neonates with
92. Laroia N, Guillet R, McBride M. Felbamate in term infants with uncontrolled seizures. Br J Clin Pharmacol. 1996;42(6):779–81.
hypoxic ischemic encephalopathy. J Pediatr Neurology. 2007;5: 111. Jammoul F, Wang Q, Nabbout R, et al. Taurine deficiency is a
301–4. cause of vigabatrin-induced retinal phototoxicity. Ann Neurol.
93. Adusumalli VE, Yang JT, Wong KK, et al. Felbamate phar- 2009;65(1):98–107.
macokinetics in the rat, rabbit, and dog. Drug Metab Dispos. 112. Wild JM, Chiron C, Ahn H, et al. Visual field loss in patients
1991;19(6):1116–25. with refractory partial epilepsy treated with vigabatrin: final
94. Adusumalli VE, Wichmann JK, Kucharczyk N, et al. Distribu- results from an open-label, observational, multicentre study.
tion of the anticonvulsant felbamate to cerebrospinal fluid and CNS Drugs. 2009;23(11):965–82.
brain tissue of adult and neonatal rats. Drug Metab Dispos. 113. Herranz JL, Arteaga R, Farr IN, et al. Dose-response study of
1993;21(6):1079–85. vigabatrin in children with refractory epilepsy. J Child Neurol
95. Pellock JM. Felbamate in epilepsy therapy: evaluating the risks. 1991; Suppl. 2: S45-51.
Drug Saf. 1999;21(3):225–39. 114. Porro GL, Wittebol-Post D. Impairment of peripheral vision and
96. Payne JA, Rivera C, Voipio J, et al. Cation-chloride co-trans- its measurement. In: Bax M, Dutton G, editors. Visual impair-
porters in neuronal communication, development and trauma. ment in children due to damage in the brain. Cambridge: Mc
Trends Neurosci. 2003;26(4):199–206. Keith Press; 2010. p. 85–97.
97. Fukuda A. Diuretic soothes seizures in newborns. Nat Med. 115. Van den Broek MP, Groenendaal F, Toet MC, et al. Pharma-
2005;11(11):1153–4. cokinetics and clinical efficacy of phenobarbital in asphyxiated
98. Kahle KT, Staley KJ. The bumetanide-sensitive Na-K-2Cl co- newborns treated with hypothermia. Clin Pharmacokinet.
transporter NKCC1 as a potential target of a novel mechanism- 2012;51:671–9.
based treatment strategy for neonatal seizures. Neurosurg Focus. 116. Van den Broek MP, Rademaker CM, van Straaten HL, et al.
2008;25(3):E22. Anticonvulsant treatment of asphyxiated newborns under
99. Dzhala VI, Talos DM, Sdrulla DA, et al. NKCC1 transporter hypothermia with lidocaine: efficacy, safety and dosing. Arch
facilitates seizures in the developing brain. Nat Med. 2005; Dis Child Fetal Neonatal Ed (in press).
11(11):1205–13.

You might also like