Art Crisis Neonatales Manejo

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Neurotherapeutics

https://doi.org/10.1007/s13311-021-01085-8

REVIEW

Novel Therapeutics for Neonatal Seizures


Julie M. Ziobro1 · Krista Eschbach2 · Renée A. Shellhaas1

Accepted: 26 June 2021


© The American Society for Experimental NeuroTherapeutics, Inc. 2021

Abstract
Neonatal seizures are a common neurologic emergency for which therapies have not significantly changed in decades.
Improvements in diagnosis and pathophysiologic understanding of the distinct features of acute symptomatic seizures and
neonatal-onset epilepsies present exceptional opportunities for development of precision therapies with potential to improve
outcomes. Herein, we discuss the pathophysiology of neonatal seizures and review the evidence for currently available treat-
ment. We present emerging therapies in clinical and preclinical development for the treatment of acute symptomatic neonatal
seizures. Lastly, we discuss the role of precision therapies for genetic neonatal-onset epilepsies and address barriers and
goals for developing new therapies for clinical care.

Keywords  Antiseizure medication · Hypoxic-ischemic encephalopathy · KCNQ2 · Phenobarbital · Levetiracetam ·


Bumetanide

Introduction this distinction may not be clear at seizure onset and some
neonates with epilepsy also have super-imposed acute brain
Neonatal seizures are a common emergency with an inci- injury [6]. Acute symptomatic seizures arise within the first
dence up to 0.95 to 2.8 per 1000 live births [1–3]. The week after an acquired brain injury and are usually self-
reported incidence of seizures in preterm infants varies with limited in the neonatal period (though post-neonatal epilepsy
the method of ascertainment, although in population-based can occur in up to ~ 20%) [7]. Neonatal-onset epilepsies are
and unselected cohorts incidence is estimated to be as high often secondary to underlying genetic or metabolic disor-
as 5% [4]. The most common causes of neonatal seizures— ders; methods to assist with early diagnosis and potential
hypoxic–ischemic encephalopathy, stroke, and intracranial for precision therapy will be discussed.
hemorrhage—are similar for both term and preterm infants Key challenges surround the incomplete efficacy of
[5]. Less common etiologies of neonatal seizures include antiseizure medications for neonatal seizures; despite
infection, electrolyte disturbances (e.g., hypoglycemia or standard treatments, more than half of neonates experience
hypocalcemia), as well as metabolic causes. Therefore, most continued seizures after initial loading doses of antiseizure
neonatal seizures are acute symptomatic seizures rather than medications [8, 9]. This is likely, in part, due to immature
a manifestation of neonatal-onset epilepsy [6]. neonatal neurophysiology which leads to excess excitation
The therapeutic approach to neonatal seizures can be and reduced inhibition. Compared to mature adult neurons,
tailored according to the key distinction between acute intracellular chloride concentration is about 20–40  mM
symptomatic seizures or neonatal-onset epilepsy. However, higher in immature neonatal neurons, a difference which
is sufficient to shift the action of gamma-aminobutyric acid
(GABA) from inhibition to excitation [10]. In mature neu-
* Julie M. Ziobro rons, activation of ­GABAA receptors by binding of GABA
ziobroj@med.umich.edu causes an influx of chloride; this results in neuronal mem-
1
brane hyperpolarization and inhibition of action poten-
Department of Pediatrics, Michigan Medicine, C.S. Mott
Children’s Hospital, University of Michigan, 1540 E.
tials. In immature neurons, ­GABAA receptor activation
Hospital Dr, Ann Arbor, MI, USA leads to an efflux of chloride, which effectively increases
2
Department of Pediatrics, Section of Neurology, Denver
the likelihood of firing an action potential. Reversal of
Anschutz School of Medicine, Children’s Hospital Colorado, this chloride gradient is mediated by the expression of the
University of Colorado, Aurora, CO 80045, USA

13
Vol.:(0123456789)
J. M. Ziobro et al.

sodium–potassium-chloride cotransporter 1 (NKCC1), are a handful of randomized, controlled clinical trials in the
which imports chloride, and the potassium-chloride cotrans- treatment of neonatal seizures, but some of these same chal-
porter 2 (KCC2), which exports chloride [11, 12]. In the lenges are still apparent. For example, in the landmark study
immature brain, expression of NKCC1 is higher than KCC2 by Painter et al., comparing phenobarbital to phenytoin,
which leads to the relatively high intracellular chloride con- continuous video-EEG monitoring was not used through-
centration in immature neurons. This results in chloride out the screening period [9]. Expert and key stakeholder
efflux and cell depolarization when GABA binds to the post- recommendations are available to assist in neonatal clinical
synaptic receptor [11] and triggers inward calcium currents trial design and address the above limitations with specific
and the removal of the voltage-dependent magnesium block solutions for each clinical trial phase. These recommenda-
from the N-methyl-D-aspartate (NMDA) receptors. The net tions are an important contribution to improve the quality
effect is promotion of calcium entry and activation of second of clinical trials in neonatal seizures[22]. A brief overview
messengers that increase brain excitability and seizure risk of proposed solutions to the limitations of current neonatal
[13, 14]. Recurrent seizures further increase the intracel- seizure treatment studies are shown in Table 1.
lular chloride concentration, resulting in a more depolar-
ized reversal potential of GABA [15]. Additional develop-
mentally related expression changes in excitatory receptors Treatment of Acute Symptomatic Seizures
that allow the brain to form appropriate activity-dependent
developmental processes also leave the immature brain sus- Phenobarbital
ceptible to hyperexcitability and seizures [13, 14, 16, 17].
Yet, despite these differences in neonatal physiology as Phenobarbital is strongly recommended by the World Health
compared to adults, current treatment practices include first- Organization for the first-line treatment of neonatal seizures
line antiseizure medications that act through the ­GABAA and is the standard of care at most institutions, although
receptor (i.e., phenobarbital and benzodiazepines), which there are limited randomized controlled trials to support this
offers a potential explanation for the high risk of incomplete distinction [23]. Use of phenobarbital for neonatal seizures
treatment response. However, prescription of other antisei- has been very consistent for decades and across geographical
zure medications, including fosphenytoin or levetiracetam, regions. Review of American hospital data from the Pediat-
has not yielded superior results. Below, we discuss in detail ric Health Information System revealed that 97% of neonates
the data for each of the current available medications, as well treated for seizures received phenobarbital and this practice
as emerging treatment options for neonatal seizures (Fig. 1). has been very consistent over time [24–27]. When antisei-
zure medications are prescribed to neonates at hospital dis-
charge, phenobarbital is also the most commonly prescribed
Evaluating Data and Clinical Trial Design medication [27, 28]. These data highlight the importance of
phenobarbital in current treatment paradigms of neonatal
The available data to guide medication selection for the seizures.
treatment of neonatal seizures are limited in quality and Phenobarbital exerts its effect as an allosteric modula-
consist primarily of retrospective studies, with very few tor of the ­GABAA receptor. Yet, activation of the ­GABAA
prospective trials. These study designs are limited by ina- receptor in neonatal neurons results in excitation, instead of
bility to adequately control for confounding variables (e.g., the typical inhibitory effect in adult models [29, 30]. These
disease severity, medication exposure, time to treatment). maturational differences in the newborn are hypothesized to
This is, in part, due to variability in seizure identification— underlie the reduced rates of response to phenobarbital as
some studies assess clinical seizures only and others compared to older children and adults with seizures.
require amplitude-integrated EEG or conventional video- The landmark study by Painter et al. was a randomized
EEG monitoring. Conventional video-EEG monitoring is controlled trial that directly compared phenobarbital to
the gold standard for neonatal seizure identification [18] and phenytoin as first-line treatment for neonatal seizures [9].
is essential for trials of treatment efficacy given high rates Complete seizure resolution occurred in 43% of infants ran-
of subclinical (electrographic only) seizures in neonates. domized to receive phenobarbital, with an 80% reduction of
Assessment of medication response has also varied. Some seizures in 80% of neonates. More recently, a randomized
studies target complete seizure cessation, while others report controlled trial evaluated neonates treated with phenobar-
percent seizure reduction or response within 1 h, etc. This bital versus levetiracetam as first-line therapy after new-
variability reduces the opportunity to compare results across onset conventional EEG-confirmed seizures (NEOLEV2;
studies. Lastly, long-term developmental outcome measures NCT01720667) [31]. This study demonstrated seizure
are commonly not assessed, despite the critical importance freedom at 24 h in 70% of patients treated with a single
of this information to families and clinicians [19–21]. There 20 mg/kg dose of phenobarbital. Notably, this phenobarbital

13
Novel Therapeutics for Neonatal Seizures

Fig. 1  Graphic illustration of sites of action for the currently available TrkB pathway is activated following ischemia, leading to hypofunc-
antiseizure medications used for neonatal seizures (black text, blue tion of the KCC2 channel. Treatment with TrkB pathway modulators
boxes) and therapeutics in preclinical development (red italics text, have been shown to improve KCC2 function in preclinical models
pink boxes). Note that in the neonatal brain, chloride efflux through of neonatal hypoxic–ischemic injury and reverse seizure refractori-
­GABAA receptors (black arrow) predominates due to the chloride ness to phenobarbital, suggesting a future role as adjunctive therapy
gradient created from relative greater expression of NKCC1 versus for refractory acute symptomatic seizures in neonates. Figure created
KCC2. In the adult brain, this chloride gradient is reversed, allow- with BioRender.com
ing for chloride influx (red arrow) through the ­GABAA receptor. The

response rate was higher than in the prior randomized con- symptomatic neonatal seizures [32]. Despite differences in
trolled trial by Painter et al. [9]. There are differences in these two randomized controlled trials, the phenobarbital
the study design between the two trials that could, in part, response rate in the NEOLEV2 study is encouraging. There
explain the higher response rate in the NEOLEV2 study. remain concerns regarding the side effects of sedation, res-
These include variability in EEG monitoring protocols and piratory suppression, and hypotension [31]. Yet, these can
use of therapeutic hypothermia for infants with hypoxic- easily be managed in the intensive care unit environment.
ischemic encephalopathy in NEOLEV2. Additionally, time The potential negative long-term effects of phenobarbital
to treatment was not reported in either study. This variable on the developing brain continue to be a discussion point in
is important due to the natural temporal evolution of acute clinical practice and research. This concern is supported in

13
J. M. Ziobro et al.

Table 1  Study limitations and proposed trial design solutions


Current study limitations Proposed trial design solution

Parents and clinicians wish to determine the best initial treatment Use current common clinical practices (i.e., phenobarbital) as active
approach for neonatal seizures, rather than waiting for the seizures to control arms, and signal clinical equipoise, for future trials
be resistant to multiple medications
It is not ethical to prescribe a placebo to a neonate with active seizures Use active treatment controls based on current clinical practice and
prior randomized controlled trials
Neonates with seizures often have additional co-morbid health con- It is essential to include a control group in the design of future clinical
cerns and it is important to separate these from treatment-related trials
adverse events
Variability in EEG monitoring practices and high rates of electro- Continuous video-EEG monitoring is the gold standard for neonatal
graphic-only seizures in neonates, especially after treatment with seizure identification and needs to be included as part of screening,
antiseizure medication treatment, and follow-up periods for clinical trials of antiseizure
medication efficacy
Inter-reader variability in the identification of neonatal seizures Application of ACNS guidelines for identification of neonatal seizures
increases interrater reliability [176], consideration of central EEG reader
for interpretation or additional standardized site investigator training
Neonatal acute symptomatic seizures have a natural temporal evolution Time to treatment should ideally be standardized, e.g., within 1 h
with an increasing then gradual decreasing pattern [32] that influ- of seizure identification, controlled for in statistical analyses, and
ences evaluation of treatment effect reported in each study; control groups are also necessary
Lack of consistency regarding definition of seizure response and out- Seizure cessation within 1 h of study treatment and lasting for at least
come measures 24 h as standardized primary outcome measure
Small sample sizes limit ability to detect a difference in treatments Multicenter collaborations are essential; well-powered designs are key
Parents and clinicians are interested in improving long-term outcomes, Goal to monitor both treatment and control groups long-term as sec-
including developmental milestones and risk for future epilepsy ondary outcome measures. Consideration of sample size (to permit
relevant conclusions), logistical and financial implications of long-
term follow-up remain challenging
ACNS American Clinical Neurophysiology Society

non-injured animal models in which exposure to phenobar- to seizure onset, did reduce incidence of seizures but was
bital resulted in apoptosis of neurons in the cortex, white not associated with a change in mortality or neurological
matter, and limbic system, as well as disruption of inhibi- outcome at hospital discharge. Long-term outcome meas-
tory synaptic development in hippocampal neurons [33–36]. ures were not assessed. Overall, the data support concerns
These changes may explain the observed anxiety, behavio- for a negative effect of phenobarbital on the immature
ral, and cognitive changes in rodent models after exposure brain and a Cochrane Review concluded that prophylactic
to phenobarbital [37, 38]. Additionally, it is proposed that exposure in infants at risk for seizures is not recommended
damage to ­GABAA receptor neurons results in an enhanced [40]. Importantly, the negative effect of untreated neonatal
excitatory network later in life [33]. seizures is considered more clinically meaningful than the
However, there has also been suggestion of a neuroprotec- potential adverse effects of phenobarbital. Thus, phenobar-
tive effect of phenobarbital. In a mouse model of hypoxia bital remains the first-line standard of care treatment option
induced seizures, treatment with phenobarbital (compared for neonatal seizures. Yet, the above concerns highlight the
with saline placebo) resulted in less hippocampal neuronal urgent need for novel therapeutic options.
death after a second-hit and later life exposure to kainic acid
[37]. In this study, there were similar rates of neuronal cell Fosphenytoin and Phenytoin
death between the control group of uninjured mice with-
out phenobarbital exposure and the phenobarbital exposed Fosphenytoin is used in up to 15% of patients with neonatal
groups (group 1—hypoxia-induced seizure model exposed seizures [25, 26] but is typically prescribed when seizure
to phenobarbital and group 2—uninjured mice with pheno- control is inadequate after initial treatment with phenobarbi-
barbital exposure), although this was not controlled for the tal. Fosphenytoin is a phosphorylated prodrug of phenytoin
initial response to phenobarbital or seizure burden [37]. Fur- and has the advantage of less cardiac toxicity and fewer local
thermore, there was a clinical trial to evaluate for a potential extravasation effects as compared to phenytoin, although
preventative neuroprotective effect of phenobarbital in new- both require careful consideration of drug interactions. The
borns with hypoxic–ischemic encephalopathy [39]. It was reduced risk of local skin reactions with fosphenytoin is due
determined that early treatment with phenobarbital, prior to the lower pH of 8.6 (compared to phenytoin pH of 11)

13
Novel Therapeutics for Neonatal Seizures

and absence of propylene glycol. Additionally, compared than phenobarbital on the newborn brain, and improved effi-
to phenytoin, fosphenytoin is more likely to be compatible cacy compared to phenobarbital in infantile epilepsy [49].
with other co-administered medications or infusions in an Initial single-center, retrospective studies of levetiracetam
intravenous line. reported efficacy in both term and preterm infants with a
Fosphenytoin is converted to phenytoin, the active complete seizure cessation widely ranging between 26 and
metabolite, and then acts to stabilize the neuronal mem- 86% of patients [50–54]. However, these studies were limited
brane by altering sodium currents. There was initial con- by primarily retrospective study designs, lack of a control
cern that parenteral fosphenytoin administration may result group, and variable outcomes measures, as discussed pre-
in reduced serum levels of free phenytoin, as early studies viously. Yet, these numerous preliminary studies provided
of pharmacokinetics were in older children and adults. How- encouraging initial data and rationale for the NEOLEV2
ever, additional studies that included neonates demonstrated study, a randomized controlled phase IIb efficacy, dose-
adequate levels of free phenytoin serum concentration after escalation study of first-line treatment with levetiracetam
loading doses of fosphenytoin and similar times to conver- compared to treatment with phenobarbital. Seizure cessa-
sion to phenytoin as adult patients[41, 42]. Despite this, an tion for 24 h occurred in significantly more patients treated
additional challenge remains with enteral administration of with phenobarbital compared to levetiracetam (80% vs 28%,
phenytoin due to inconsistent (often limited) absorption and p < 0.001) [31]. After dose escalation, with an additional
variability in serum free phenytoin levels [43]. 20 mg/kg of levetiracetam (total dose 60 mg/kg), the seizure
Studies comparing phenytoin or fosphenytoin to phe- freedom rate at 24 h increased to 35%. Despite some efficacy
nobarbital in newborns demonstrate similar efficacy with demonstrated by levetiracetam, this study provides strong
45–56% of patients becoming seizure free after initial treat- evidence of the superiority of phenobarbital as a first-line
ment with phenytoin/fosphenytoin [9, 44]. A single retro- antiseizure medication and lack of seizure control with up to
spective study reported a response rate of phenytoin for neo- 60 mg/kg in loading doses of levetiracetam as first-line treat-
natal acute symptomatic seizures of 85.7% [45], although ment for neonatal seizures. Although not powered to evalu-
the details about seizure burden, and timing of treatment ate levetiracetam as a second-line antiseizure medication for
were not reported. There are a few case reports on the use neonatal seizures, it is notable that none of the neonates with
of fosphenytoin in preterm infants with no adverse effects seizures refractory to two loading doses of phenobarbital
reported [46, 47]. responded to levetiracetam. Thus, the NEOLEV2 study pro-
There is also concern of a neuronal apoptotic effect with vides quite conclusive data that the secular trend of prescrib-
phenytoin that is dose dependent but triggered at typical ing levetiracetam for neonatal seizures is not justified. As a
loading doses [48]. However, a long-term follow-up study result, phenobarbital remains the first-line standard of care.
demonstrated that newborns who received initial treatment Animal studies have not clearly demonstrated neuronal
with fosphenytoin were more likely to have normal or only apoptosis with average doses of levetiracetam, although
mild developmental delay at 18–24 months, compared with there is a possible dose-dependent effect with neuronal
newborns treated with phenobarbital [44]. This study was apoptosis after exposure to high doses (70 mg/kg) of leveti-
limited by the retrospective design and relatively small sam- racetam as compared to low doses (7 mg/kg) in a neonatal
ples size in the fosphenytoin group (n = 23 in fosphenytoin hypoxia mouse model [55]. However, long-term outcomes of
group; n = 80 in phenobarbital group) but may suggest an high-dose levetiracetam exposure were not evaluated. Short-
alternative to the potential negative effects of phenobarbital term follow-up studies have reported improved tone and pos-
on the developing brain. ture among infants treated with levetiracetam compared with
neonates treated with phenobarbital [56], although prelimi-
Levetiracetam nary data suggest long-term neurodevelopmental outcomes
appear similar [57].
Levetiracetam acts through the synaptic vesicle glycopro-
tein 2A (SV2A), which is a protein involved in the release
of neurotransmitters, although the exact mechanism is not
known. Levetiracetam is used less commonly than pheno- Treatment of Status Epilepticus in Neonates
barbital for the initial treatment of neonatal seizures. Over
the past decade, use of levetiracetam increased from < 10% Guidelines from the American Clinical Neurophysiology
of all neonates with seizures to nearly 40%, surpassing the Society define neonatal status epilepticus as seizure duration
use of fosphenytoin/phenytoin [26, 28]. Recent interest in totaling > 50% of a 1-h epoch [58]. The causes of neonatal
levetiracetam is likely multifactorial and includes its avail- status epilepticus are comparable to neonatal seizures in
ability for parenteral administration, relative safety profile, general, with hypoxic–ischemic encephalopathy or intrac-
minimal drug interactions, likelihood of less adverse effects ranial hemorrhage most common. There are high rates of

13
J. M. Ziobro et al.

morbidity and mortality associated with neonatal status approximately 20% of infants had a sustained response and
epilepticus, with death occurring in up to half of preterm did not require additional antiseizure medications after sec-
neonates [59]. ond line treatment with lidocaine infusion[69]. Additionally,
data suggest lidocaine may be more effective in term than
Benzodiazepines preterm infants [69].
Large retrospective studies have identified cardiac
Benzodiazepines are first-line treatment for pediatric sta- arrhythmias in up to 4.8% of neonates during infusion with
tus epilepticus, but there are limited data to guide their use lidocaine [70], although with a reduced dosing regimen
in neonatal status epilepticus. Benzodiazepines exert effect this number decreased to < 1% [71]. This highlights the
through the G ­ ABAA receptors as allosteric agonists, with importance of continuous cardiac monitoring during treat-
the same challenges related to the maturational pattern in ment with lidocaine. Future well-designed studies would be
the newborn brain as phenobarbital. Additionally, similar to beneficial to define the role of lidocaine in the treatment
phenobarbital, animal models demonstrate that midazolam of neonatal seizures. Until such data become available, we
may increase status epilepticus-associated neuronal injury suggest consideration of lidocaine for neonates with status
with no increase in animal survival [36, 60]. epilepticus despite loading doses of ≥ 40 mg/kg of pheno-
Available data regarding the use of benzodiazepines in barbital. Note that exposure to fosphenytoin/phenytoin and
neonates for seizures are primarily limited to retrospective presence of congenital heart disease are contraindications
studies. The response rate is varied with complete seizure due to the risk of arrhythmia. Additionally, the clinical team
response occurring in < 20% of neonates treated with 0.1 mg/ must have a plan for subsequent treatment after the 48 h of
kg midazolam as first-line treatment [61] and between 0 and lidocaine are completed.
100% when midazolam or clonazepam are used as second-
line treatment for ongoing seizures [62–64]. Nearly half of
the neonates treated with midazolam as first-line treatment Duration of Treatment with Antiseizure
had continued seizures [61]; given the limitations in study Medication
design this number may, in fact, be higher.
In clinical studies, hypotension was the most frequently It is important to also consider the duration of treatment
reported adverse event, and occurred in nearly 10% of with either current or emerging therapeutics for neonatal
infants, although transient hypoxia, apnea, decreased urinary seizures in order to reduce any potential risk of treatment on
output, and tachycardia also can occur [61, 64]. Additionally, the immature neonatal brain. Neonatal seizures are predomi-
in very preterm neonates (24–32 weeks gestation) exposure nantly acute symptomatic seizures and the highest seizure
to midazolam was found to be inversely associated with hip- burden is typically in the first 24 h after onset [32]. Seizures
pocampal volume and cognitive outcomes [65]. The incon- usually resolve within several days, but up to 20% go on to
sistent response to benzodiazepines in neonates, as well as develop post-neonatal epilepsy [137, 138]. The incidence of
the potential additive neurotoxic effect on the newborn brain, post-neonatal epilepsy and of abnormal neurodevelopmental
highlights the need for alternative treatment options for neo- outcomes does not appear to be reduced by longer dura-
natal status epilepticus. tion of treatment with antiseizure medications [137, 139].
A recent prospective, multicenter comparative effectiveness
Lidocaine study by the Neonatal Seizure Registry confirmed that dis-
continuation of antiseizure medication after resolution of
Lidocaine is commonly used as a local anesthetic but has acute symptomatic seizures and prior to hospital discharge
been employed for second and third-line treatment of neo- does not alter future epilepsy risk, rates of cerebral palsy, or
natal seizures [26]. It is also an anti-arrhythmia drug and 2-year neurodevelopmental outcome [140].
has a narrow therapeutic window. Additionally, lidocaine The World Health Organization (WHO) recommends
is metabolized in the liver by the cytochrome p450 system, discontinuation of antiseizure medication in neonates with
which can be influenced by gestational age and therapeu- a normal neurological exam and/or normal EEG after at
tic hypothermia; however, this can be mitigated with the least 72 h of seizure freedom [23]. This recommendation
proposed dosing regimens available for preterm and term is intended for neonates with acute symptomatic seizures,
neonates [66–68]. and not for infants with neonatal-onset epilepsy. However,
Retrospective and small clinical studies demonstrate effi- in neonates with acute symptomatic seizures, there remains
cacy of lidocaine in neonates for whom phenobarbital failed significant variability in how long antiseizure medications
to control seizures. The response rate to lidocaine infusions are continued, with medications often maintained after hos-
has been reported to be approximately 50–70% [63, 64, 69]. pital discharge at most centers [27]. A longer duration of
However, in the largest retrospective study (n = 413) only seizures in the neonatal period, abnormal MRI brain, and

13
Novel Therapeutics for Neonatal Seizures

some seizure etiologies are risk factors for longer duration significant reduction in seizure burden with escalating doses
of treatment [27, 138, 141]. It is also known that the hospital of bumetanide (0.1, 0.2, or 0.3 mg/kg) compared to the con-
site influences duration of treatment, as well as availabil- trol group; however, the interpretation was complicated as
ity of a neonatal neurocritical care service [27, 142]. This seizure severity was unexpectedly not randomly distributed
highlights the importance of institutional efforts to stand- across groups. Yet, it appeared that infants with high sei-
ardize the identification and treatment of neonatal seizures zure burden had more than expected reduction in seizures
and reduce unnecessarily prolonged exposure to antiseizure after bumetanide. Importantly, this was a pilot safety trial
medication. Most neonates with acute symptomatic neonatal not intended to assess primary efficacy outcomes. However,
seizures do not require continued medication after hospital this study revealed important initial data regarding tolerated
discharge. Conversely, infants with neonatal-onset epilepsy bumetanide doses and safety data for future trials design.
usually do require long-term treatment.
Topiramate

Emerging Treatment Options Topiramate has multiple mechanisms of action, including


glutamate-receptor inhibition and blockade of Na + chan-
Bumetanide nels, high-voltage Ca + currents, and carbonic anhydrase
isoenzymes. These characteristics have raised the potential
Bumetanide is a loop diuretic that blocks the Na–K-Cl of a neuroprotective effect of topiramate through reduction
co-transporter. Early animal models suggested a potential of excitatory pathways. This has been supported by both
benefit when combined with phenobarbital for treatment of in vitro and in vivo clinical studies, including models in the
neonatal seizures [72], although this was not a completely newborn such as periventricular leukomalacia models [78].
consistent result and it has not been replicated in all rodent There is also report of increased safety in animal models
models of birth asphyxia [73]. Additionally, there is concern as compared to phenobarbital or benzodiazepines, as well
regarding the ability of bumetanide to cross the blood–brain neuronal apoptosis only with doses significantly higher than
barrier and reach intended targets [74]. currently used treatment doses [79, 80]. Two randomized
In an initial open-label, dose escalation, feasibility study clinical trials of topiramate initiation at the start of therapeu-
(NEMO trial; NCT01434225) of bumetanide administered tic hypothermia compared to hypothermia alone revealed a
with a second dose of phenobarbital as second-line therapy decrease in seizure burden and need for less antiseizure med-
after phenobarbital failure (N = 14), the primary seizure ications in the first 24 h, although the latter was not assessed
efficacy endpoint (80% seizure reduction) occurred in less for significance [81]. There was no significant effect on mor-
than half the cohort [75]. However, 5 of the neonates did tality or neurological disability [81, 82]. While the rate of
not have seizures in the baseline period. It has been argued post-neonatal epilepsy was lower among neonates treated
that if these neonates were excluded from efficacy analysis, with topiramate plus hypothermia compared to hypothermia
then 5/9 neonates demonstrated a > 80 seizure reduction and alone, this was not statistically significant (14.3% vs 30.4%,
reached the primary clinical efficacy endpoint of a > 50% P = 0.21) [82]. The first study by Filippi et al. included 21
response rate [76]. Additionally, 3 of the 14 neonates dem- neonates in the topiramate group, while the subsequent
onstrated hearing loss, although co-treatment with amino- study by Nunez-Ramiro et al. was slightly larger with 57
glycoside was common and there was no control group to neonates in the topiramate group [81, 82]. These relatively
compare the rate of hearing loss in neonates who received small sample sizes are a limiting factor and future studies
standard treatment without bumetanide. The study was are warranted, especially as no adverse events were attrib-
ended early due to insufficient efficacy and concern about uted to topiramate administration. There is an ongoing clini-
the potential increased prevalence of hearing loss. Subse- cal trial of topiramate in newborns with hypoxic–ischemic
quently, a randomized, double-blind, dose-escalation trial encephalopathy treated with therapeutic hypothermia that
(NCT00830531) designed to assess the safety and pharma- is currently recruiting participants (NCT01765218) with an
cokinetics of bumetanide did not show a significant differ- expected completion date of May 2022.
ence in hearing impairment between control (phenobarbital There are few data regarding topiramate for the acute
alone) and bumetanide treated groups [77]. treatment of seizures in neonates. Its use is, in large part,
In the randomized control trial, neonates (postmenstrual limited by absence of an intravenous formulation. In a recent
age 34–44 weeks) with persistent seizures despite initial large multi-center review of practice variation in treatment
loading doses of 20–40 mg/kg phenobarbital were rand- of neonatal seizures, topiramate was used < 1% of the time;
omized to subsequent treatment with either bumetanide plus however, the paper did not specify whether topiramate was
a 5–10 mg/kg phenobarbital dose or phenobarbital 5–10 mg/ prescribed for acute symptomatic seizures or neonatal epi-
kg alone (with saline as a control). There was a statistically lepsy [26]. A retrospective case series of neonates treated

13
J. M. Ziobro et al.

with topiramate suggests tolerability and safety, as well as are important to help guide clinical use. Review of the lit-
efficacy in some patients [83], but larger prospective studies erature demonstrates two patients who received lacosamide
are needed. at age 1 month with no adverse events reported [92, 93].
There is a planned, multi-center, open label randomized
Brivaracetam active comparator study to evaluate the efficacy, safety, and
pharmacokinetics of lacosamide in neonates with repeated
Brivaracetam is a n-propyl analogue of levetiracetam electroencephalographic neonatal seizures (NCT04519645).
designed with high affinity for the synaptic vesicle protein
2A (SV2A), a glycoprotein present in the membranes of syn- Ketamine
aptic vesicles. Brivaracetam is well tolerated in the setting of
renal impairment, has minimal interactions with other medi- Ketamine is a non-competitive N-methyl-D-aspartate
cations, and is available in intravenous formulation. Like (NMDA) receptor antagonist that crosses the blood–brain
levetiracetam, the precise mechanism of this medication is barrier and is clinically used as a dissociative anesthetic.
not known, and these drugs may each act differently on the During refractory status epilepticus there is loss of GABA
SV2A protein. Animal studies suggest that there may be receptor inhibition as GABA receptors internalize and the
developmental differences in SV2A with reduced expression concentration of NMDA receptors increases [94, 95]. This
in newborn models, although antiseizure properties of bri- loss of GABA inhibition and activation of NMDA receptors
varacetam are demonstrated in these models [84]. There are can further propagate seizure activity and contribute to self-
limited clinical data regarding brivaracetam in the newborn sustained status epilepticus [96]. In animal models, treat-
period, although this medication has been well-tolerated ment with ketamine can be used to halt this response and
in pediatric studies with side effects of mild somnolence, resolve self-sustaining refractory status epilepticus [96, 97].
psychosis/behavior changes, and stomach upset/decreased In a rat model of perinatal brain injury, exposure to ket-
appetite [85–87]. There is an active, open-label, single-arm amine resulted in a partial neuroprotective response with
study to evaluate the pharmacokinetics, efficacy, and safety reduced energy requirements and restored levels of ATP
of brivaracetam in neonates with repeated electroencepha- compared to controls [98]. However, additional in vivo ani-
lographic seizures despite initial antiseizure medication mal studies suggest neuronal apoptosis with ketamine expo-
(NCT03325439). The expected completion date is January sure and reduced cognitive testing outcomes in follow-up
2022. studies, although the exact dose and vulnerable ages are not
known [99, 100].
Lacosamide Ketamine is also used as an analgesic. Data derived from
its use in the delivery room of preterm infants at the time of
Lacosamide is a sodium-channel blocking agent with a intubation revealed no difference in neurological outcomes
novel mechanism through slow inactivation of voltage-gated at age 2 years compared with controls [101]. There may be a
sodium channels. It was introduced for adult epilepsy treat- ketamine dosing threshold and duration of treatment neces-
ment in 2008 and studies report its use in pediatric epilepsy sary for negative effect and neuronal apoptosis.
management with limited side effects even with intravenous The use of ketamine for treatment of neonatal seizures
administration [88]. Due to its action on the sodium channel, is primarily based on case reports with little information
there can be associated PR interval prolongation, although regarding dosing, safety, or efficacy [102, 103]. There are
this has rarely occurred with intravenous administration in more extensive data regarding the use of ketamine in chil-
children. dren and adults, although there are no randomized controlled
There was a potential neuroprotective effect of lacosa- trials of this agent. A review of ketamine for the treatment of
mide demonstrated in animal models of traumatic brain status epilepticus in children suggested an overall response
injury and status epilepticus [89, 90]. A model of neona- rate of 73% in children and only rare adverse effects [104].
tal arterial ischemic infarction revealed reduced areas of These encouraging data highlight the importance of future
ischemia after pretreatment with oral lacosamide, although studies to further evaluate ketamine for the treatment of
there was a dose-dependent response with higher rates of recurrent neonatal seizures and status epilepticus.
toxicity and mortality seen with dose escalation [91]. It
remains unclear whether there is a neuroprotective effect
in the newborn brain, or any associated neuronal apoptosis Preclinical Studies
associated with exposure.
There are limited data available regarding the use of Given the unique pathophysiology of neonatal seizures
lacosamide in the neonatal period. Studies aimed at charac- [14], identification of potential treatments of neonatal sei-
terizing the pharmacokinetics in term and preterm neonates zures cannot rely solely on extrapolation of antiseizure

13
Novel Therapeutics for Neonatal Seizures

medication data from older children and adults. Animal Flupiritine has been approved in Europe for decades as a
models in various species have been developed to mimic the non-opioid analgesic. It has been shown to activate G-protein
consequences of neonatal hypoxic-ischemic injury (reviewed regulated inward rectifying K + (GIRK) channels, which may
in [105]). However, the majority of hypoxia-related seizure indirectly inhibit NMDA receptor activity. Activation of GIRK
models are performed with rodents. In rats, hypoxic seizures channels leads to hyperpolarization of the neuronal membrane,
are induced only during the critical developmental window with resulting stabilization of the resting neuronal membrane
between post-natal day (P) 6–12, which roughly corresponds [114]. A series of eloquent experiments by Raol and colleagues
to term brain development in humans [106–108]. Neonatal examined the effects of flupiritine in several models of neona-
mice have been used at the P7-9, during the maximal transi- tal seizures. They initially reported flupiritine to be more effi-
tion in synaptic receptor maturation, similar to term human cacious in preventing and stopping chemoconvulsant induced
brain development [106]. seizures than phenobarbital or diazepam in neonatal rats [115].
In the hypoxia model, acute seizures are induced with They later showed efficacy in treating seizures in a hypoxia
exposure to varying degrees of global hypoxia for varying model [116] and hypoxia–ischemia model [117] of neonatal
durations. Several models display spontaneous recurrent seizures in rats. In follow-up studies, they also reported that
seizures, without significant neuronal death, following the flupiritine treatment reduced injury induced volume loss and
acute insult [106]. In contrast, hypoxia–ischemia models partially reversed learning and memory impairments [118].
consist of unilateral ligation of the common carotid artery Unfortunately, the withdrawal of flupiritine from the Euro-
in addition to a period of hypoxia exposure. This provides pean market was recommended in 2018 due to the risk of liver
a more severe insult and produces more substantial tissue injury with prolonged use [119].
damage in addition to more severe recurrent seizures in the Retigabine/ezogabine is chemically similar to flupiritine
long-term compared with the hypoxia model [106, 109]. and also showed antiseizure effects in limited preclinical
This results in the recapitulation of many of the features seen models of chemoconvulsant induced seizures in neonatal
in human neonatal brain injury, including tissue pathology, rats [120]. Ezogabine was previously FDA approved as an
cognitive/behavioral deficits, and seizures during and after adjunctive treatment for focal seizures, though it was with-
the insult [106, 110]. In addition, chemoconvulsant seizures drawn from clinical use in 2017 due to skin and eye abnor-
have been induced with kainic acid or flurothyl to assess effi- malities. Prior to withdrawal, ezogabine was explored as a
cacy of various antiseizure medications in neonatal rodents potential precision therapy for KCNQ2 developmental and
[111, 112]. epileptic encephalopathy (KCNQ2-DEE), potentially show-
Chemoconvulsant modeling is technically easier to per- ing some benefit in patients treated prior to age 6 months
form than current hypoxic-ischemic models, with consistent [121], though none of the patients received ezogabine treat-
elicitation of seizures by exposure to the chemoconvulsant ment in the neonatal period. Targeting potassium channels in
agent. This approach may allow for relatively high-throughput the acute neonatal period may be of future benefit in treating
screening of candidate anti-seizure compounds, while acute symptomatic neonatal seizures in addition to target-
providing insight into mechanisms required for appropri- ing specific genetic etiologies. A phase 3 clinical trial of
ate treatment of neonatal seizures. However, care should be a pediatric re-formulated ezogabine (XEN496) is expected
taken in interpretation of results derived from this appraoch, in 2021 as an orphan drug specifically for KCNQ2-DEE
as these models do not recapitulate the same complex cas- (NCT04639310).
cade of cellular and molecular mechanisms that lead to
hypoxic-ischemic seizures in neonates; candidate drug effi- Tyrosine Receptor Kinase B Modulators
cacy should also be examined in more physiologically rel-
evant models of hypoxic–ischemic injury. The age-dependent upregulation of the K + -Cl- co-transporter
(KCC2) plays a role in the shift of GABAergic signaling
Potassium‑Channel Openers from depolarizing to hyperpolarizing [11, 12] and thus
contributes to pharmacoresistant seizures. Excitotoxic injury
Compared with the mature brain, GABAergic inhibition has been shown to phosphorylate tyrosine receptor kinase
is decreased in the neonatal brain. Given this, potassium B (TrkB) pathway signaling [122]. Recent preclinical stud-
channels play a unique role in controlling excitability dur- ies have sought to modulate KCC2 to improve the efficacy
ing early development, as discussed above [11, 12] and of ­GABAA agonists in neonatal seizures via antagonism of
likely relate to the development and subsequent resolution TrkB signaling. In a mouse model of phenobarbital-refractory
of KCNQ2/3 related self-limited familial neonatal epilepsies postischemic seizures, the postischemic activation of
[113]. This suggests that use of potassium channel openers the TrkB pathway resulted in the hypofunction of KCC2
may be an effective approach to enhance inhibition and treat [123]. This study utilized a carotid-ligation ischemic sei-
neonatal seizures. zure model in which seizures were found to be refractory

13
J. M. Ziobro et al.

to phenobarbital at P7, but responsive to phenobarbital at Later studies examined treatment with cannabidivarin
P10, further under-scoring the age-dependent nature of neo- (CBDV), the propyl analog of cannabidiol (CBD). The
natal seizure pathophysiology. Administration of a small- effects of CBDV were age- and model-dependent, with lit-
molecule TrkB antagonist, ANA12, reversed phenobarbi- tle efficacy at P10, but significant antiseizure effects at P20
tal resistant seizures at P7 by preventing activation of the in the maximal electric shock model and DMCM-evoked
TrkB/phospholipase C, gamma 1 (PLCgamma1) pathway seizures [128]. These results suggest that targeting of the
in a dose-dependent manner, though it showed no change endocannabinoid system for the treatment of neonatal sei-
in seizure rescue at P10 [123]. Significantly, treatment with zures deserves further attention in pre-clinical studies, with
ANA12 also led to long-term benefits in improving sleep additional focus on toxicity of manipulating the endocan-
dysfunction and activity-dependent markers of cognition nabinoid system in the developing brain, given the known
[124]. Similar results were reported from follow-up stud- deleterious neurodevelopmental effects of in utero cannabis
ies utilizing a brain-derived neurotrophic factor (BDNF) exposure [129].
memetic, LM22A-4, also prevented BDNF-mediated TrkB/
PLCgamma1 pathway activation and KCC2 hypofunction Other Agents
[125]. Interestingly, the full TrkB agonists, HIOC, and
deoxygedunin also significantly rescued the phenobarbital The unique physiology of the neonatal brain raises the pos-
refractory phenotype and prevented postischemic degrada- sibility of targeting unique mechanisms to dampen hyperex-
tion of KCC2 [125]. citability. The mechanisms of pharmacoresistant status epi-
Taken together, these studies suggest that inhibition of lepticus are poorly understood, but there is a need to explore
cascades associated with endogenous BDNF/TrkB pathway mechanisms beyond regulating GABA or sodium channels,
activation may prevent the emergence of refractory neonatal which are currently the first- and second-line approaches in
seizures. Indeed, the role of TrkB activation in epileptogene- managing neonatal status epilepticus [26, 27, 130]. A recent
sis was shown in a study utilizing the TrkB inhibitor, lestaur- study in neonatal rats aimed to alter metabolism for seizure
tinib (CEP-701), to reverse TrkB phosphorylation following control, arguing that neuronal activity is highly energy-
hypoxia. This prevented seizure susceptibility to a “second dependent and requires significant glucose metabolism
hit” with a chemoconvulsant [126]. These studies provide [131]. The investigators aimed to utilize a glycolysis inhibi-
promising evidence that targeting the TrkB pathway may be tor, 2-deoxyglucose (2-DG) to compete with glucose for cell
a successful strategy for prevention of refractory seizures in entry and inhibit glycolysis and neuronal ATP production. In
neonates and could lead to improved neurodevelopmental the rat model of pilocarpine-induced neonatal status epilep-
outcomes following hypoxic–ischemic injury. ticus, 2-DG was as effective at aborting status epilepticus as
phenobarbital or levetiracetam [131]. Though this appears
to be an intriguing target in the treatment of refractory sta-
Cannabinoid‑Related Agents tus epilepticus, significant work needs to be done to ensure
that these results translate to more physiologic models of
Recent interest in cannabidiol as a novel antiseizure medica- hypoxic-ischemic induced refractory seizures and to exam-
tion has led to further examination of the manipulation of the ine adverse effects of systemic inhibition of glycolysis, even
endogenous cannabinoid (endocannabinoid) system in the if administered only for a short time.
treatment of neonatal seizures. The endocannabinoid system An additional recent study has implicated chloride chan-
is thought to modulate neuronal hyperexcitability in adult nels, specifically the voltage-dependent CLC-3 Cl- channels
brains but targeting the cannabinoid system in developing as playing a significant role in neonatal seizures [132]. The
brains is still poorly understood. investigators reported that CLC-3 Cl- channels mediated
A study designed to examine cannabinoid manipulation a large voltage-dependent outward rectifying Cl- current
in chemoconvulsant and hypoxia-induced seizure models in in neonatal, but not adult, mouse brains, and this current
P10 and P20 rats demonstrated that cannabinoid agonists was blocked with the administration of sodium-gluconate
WIN 55,212–2 (WIN) and arachidonyl-2′-chloroethylamide in vitro. Intriguingly, this in vitro finding was translated to
(ACEA) exerted anticonvulsant effects in a benzodiazepine several in vivo models. Administration of sodium gluconate
inverse agonist model of clonic seizures utilizing the chem- blocked kainic acid induced status epilepticus in neonatal,
oconvulsant methyl-6,7-dimethoxy-4-ethyl-beta-carboline- but not adult rats. Additionally, in the hypoxia–ischemia
3-carboxylate (DMCM). WIN also exhibited modest, but model of neonatal seizures, administration suppressed epi-
significant, antiseizure effects in PTZ- and acute hypoxia- leptiform bursts as well as phenobarbital and their effects
induced seizures [127]. Conversely, CB1 receptor antago- were synergistic when co-administered [132]. These results
nism increased seizure severity. are impressive and deserve further attention in the human
condition, as gluconate is already FDA approved as a food

13
Novel Therapeutics for Neonatal Seizures

additive. The authors noted that the gluconate concentrations Neonatal‑Onset Epilepsy
needed to treat seizures were rather high and that brain pen-
etration and side effects would need to be evaluated further. Though neonatal-onset epilepsies make up only 13% of
Kainate and AMPA receptors may be additional targets patients with neonatal seizures in the neonatal intensive care
for neonatal seizures, as they show peak brain expression unit [6], the early identification of epilepsy rather than acute
during late embryonic and early postnatal periods [14]. symptomatic seizures can have significant consequences for
Indeed, knocking out the GluK2 kainic acid receptor subunit guidance of management and informing prognosis in these
or blocking kainic acid receptors by a novel drug, UBP310, cases. As rapid genetic testing becomes more widely avail-
blocked excitatory neurotransmission in vitro [133]. In vivo, able, our future ability to deliver precision medicine will
GluK2 knockout mice and those treated with UBP310 were provide opportunities to improving outcomes for children
less susceptible to seizures during reoxygenation follow- with neonatal-onset epilepsies. However, until such time that
ing hypoxia [133]. Given the importance of kainate recep- rapid genetic testing is standard of care, clinicians must be
tor signaling on brain development, blocking kainic acid aware of clinical features that may indicate a genetic etiol-
receptors may result in significant toxicity, which has not ogy that may be responsive to targeted treatment. Neonatal-
been fully assessed. The noncompetitive AMPA antagonist, onset epilepsies with a genetic etiology may be related to
talampanel, suppressed hypoxia-induced neonatal seizures structural brain abnormalities, metabolic diseases, or other
and also prevented enhanced seizure susceptibility later in genetic syndromes [143]. Targeted therapies are available
life [134]. Notably, treatment with talampanel did not result for some genetic etiologies of neonatal-onset epilepsy, with
in any increase in cell death, suggesting that AMPA receptor others in preclinical development (Table 2).
antagonism may be a safe and effective target for hypoxia-
induced neonatal seizures. Structural Brain Abnormalities
Lastly, bumepamine, a lipophilic benzylamine derivative
of bumetanide has been considered for use in neonatal sei- Structural brain anomalies may be identified by in utero
zures in preclinical models. Bumepamine was first devel- imaging or with early magnetic resonance imaging (MRI)
oped in 1978 by Nielsen and Feit [135]. It demonstrates 80% during initial seizure work-up. A recent report of neona-
lower diuretic activity and lower ionization rate at physiolog- tal epilepsy found that ~ 29% of neonatal-onset epilepsies
ical pH compared to bumetanide. Additionally, bumepamine were related to congenital brain malformations [6]. Con-
has shown a more potent dose dependent effect as compared genital malformations of brain development may be mul-
to bumetanide in potentiating the effect of phenobarbital in tifactorial, though multiple pathogenic genes and chromo-
animal models of chronic epilepsy, although the mechanism somal anomalies have been implicated. Early imaging is
of this effect is not fully understood [136]. These factors essential to narrowing the differential diagnosis, as many
have prompted interest in this compound as a treatment for of these malformations are due to genetic syndromes that
neonatal seizures. However, data at this time are limited to may be associated with extracerebral congenital anoma-
animal models. lies. Early consultation with a geneticist is recommended

Table 2  Select genetic neonatal epilepsies with targeted treatments


Genetic disorder Current targeted treatments Emerging treatments

Tuberous sclerosis complex (TSC1/TSC2) Vigabatrin Everolimus


mTOR-related structural epilepsies (DEPDC5, Everolimus, consider early surgery Rapamycin
TSC1, TSC2, AKT3, MTOR) for infants with structural seizure
etiology
Pyridoxine-dependent epilepsy (ALDH7A1) Pyridoxine, lysine restriction, arginine ALDH7A1 directed antisense oligonucleotide
(ASO); upstream reduction therapy
Cerebral folate transport deficiency (FOLR1) Folinic acid
Molybdenum cofactor deficiency (MOCS1) Cyclic pyranopterin monophosphate (cPNP), viral
vectors to increase hepatic expression of molyb-
dopterin synthase
KCNQ2-DEE Phenytoin, carbamazepine Ezogabine
SCN1A-DEE (with Thr226Met pathogenic variant) Phenytoin, carbamazepine
SCN2A Phenytoin, carbamazepine, lacosamide
KCNT1 Quinidine Bepridil

DEE developmental and epileptic encephalopathy

13
J. M. Ziobro et al.

to ensure that the appropriate diagnostic tests are pur- treatment-resistant focal seizures due to a structural etiol-
sued [129]. Isolated structural malformations can lead to ogy, even those with a genetic basis [149].
neonatal seizures due to focal or diffuse abnormalities in
brain structure or neuronal migration, including agenesis Metabolic Disorders
of the corpus callosum (L1CAM, ZEB2, TUBA1, TUBB2A,
etc.), polymicrogyria (ADGRG1, AKT2, MTOR, PTEN, Though rare, it is important to recognize treatable metabolic
etc.), lissencephaly (PAFAH1B1, TUBA1A, DCX, ARX, neonatal epileptic encephalopathies (reviewed in [150]).
FKTN), schizencephaly (EMX2, SIX3, SHH, COL4A1), Clinical features that may raise suspicion for an underly-
megalencephaly (MTOR, SHH, PIK3CA, LYK5/STRADA), ing metabolic disorder include an initial symptom-free
holoprosencephaly (SHH, ZIC2, SIX3, TGIF1), and het- period followed by feeding difficulty and lethargy, hypoto-
erotopias (RELN, DCX, TUBBA1A, etc.) [144]. Though nia, respiratory distress and abnormal laboratory test result
age of seizure onset can vary in patients with structural including hypoglycemia, non-anion gap metabolic acidosis,
abnormalities, most with an early presentation of seizures ketosis, hyperammonemia, elevated lactate, and transamini-
have a disorder of neuronal migration [6]. Neonates with tis [136, 137, 145]. Seizures in metabolic disorders may be
epilepsy due to brain malformations are more likely to die either a primary manifestation or secondary to metabolic
or to be discharged to hospice care than those with other abnormalities and EEG may show a burst-suppression or
genetic epileptic encephalopathies [6], underscoring the hypsarrhythmia pattern [145]. Imaging may show evidence
severity of their epilepsy and need for additional thera- of severe injury without a clinical history to suggest an
peutic interventions. obvious hypoxic injury at birth [145]. Magnetic resonance
Targeted therapeutics in structurally related neonatal- spectrography (MRS) may be an essential tool in identifying
onset epilepsies are limited. Though people with tuberous suspected metabolic disorders [137].
sclerosis complex rarely have seizures starting in the neo- Potentially treatable causes of metabolic neonatal-onset
natal period, the ability to identify these infants as high-risk seizures include pyridoxine-dependent epilepsy, PNPO defi-
patients may allow for early intervention to prevent epilep- ciency, nonketotic hyperglycinemia, organic acidurias, urea
togenesis. A recent study of infants with tuberous sclero- cycle disorders, peroxisomal disorders, folinic acid-responsive
sis complex (TSC1, TSC2) aimed to improve outcomes at seizures, biotinidase deficiency, molybdenum cofactor
24 months by treating with vigabatrin prior to seizure onset, deficiency, sulphite oxidase deficiency, and congenital
but after the first interictal discharges were detected, versus disorders of glycosylation (reviewed in [144]). Even with
conventional treatment once seizures had developed [145]. appropriate treatments, responses are often varied; metabolic
Children in the early treatment group, compared with con- epileptic encephalopathies are frequently associated with
trols, had a later onset of epilepsy and a decreased incidence poor outcomes. This underscores the need for early identi-
of infantile spasms and refractory epilepsy. Interestingly, fication and the development of more precise and effective
there was no significant difference in developmental out- targeted therapies.
comes at 24 months of age, suggesting that developmental Preclinical and clinical studies of therapeutics for meta-
delays related to tuberous sclerosis complex may be some- bolic epileptic encephalopathies are limited. There are cur-
what independent from seizure severity [145]. However, this rent clinical trials of cyclic pyranopterin monophosphate
is contradictory to larger long-term studies that suggest that (cPNP) for the treatment of Molybdenum cofactor deficiency
early onset and severe epilepsy in the first 2 years of life is (NCT02629393) after treatment in an initial compassionate-
associated with long-term intellectual disabilities in indi- use cohort showed safety and efficacy, most notably in the
viduals with tuberous sclerosis complex [146]. Given this patients who were treated before the onset of recurrent sei-
disconnect, longer-term evaluation of children who received zures [151]. Preclinical studies are also examining the use of
early treatment with vigabatrin deserves evaluation, as the viral vectors to increase hepatic expression of molybdopterin
suppression of epileptogenesis with early treatment remains synthase [152].
intriguing. Current treatments of pyridoxine-dependent epilepsy
The mammalian target of rapamycin (mTOR) pathway have focused on pyridoxine supplementation, with some
includes several genes that may be related to early-onset evidence supporting additional lysine restriction and argi-
structural epilepsies including DEPDC5, TSC1, TSC2, nine supplementation [153]. Emerging therapeutic strate-
AKT3, and MTOR. The use of mTor inhibitors, such as gies for pyridoxine-dependent epilepsy include ALDHA7A1
everolimus and rapamycin, have shown limited success in directed antisense therapy [154] and targeting of upstream
the treatment of epilepsy in these disorders, though they lysine degradation pathways to prevent the build-up of toxic
have not been fully explored in the neonatal period or prior metabolites [153, 155]. Gene therapies for inherited neuro-
to seizure onset [147, 148]. Additionally, early evaluation metabolic diseases continue to be investigated as a means
for epilepsy surgery may be considered in patients with of precision therapy for these rare diseases. Importantly,

13
Novel Therapeutics for Neonatal Seizures

immune tolerance to foreign proteins in the neonate may be sodium-channel blockers act in a potassium channel muta-
an important factor in successful gene or protein replace- tion, though the authors suggested that voltage-gated sodium
ment therapies, making early diagnosis essential for success- channels co-localize with KCNQ potassium channels and
ful treatment [156, 157]. that modulation of one channel may significantly affect the
function of the channel complex [161].
Genetic Neonatal Epilepsy Syndromes Pathogenic variants in voltage-gated sodium channel
subunits, including SCN1A and SCN2A, are associated with
Neonatal epilepsy syndromes often present in the first days several neonatal-onset epilepsy syndromes. Loss-of-function
to weeks of life. Clinical identification of a genetic neona- mutations in SCN1A are most commonly implicated in Dravet
tal epilepsy syndrome is useful to establish a diagnosis and syndrome with seizure onset in the first year of life. How-
etiology, understand the pathophysiology, guide treatment, ever, SCN1A is rarely implicated in epilepsy of infancy with
estimate prognosis, and identify expected comorbidities migrating focal seizures (EIMFS) and early onset SCN1A-
[158]. Interestingly, neonatal-onset epilepsies have a broad DEE with seizure onset in the neonatal period [162]. It is
range of phenotypic severity, with some self-limited and notable that in SCN1A-DEE, most patients have a recurrent
others severe and progressive, even though they may have de novo missense mutation, Thr226Met variant, which is
common genetic loci. Monogenetic causes of neonatal-onset proposed to create a dual-action, gain-of-function and loss-
epilepsies have been reviewed extensively as our diagnos- of-function phenotype, contrary to the loss-of-function phe-
tic testing and understanding of specific genetic disorders notype most commonly seen in Dravet syndrome [163, 164].
continue to expand [130, 144]. Yet, targeted therapeutic This would suggest that children with SCN1A-DEE may have
options remain inadequate and only clinically relevant to a seizures responsive to sodium-channel blockers, even though
few known disorders. these medications are contraindicated in SCN1A-associated
In a recent study of neonatal-onset epilepsies, autoso- Dravet syndrome. However, this has not been adequately sup-
mal dominant pathogenic variants in KCNQ2 were the most ported in the limited published literature [163].
common finding on genetic testing [6]. KCNQ2 encodes a Pathogenic variants in SCN2A have been associated with
voltage-gated potassium channel (Kv7.2) responsible for self-limited familial or nonfamilial neonatal epilepsies as
dichotomous epilepsy syndromes. Self-limited familial well as more severe developmental and epileptic encepha-
neonatal epilepsy due to (often familial) pathogenic vari- lopathies with neonatal-onset, in addition to other neurologic
ants in KCNQ2 often presents in the first few days of life disorders with onset beyond the neonatal period [165]. There
with focal tonic seizures, often with apnea, vocalizations, is wide phenotypic variation associated with SCN2A patho-
or autonomic changes [154]. Seizures are typically brief, genic variants, though missense variants in epileptic enceph-
but cluster and require treatment. Interictal EEG is normal. alopathies are often activating, suggesting gain of function
Seizures abate within the first year of life, though up to 15% physiology [166]. As such, these children’s seizures often
of individuals may develop epilepsy later in life [159]. Con- respond to sodium-channel blockers (e.g., phenytoin and
versely, KCNQ2 related neonatal encephalopathy presents carbamazepine [166]). Another recent case series showed
with a much more severe phenotype. Patients present with response to lacosamide in two children with SCN2A associ-
early onset, often tonic seizures that are difficult to control, ated infantile onset epilepsy after they had both shown an
and have a severely abnormal interictal EEG that may dis- incomplete response to phenytoin [93].
play a burst suppression pattern or multifocal discharges. The syndrome of epilepsy of infancy with migrating focal
Seizure frequency tends to decrease over time, but chil- seizures (EIMFS) is most commonly associated with patho-
dren have poor developmental outcomes. Targeted therapy genic variants in the KCNT1 gene, which encodes a subunit
with the potassium channel opener, ezogabine, was poten- of a sodium-dependent potassium channel [167]. Seizures
tially beneficial in treating refractory seizures in patients in EIMFS often begin in the neonatal period and are char-
with KCNQ2 encephalopathy, especially in patients treated acterized by focal seizures that appear to migrate from one
before 6 months of age, whereas patients treated at older region to another on EEG. Seizures are often refractory to
ages showed a less robust response [121]. Unfortunately, antiseizure medications and there is associated arrest of
ezogabine was discontinued by the manufacturer in 2017 psychomotor development. Pathogenic variants in KCNT1
due to skin discoloration and potential vision loss due to typically have a gain-of-function effect. Precision treatment
retinal pigmentation [160]. Interestingly, treatment with with the potassium-channel blocker quinidine has been
sodium channel blockers, such as carbamazepine or phe- reported to be effective in controlling seizures in a small
nytoin, has been reported to be relatively effective in treat- subset of patients with KCNT1 [168–170] although more
ing seizures in KCNQ2-DEE. In a recent study of children recent, larger case series have reported mixed effects [171,
with KCNQ2-DEE, 53% were seizure free on carbamazepine 172]. Additional in vitro studies have been conducted with
and 33% responded the phenytoin [161]. It is unclear how the KCNT1 current blocker, bepridil, showing more potent

13
J. M. Ziobro et al.

KCNT1 blockade than quinidine for two patient-associated The need for new treatments for neonatal seizures is evi-
KCNT1 variants, though it was not trialed in patients [173]. dent. However, despite limited new antiseizure medications
The authors suggested that bepridil may provide another for neonatal seizures in the past, there is hope for the future.
agent for precision medicine in KCNT1 associated EIMFS, Emerging clinical trials and preclinical data are encouraging,
though it must be noted that functional evaluation of spe- as is the expanding genetic landscape and potential for pre-
cific mutations should be considered prior to treatment with cision therapy. Long-term developmental outcomes remain
KCNT1-blocking agents [169, 173]. of paramount importance and our hope is that in the next
decade we will have antiseizure medications that are not only
more effective than our current treatment options for neona-
tal seizures, but also decrease the risk of neuronal injury in
Conclusions the developing brain. The combination of these features will
provide the best hope for the future of neonates with seizures.
The treatment of neonatal seizures presents a unique chal-
lenge in pediatric neurology and neonatology due to the Supplementary Information  The online version contains supplemen-
unique pathophysiologic features of the immature brain. tary material available at https://​doi.​org/​10.​1007/​s13311-​021-​01085-8
Despite significant advances in preclinical research, cur-
rent available therapeutics for acute symptomatic neonatal Required Author Forms  Disclosure forms provided by the authors are
seizures have not changed substantially in decades. By con- available with the online version of this article.
trast, the Food and Drug Administration (FDA) and Euro-
pean Medicines Agency (EMA) have approved greater than
20 new antiseizure medications for adults, none of which
are approved for newborn infants [174]. The challenges in References
bringing new drug treatments to clinical trials for neonatal
seizures are great. There are ethical considerations in balanc- 1. Lanska MJ, Lanska DJ. Neonatal Seizures in the United States:
ing the risks and benefits in administering a new drug when Results of the National Hospital Discharge Survey, 1980-1991.
Neuroepidemiology. 1996;15:117–125.
safety and efficacy data from adults cannot be adequately 2. Saliba RM, Annegers JF, Waller DK, et al. Incidence of neonatal
extrapolated to neonates. Logistic considerations are even seizures in Harris County, Texas, 1992-1994. Am J Epidemiol.
greater, as participants are recruited within hours to days 1999;150:763–769.
following birth, with consent provided by parents who have 3. Glass HC, Pham TN, Danielsen B, et al. Antenatal and Intrapar-
tum Risk Factors for Seizures in Term Newborns: A Population-
likely also gone through a series of parallel medically and Based Study, California 1998-2002. J Pediatr. 2009;154:24-28.
emotionally traumatic events. In addition, adequate EEG e1.
monitoring—with real-time interpretation—and ability 4. Lloyd RO, O’Toole JM, Pavlidis E, et al. Electrographic Seizures
to perform frequent blood monitoring is necessary to per- during the Early Postnatal Period in Preterm Infants. J Pediatr.
2017;187:18–25.e2.
form high quality trials. It is also recommended to include 5. Glass HC, Shellhaas RA, Wusthoff CJ, et al. Contemporary
a control arm; either with current first-line therapy (i.e., Profile of Seizures in Neonates: A Prospective Cohort Study. J
phenobarbital) or to design trials that directly compare sec- Pediatr. 2016;174:98-103.e1.
ond-line treatment options for seizures that persist despite 6. Shellhaas RA, Wusthoff CJ, Tsuchida TN, et al. Profile of neo-
natal epilepsies. Neurology. 2017;89:893–899.
phenobarbital. These steps require close multidisciplinary 7. Uria-Avellanal C, Marlow N, Rennie JM. Outcome following
collaboration. neonatal seizures. Semin Fetal Neonatal Med. 2013;18:224–232.
Given these challenges, collaboration across multi- 8. Glass HC, Soul JS, Chu CJ, et al. Response to antiseizure medi-
ple centers is essential for future trials. Rapid distinction cations in neonates with acute symptomatic seizures. Epilepsia.
2019;60:e20–e24.
between acute symptomatic seizures versus neonatal-onset 9. Painter MJ, Scher MS, Stein AD, et al. Phenobarbital Compared
epilepsy with early appropriate imaging, laboratory and with Phenytoin for the Treatment of Neonatal Seizures. N Engl
genetic testing is crucial as we move toward precision medi- J Med. 1999;341:485–489.
cine. Use of electronic health records and learning health- 10. Yamada J, Okabe A, Toyoda H, et al. Cl- uptake promoting depo-
larizing GABA actions in immature rat neocortical neurones is
care systems [175]—particularly for identification of chil- mediated by NKCC1. J Physiol. 2004;557:829–841.
dren with rare neonatal-onset epilepsies or for long-term 11. Dzhala VI, Talos DM, Sdrulla DA, et  al. NKCC1 trans-
follow-up data—could allow study sites to maintain consist- porter facilitates seizures in the developing brain. Nat Med.
ent data collection and lead to more robust interpretation of 2005;11:1205–1213.
12. Rivera C, Voipio J, Payne JA, et al. The K+/Cl− co-transporter
the gathered data. The learning healthcare system construct KCC2 renders GABA hyperpolarizing during neuronal maturation.
could also be applied for assessment of implementation of Nature. 1999;397:251–255.
new trial results in real-world clinical practice.

13
Novel Therapeutics for Neonatal Seizures

13. Carrasco M, Stafstrom CE. How Early Can a Seizure Happen? 33. Al-Muhtasib N, Sepulveda-Rodriguez A, Vicini S, et al. Neonatal
Pathophysiological Considerations of Extremely Premature phenobarbital exposure disrupts GABAergic synaptic maturation
Infant Brain Development. Dev Neurosci. 2018;40:417–436. in rat CA1 neurons. Epilepsia. 2018;59:333–344.
14. Nardou R, Ferrari DC, Ben-Ari Y. Mechanisms and effects of 34. Kaushal S, Tamer Z, Opoku F, et al. Anticonvulsant drug-
seizures in the immature brain. Semin Fetal Neonatal Med. induced cell death in the developing white matter of the rodent
2013;18:175–184. brain. Epilepsia. 2016;57:727–734.
15. Dzhala VI, Kuchibhotla KV., Glykys JC, et  al. Progressive 35. Forcelli PA, Kim J, Kondratyev A, et al. Pattern of antiepileptic
NKCC1-Dependent Neuronal Chloride Accumulation during drug-induced cell death in limbic regions of the neonatal rat
Neonatal Seizures. J Neurosci. 2010;30:11745–11761. brain. Epilepsia. 2011;52:e207–e211.
16. Stafstrom CE, Thompson JL, Holmes GL. Kainic acid seizures in 36. Noguchi KK, Fuhler NA, Wang SH, et al. Brain pathology
the developing brain: status epilepticus and spontaneous recur- caused in the neonatal macaque by short and prolonged
rent seizures. Dev Brain Res. 1992;65:227–236. exposures to anticonvulsant drugs. Neurobiol Dis [Internet].
17. Miller S, Goasdoue K, Björkman St. Neonatal seizures and 2021;149:105245. Available from: https://​doi.​org/​10.​1016/j.​
disruption to neurotransmitter systems. Neural Regen Res. nbd.​2020.​105245. Accessed February 20, 2021. 
2017;12:216-217. 37. Quinlan SMM, Rodriguez-Alvarez N, Molloy EJ, et al. Com-
18. Shellhaas RA, Chang T, Tsuchida T, et al. The American Clinical Neuro- plex spectrum of phenobarbital effects in a mouse model
physiology Societyʼs Guideline on Continuous Electroencephalogra- of neonatal hypoxia-induced seizures. Sci Rep [Internet].
phy Monitoring in Neonates. J Clin Neurophysiol. 2011;28:611–617. 2018;8:9986. Available from: https:// ​ w ww. ​ n ature. ​ c om/
19. Lemmon ME, Donohue PK, Parkinson C, et al. Communication ​artic​les/​s41598-​018-​28044-2. Accessed January 1, 2021. 
Challenges in Neonatal Encephalopathy. Pediatrics [Internet]. 38. Forcelli PA, Kozlowski R, Synder C, et al. Effects of Neo-
2016;138:e20161234. Available from: www.​aappu​blica​tions.​org/​ natal Antiepileptic Drug Exposure on Cognitive, Emotional,
news. Accessed January 1, 2021  and Motor Function in Adult Rats. J Pharmacol Exp Ther
20. Lemmon ME, Donohue PK, Parkinson C, et  al. Parent [Internet]. 2012 [cited 2020 Dec 30];340:558–566. Available
Experience of Neonatal Encephalopathy. J Child Neurol. from: https://​doi.​org/​10.​1124/​jpet.​111.​188862.
2017;32(3):286–292. 39. Singh D, Kumar P, Narang A. A randomized controlled trial of
21. Hill E, Glass HC, Kelley K, et al. Seizures and Antiseizure Medi- phenobarbital in neonates with hypoxic ischemic encephalopa-
cations are Important to Parents of Newborns With Seizures. thy. J Matern Neonatal Med. 2005;18:391–395.
Pediatr Neurol. 2017;67:40–44. 40. Young L, Berg M, Soll R. Prophylactic barbiturate use for the pre-
22. Soul JS, Pressler R, Allen M, et al. Recommendations for the vention of morbidity and mortality following perinatal asphyxia.
design of therapeutic trials for neonatal seizures. Pediatr Res. Cochrane Database Syst. Rev.[Internet]; 2016:5(5):CD001240.
2019;85:943–954. Available from: https://​doi.​org/​10.​1002/​14651​858.​CD001​240.​
23. WHO Department of Mental Health and Substance Abuse, WHO pub3/​full. Accessed December 30, 2020. 
Department of Maternal Newborn Child and Adolescent Health, 41. Bohannon KK, Leung N, Cook AM, et al. Evaluation of two
OASI Department of Mental Retardation Unit of Neurology and fosphenytoin loading dose regimens and monitoring in infants
Clinical Neurophysiopathology. Guidelines on neonatal seizures. and neonates less than six months of age. J Pediatr Pharmacol
[Internet]. World Health Organ. 2011. Available from: https://​apps.​ Ther. 2020;25:617–622.
who.​int/​iris/​handle/​10665/​77756. Accessed November 30, 2020. 42. Morton LD. Clinical experience with fosphenytoin in children.
24. Blume HK, Garrison MM, Christakis DA. Neonatal seizures: J Child Neurol. 1998;13:19–22.
Treatment and treatment variability in 31 United States pediatric 43. Al Za’abi M, Lanner A, Xiaonian X, et al. Application of routine
hospitals. J Child Neurol. 2009;24:148–154. monitoring data for determination of the population pharmacoki-
25. Dizon MLV, Rao R, Hamrick SE, et al. Practice variation in anti- netics and enteral bioavailability of phenytoin in neonates and
epileptic drug use for neonatal hypoxic-ischemic encephalopa- infants with seizures. Ther Drug Monit. 2006;28:793-799.
thy among regional NICUs. BMC Pediatr. 2019;19:67. Accessed 44. Alix V, James M, Jackson AH, et al. Efficacy of Fosphenytoin as
November 30, 2021. First-Line Antiseizure Medication for Neonatal Seizures Com-
26. Ahmad KA, Desai SJ, Bennett MM, et al. Changing antiepileptic pared to Phenobarbital. J Child Neurol. 2021;36:30-37.
drug use for seizures in US neonatal intensive care units from 45. Sicca F, Contaldo A, Rey E, et al. Phenytoin administration in
2005 to 2014. J Perinatol. 2017;37:296–300. the newborn and infant. Brain Dev. 2000;22:35–40.
27. Shellhaas RA, Chang T, Wusthoff CJ, et al. Treatment Duration 46. Kriel RL, Cifuentes RF. Fosphenytoin in infants of extremely low
After Acute Symptomatic Seizures in Neonates: A Multicenter birth weight. Pediatr Neurol. 2001;24:219–221.
Cohort Study. J Pediatr. 2017;181:298-301.e1. 47. Takeoka M, Kishnamoorthy K, Soman TB, et al. Fosphenytoin
28. Le VT, Abdi HH, Sánchez PJ, et al. Neonatal Antiepileptic Medi- in Infants. J Child Neurol. 1997;13:537–540.
cation Treatment Patterns: A Decade of Change. Am J Perinatol. 48. Bittigau P, Sifringer M, Ikonomidou C. Antiepileptic drugs
2021;38:469-476. and apoptosis in the developing brain. Ann N Y Acad Sci.
29. Ben-Ari Y, Cherubini E, Corradetti R, et  al. Giant synaptic 2003;993:103-114.
potentials in immature rat CA3 hippocampal neurones. J Physiol. 49. Grinspan ZM, Shellhaas RA, Coryell J, et al. Comparative effec-
1989;416:303–325. tiveness of levetiracetam vs phenobarbital for infantile epilepsy.
30. Cherubini E, Rovira C, Gaiarsa JL, et al. GABA mediated excita- JAMA Pediatr. 2018;172:352-360.
tion in immature rat CA3 hippocampal neurons. Int J Dev Neu- 50. Abend NS, Gutierrez-Colina AM, Monk HM, et  al. Lev-
rosci. 1990;8:481–490. etiracetam for treatment of neonatal seizures. J Child Neurol.
31. Sharpe C, Reiner GE, Davis SL, et al. Levetiracetam Versus 2011;26:465–470.
Phenobarbital for Neonatal Seizures: A Randomized Controlled 51. Ramantani G, Ikonomidou C, Walter B, et al. Levetiracetam:
Trial. Pediatrics [Internet]. 2020;145:e20193182. Available at: Safety and efficacy in neonatal seizures. Eur J Paediatr Neurol.
https://p​ ediat​ rics.a​ appub​ licat​ ions.o​ rg. Accessed January 1, 2021. 2011;15:1–7. 
32. Lynch NE, Stevenson NJ, Livingstone V, et al. The temporal 52. Khan O, Cipriani C, Wright C, et al. Role of intravenous lev-
evolution of electrographic seizure burden in neonatal hypoxic etiracetam for acute seizure management in preterm neonates.
ischemic encephalopathy. Epilepsia. 2012;53:549–557. Pediatr Neurol. 2013;49:340–343.

13
J. M. Ziobro et al.

53. Kurtom W, Courchia B, Pensirikul A, et al. Lack of response 71. Weeke LC, Schalkwijk S, Toet MC, et  al. Lidocaine-Asso-
to treatment with levetiracetam in extreme preterm infants with ciated Cardiac Events in Newborns with Seizures: Incidence,
seizures. J Perinatol. 2019;39:1480–1484. Symptoms and Contributing Factors. Neonatology [Internet].
54. Han JY, Moon CJ, Youn YA, et al. Efficacy of levetiracetam for 2015;108:130–136. Available from: www.​k arger.​c om/​n eo.
neonatal seizures in preterm infants. BMC Pediatr. 2018;18:1–6. Accessed January 1, 2021.
55. Strasser K, Lueckemann L, Kluever V, et al. Dose-dependent 72. Dzhala VI, Brumback AC, Staley KJ. Bumetanide enhances
effects of levetiracetam after hypoxia and hypothermia in the phenobarbital efficacy in a neonatal seizure model. Ann Neurol.
neonatal mouse brain. 2016 [cited 2020 Dec 30]; Available from: 2008;63:222–235.
https://​doi.​org/​10.​1016/j.​brain​res.​2016.​05.​040. 73. Johne M, Römermann K, Hampel P, et al. Phenobarbital and
56. Falsaperla R, Mauceri L, Pavone P, et al. Short-term neurodevel- midazolam suppress neonatal seizures in a noninvasive rat model
opmental outcome in term neonates treated with phenobarbital of birth asphyxia, whereas bumetanide is ineffective. Epilepsia.
versus levetiracetam: A single-center experience. Behav Neu- 2021;62:920–934.
rol [Internet]. 2019;2019:3683548. Available at: https://​www. 74. Puskarjov M, Kahle KT, Ruusuvuori E, et al. Pharmacothera-
​hinda​wi.​com. Accessed January 1, 2021.  peutic targeting of cation-chloride cotransporters in neonatal
57. Arican P, Olgac Dundar N, Mete Atasever N, et al. Comparison seizures. Epilepsia. 2014;55:806–818.
of the neurocognitive outcomes in term infants treated with lev- 75. Pressler RM, Boylan GB, Marlow N, et al. Bumetanide for the
etiracetam and phenobarbital monotherapy for neonatal clinical treatment of seizures in newborn babies with hypoxic ischaemic
seizures. Seizure [Internet]. 2020;80:71–74. Available at: https://​ encephalopathy (NEMO): An open-label, dose finding, and fea-
www.​seizu​re-​journ​al.​com. Accessed November 30, 2020. sibility phase 1/2 trial. Lancet Neurol. 2015;14:469–477.
58. Tsuchida TN, Wusthoff CJ, Shellhaas RA, et al. American clini- 76. Thoresen M, Sabir H. Epilepsy: Neonatal seizure still lack safe
cal neurophysiology society standardized EEG terminology and and effective treatment. Nat Rev - Neurol. 2015;11:311–312.
categorization for the description of continuous eeg monitoring 77. Soul JS, Bergin AM, Stopp C, et al. A Pilot Randomized, Con-
in neonates: Report of the american clinical neurophysiology trolled, Double‐Blind Trial of Bumetanide to Treat Neonatal
society critical care monitoring committee. J Clin Neurophysiol. Seizures. Ann Neurol. 2021;89:327-340.
2013;30:161–173. 78. Follett PL, Deng W, Dai W, et al. Glutamate Receptor-Mediated
59. Pavlidis E, Spagnoli C, Pelosi A, et al. Neonatal status epi- Oligodendrocyte Toxicity in Periventricular Leukomalacia: A
lepticus: Differences between preterm and term newborns. Protective Role for Topiramate. J Neurosci. 2004;24:4412–4420.
Eur J Paediatr Neurol [Internet]. 2015;19:314–319. Available 79. Glier C, Dzietko M, Bittigau P, et  al. Therapeutic doses of
from:  https://​doi.​org/​10.​1016/j.​ejpn.​2015.​01.​002. Accessed topiramate are not toxic to the developing rat brain. Exp Neurol.
November 30, 2020. 2004;187:403–409.
60. Torolira D, Suchomelova L, Wasterlain CG, et al. Phenobarbital 80. Silverstein FS, Ferriero DM. Off-Label Use of Antiepileptic
and midazolam increase neonatal seizure-associated neuronal Drugs for the Treatment of Neonatal Seizures. Pediatr Neurol.
injury. Ann Neurol. 2017;82:115–120. 2008;39:77–79.
61. Dao K, Giannoni E, Diezi M, et al. Midazolam as a first-line 81. Nuñez-Ramiro A, Benavente-Fernández I, Valverde E, et al.
treatment for neonatal seizures: Retrospective study. Pediatr Int. Topiramate plus cooling for hypoxic-ischemic encephalopathy:
2018;60:498–500. A randomized, controlled, multicenter, double-blinded trial. Neo-
62. Castro Conde JR, Hernández Borges AA, Doménech Martínez natology. 2019;116:76–84.
E, et al. Midazolam in neonatal seizures with no response to 82. Filippi L, Fiorini P, Catarzi S, et al. Safety and efficacy of topira-
phenobarbital. Neurology. 2005;64:876-879. mate in neonates with hypoxic ischemic encephalopathy treated
63. Boylan GB, Rennie JM, Chorley G, et al. Second-line anticon- with hypothermia (NeoNATI): a feasibility study. J Matern Neo-
vulsant treatment of neonatal seizures: A video-EEG monitoring natal Med. 2018;31:973–980.
study. Neurology. 2004;62:486-488. 83. Glass HC, Poulin C, Shevell MI. Topiramate for the treatment of
64. Shany E, Benzaqen O, Watemberg N. Comparison of continuous neonatal seizures. Pediatr Neurol. 2011;44:439–442.
drip of midazolam or lidocaine in the treatment of intractable 84. Dupuis N, Matagne A, Staelens L, et al. Anti-ictogenic and antie-
neonatal seizures. J Child Neurol. 2007;22:255–259. pileptogenic properties of brivaracetam in mature and immature
65. Duerden EG, Guo T, Dodbiba L, et  al. Midazolam Dose rats. Epilepsia. 2015;56:800–805.
Correlates with Abnormal Hippocampal Growth and Neu- 85. Nissenkorn A, Tzadok M, Bar-Yosef O, et al. Treatment with
rodevelopmental Outcome in Preterm Infants. Ann Neurol. brivaracetam in children – The experience of a pediatric epilepsy
2016;79:548-559. center. Epilepsy Behav. 2019;101(PtA):106541.
66. Favié LMA, Huitema ADR, van den Broek MPH, et al. Lido- 86. Liu E, Dilley D, McDonough B, et  al. Safety and Toler-
caine as treatment for neonatal seizures: Evaluation of previously ability of Adjunctive Brivaracetam in Pediatric Patients < 16
developed population pharmacokinetic models and dosing regi- Years with Epilepsy: An Open-Label Trial. Pediatr Drugs.
men. Br J Clin Pharmacol. 2020;86:75–84. 2019;21:291–301.
67. Malingré MM, Van Rooij LGM, Rademaker CMA, et al. Devel- 87. McGuire S, Silva G, Lal D, et al. Safety and Efficacy of Brivar-
opment of an optimal lidocaine infusion strategy for neonatal acetam in Pediatric Refractory Epilepsy: A Single-Center Clini-
seizures. Eur J Pediatr. 2006;165:598–604. cal Experience. J Child Neurol. 2020;35:102–105.
68. Van Den Broek MPH, Huitema ADR, Van Hasselt JGC, et al. 88. Welsh SS, Lin N, Topjian AA, et  al. Safety of intravenous
Lidocaine (lignocaine) dosing regimen based upon a population lacosamide in critically ill children. Seizure. 2017;52:76–80.
pharmacokinetic model for preterm and term neonates with sei- 89. Licko T, Seeger N, Zellinger C, et al. Lacosamide treatment
zures. Clin Pharmacokinet. 2011;50:461–469. following status epilepticus attenuates neuronal cell loss and
69. Weeke LC, Toet MC, Van Rooij LGM, et al. Lidocaine response alterations in hippocampal neurogenesis in a rat electrical sta-
rate in aEEG-confirmed neonatal seizures: Retrospective study of tus epilepticus model. Epilepsia. 2013;54:1176–1185.
413 full-term and preterm infants. Epilepsia. 2016;57:233–242. 90. Wang B, Dawson H, Wang H, et al. Lacosamide improves out-
70. Van Rooij LGM, Toet MC, Rademaker KMA, et al. Cardiac come in a murine model of traumatic brain injury. Neurocrit
arrhythmias in neonates receiving lidocaine as anticonvulsive Care. 2013;19:125–134.
treatment. Eur. J. Pediatr. 2004;163:637–641.

13
Novel Therapeutics for Neonatal Seizures

91. Kim GH, Byeon JH, Eun BL. Neuroprotective effect of lacosa- 113. Chapman KE, Raol YH, Brooks-Kayal A. Neonatal seizures:
mide on hypoxic-ischemic brain injury in neonatal rats. J Clin Controversies and challenges in translating new therapies from
Neurol. 2017;13:138–143. the lab to the isolette. Eur J Neurosci. 2012;35:1857–1865.
92. Arkilo D, Gustafson M, Ritter FJ. Clinical experience of intra- 114. Harish S, Bhuvana K, Bengalorkar GM, et  al. Flupirtine:
venous lacosamide in infants and young children. Eur J Paedi- Clinical pharmacology. J Anaesthesiol Clin Pharmacol.
atr Neurol. 2016;20:212–217. 2012;28:172–177.
93. Hadar F-H, Eli H, Ayelet L, et al. Lacosamide for SCN2A- 115. Raol YSH, Lapides DA, Keating JG, et al. A KCNQ channel
related intractable neonatal and infantile seizures. Epileptic opener for experimental neonatal seizures and status epilepticus.
Disord. 2018;20:440–446. Ann Neurol. 2009;65:326–336.
94. Naylor DE, Liu H, Wasterlain CG. Trafficking of GABAA 116. Sampath D, Shmueli D, White AM, et al. Flupirtine effectively
receptors, loss of inhibition, and a mechanism for pharmacore- prevents development of acute neonatal seizures in an animal
sistance in status epilepticus. J Neurosci. 2005;25:7724–7733.  model of global hypoxia. Neurosci Lett. 2015;607:46–51.
95. Chen J, Naylor DE, Wasterlain CG. Advances in the patho- 117. Sampath D, Valdez R, White AM, et al. Anticonvulsant effect
physiology of status epilepticus. Acta Neurol Scand. of flupirtine in an animal model of neonatal hypoxic-ischemic
2007;186:7–15. encephalopathy. Neuropharmacology. 2017;123:126–135.
96. Mazarati AM, Wasterlain CG. N-Methyl-D-asparate receptor 118. Sampath D, Lam PM, Laoprasert M, et al. Effects of a potassium
antagonists abolish the maintenance phase of self-sustaining channel opener on brain injury and neurologic outcomes in an
status epilepticus in rat. Neurosci Lett. 1999;265:187–190. animal model of neonatal hypoxic–ischemic injury. Pediatr Res.
97. Wasterlain CG, Chen JWY. Mechanistic and pharmacologic 2020;88:202–208.
aspects of status epilepticus and its treatment with new antiepi- 119. European Medicines Agency. Withdrawal of pain medicine flu-
leptic drugs. Epilepsia. 2008;49:63-73. pirtine endorsed [Internet]. 2018. Available from: https://​www.​
98. Spandou E, Karkavelas G, Soubasi V, et al. Effect of ketamine ema.​e uropa.​e u/​e n/​d ocum​e nts/​r efer ​r al/​f lupi​r tine-​a rtic​l e-​3 1-​
on hypoxic–ischemic brain damage in newborn rats. Brain Res. refer ​ral-​withd​rawal-​pain-​medic​ine-​flupi​r tine-​endor​sed_​en.​pdf.
1999;819:1–7. Accessed January 2, 2021.
99. Brambrink AM, Evers AS, Avidan MS, et al. Ketamine-induced 120. Forcelli PA, Soper C, Lakhkar A, et al. Anticonvulsant effect of
Neuroapoptosis in the Fetal and Neonatal Rhesus Macaque retigabine during postnatal development in rats. Epilepsy Res.
Brain. Anesthesiology. 2012;116:372–384.  2012;101:135–140.
100. Paule MG, Li M, Allen RR, et al. Ketamine anesthesia during 121. Millichap JJ, Park KL, Tsuchida T, et al. KCNQ2 encephalopa-
the first week of life can cause long-lasting cognitive deficits in thy: Features, mutational hot spots, and ezogabine treatment of
rhesus monkeys. Neurotoxicol Teratol. 2011;33:220–230. 11 patients. Neurol Genet. 2016;2;e96.
101. Elalouf C, Le Moing AG Le, Fontaine C, et al. Prospective follow- 122. Kang SK, Johnston M V., Kadam SD. Acute TrkB inhibition
up of a cohort of preterm infants < 33 WG receiving ketamine for rescues phenobarbital-resistant seizures in a mouse model of
tracheal intubation in the delivery room: Neurological outcome neonatal ischemia. Eur J Neurosci. 2015;42:2792–2804.
at 1 and 2 years. Arch Pediatr. 2018;25:295–300. 123. Carter BM, Sullivan BJ, Landers JR, et al. Dose-dependent rever-
102. Tarocco A, Ballardini E, Garani G. Use of ketamine in a newborn sal of KCC2 hypofunction and phenobarbital-resistant neonatal
with refractory status epilepticus: A case report. Pediatr Neurol. seizures by ANA12. Sci Rep. 2018;8:11987.
2014;51:154–156. 124. Kang SK, Ammanuel S, Adler DA, et al. Rescue of PB-resistant
103. Freibauer A, Jones K. KCNQ2 mutation in an infant with enceph- neonatal seizures with single-dose of small-molecule TrkB antag-
alopathy of infancy with migrating focal seizures. Epileptic Dis- onist show long-term benefits. Epilepsy Res. 2020;159:106249.
ord. 2018;20:541–544. 125. Kipnis PA, Sullivan BJ, Carter BM, et al. TrkB agonists prevent
104. Höfler J, Trinka E. Intravenous ketamine in status epilepticus. postischemic emergence of refractory neonatal seizures in mice.
Epilepsia. 2018;59:198-206. JCI Insight. 2020;5:e136007.
105. Yager JY, Ashwal S. Animal Models of Perinatal Hypoxic- 126. Obeid M, Rosenberg EC, Klein PM, et al. Lestaurtinib (CEP-701)
Ischemic Brain Damage. Pediatr Neurol. 2009;40:156–167. attenuates “second hit” kainic acid-induced seizures following
106. Sun H, Juul HM, Jensen FE. Models of hypoxia and ischemia- early life hypoxic seizures. Epilepsy Res. 2014;108:806–810.
induced seizures. J Neurosci Methods. 2016;260:252–260. 127. Huizenga MN, Wicker E, Beck VC, et al. Anticonvulsant effect of
107. Chiba S. Long-term effect of postnatal hypoxia on the seizure cannabinoid receptor agonists in models of seizures in develop-
susceptibility in rats. Life Sci. 1985;37:1597–1604. ing rats. Epilepsia. 2017;58:1593–1602.
108. Jensen FE, Applegate CD, Holtzman D, et al. Epileptogenic 128. Huizenga MN, Sepulveda-Rodriguez A, Forcelli PA. Preclini-
effect of hypoxia in the immature rodent brain. Ann Neurol. cal safety and efficacy of cannabidivarin for early life seizures.
1991;29:629–637. Neuropharmacology. 2019;148:189–198.
109. Levine S. Anoxic-Ischemic Encephalopathy in Rats. Am J Pathol. 129. Badowski S, Smith G. Cannabis use during pregnancy and post-
1960;36:1–17. partum. Can Fam Physician. 2020;66:98–103.
110. Rice JE, Vannucci RC, Brierley JB. The influence of immatu- 130. Ziobro J, Shellhaas RA. Neonatal Seizures: Diagnosis, Etiolo-
rity on hypoxic-ischemic brain damage in the rat. Ann Neurol. gies, and Management. Semin Neurol. 2020;40:246–256.
1981;9:131–141. 131. Janicot R, Stafstrom CE, Shao LR. 2-Deoxyglucose terminates
111. Zhang Y, Zhang M, Liu S, et  al. Xenon exerts anti-seizure pilocarpine-induced status epilepticus in neonatal rats. Epilepsia.
and neuroprotective effects in kainic acid-induced status epi- 2020;61:1528–1537.
lepticus and neonatal hypoxia-induced seizure. Exp Neurol. 132. Wu Z, Huo Q, Ren L, et al. Gluconate suppresses seizure activity
2019;322:113054. in developing brains by inhibiting CLC-3 chloride channels. Mol
112. Nolan SO, Hodges SL, Condon SM, et al. High seizure load Brain. 2019;12:50.
during sensitive periods of development leads to broad shifts 133. Grosenbaugh DK, Ross BM, Wagley P, et al. The Role of Kainate
in ultrasonic vocalization behavior in neonatal male and female Receptors in the Pathophysiology of Hypoxia-Induced Seizures
C57BL/6J mice. Epilepsy Behav. 2019;95:26–33. in the Neonatal Mouse. Sci Rep. 2018;8:7035.

13
J. M. Ziobro et al.

134. Aujla PK, Fetell MR, Jensen FE. Talampanel suppresses the 155. Al Teneiji A, Bruun TUJ, Cordeiro D, et al. Phenotype, biochem-
acute and chronic effects of seizures in a rodent neonatal seizure ical features, genotype and treatment outcome of pyridoxine-
model. Epilepsia. 2009;50:694–701. dependent epilepsy. Metab Brain Dis. 2017;32:443–451.
135. Nielsen O., Feit PW. Structure-activity relationships of amin- 156. Poletti V, Biffi A. Gene-Based Approaches to Inherited Neuro-
obenzoic acid diuretics and related compounds (1). Diuret Agens. metabolic Diseases. Hum Gene Ther. 2019;30:1222–1235.
1978;83:12–23. 157. Hinderer C, Bell P, Louboutin J-P, et al. Neonatal tolerance
136. Brandt C, Seja P, Töllner K, et al. Bumepamine, a brain-permeant induction enables accurate evaluation of gene therapy for MPS I
benzylamine derivative of bumetanide, does not inhibit NKCC1 in a canine model. Mol Genet Metab. 2016;119:124–130.
but is more potent to enhance phenobarbital’s anti-seizure efficacy. 158. Pearl PL. Epilepsy Syndromes in Childhood. Continuum.
Neuropharmacology. 2018;143:186–204. 2018;24:186–209.
137. Guillet R, Kwon J. Seizure recurrence and developmental dis- 159. Maljevic S, Lerche H. Potassium channel genes and benign famil-
abilities after neonatal seizures: Outcomes are unrelated to use ial neonatal epilepsy. Prog Brain Res. 2014;213:17-53.
of phenobarbital prophylaxis. J Child Neurol. 2007;22:389–395. 160. Reif PS, Tsai MH, Helbig I, et  al. Precision medicine in
138. Natarajan N, Beatty CW, Gust J, et al. Provider Practices of Phe- genetic epilepsies: break of dawn? Expert Rev Neurother.
nobarbital Discontinuation in Neonatal Seizures. J Child Neurol. 2017;17:381–392.
2018;33:153–157. 161. Pisano T, Numis AL, Heavin SB, et al. Early and effective treat-
139. Fitzgerald MP, Kessler SK, Abend NS. Early discontinuation ment of KCNQ2 encephalopathy. Epilepsia. 2015;56:685–691.
of antiseizure medications in neonates with hypoxic–ischemic 162. Scheffer IE, Nabbout R. SCN1A-related phenotypes: Epilepsy
encephalopathy. Epilepsia. 2017;58:1047–1053. and beyond. Epilepsia. 2019;60:S17–S24.
140. Glass HC, Soul JS, Chan T, et al. Duration of antiseizure medi- 163. Sadleir LG, Mountier EI, Gill D, et al. Not all SCN1A epi-
cation treatment and 2-year outcome after acute symptomatic leptic encephalopathies are Dravet syndrome. Neurology.
neonatal seizures - a Neonatal Seizure Registry study. 16th Int 2017;89:1035–1042.
Child Neurol Virtual Congr. 2020. 164. Berecki G, Bryson A, Terhag J, et al. SCN1A gain of function in
141. Bartha AI, Shen J, Katz KH, et al. Neonatal Seizures: Multi- early infantile encephalopathy. Ann Neurol. 2019;85:514–525.
center Variability in Current Treatment Practices. Pediatr Neurol. 165. Reynolds C, King MD, Gorman KM. The phenotypic spec-
2007;37:85–90. trum of SCN2A-related epilepsy. Eur J Paediatr Neurol.
142. Wietstock SO, Bonifacio SL, McCulloch CE, et al. Neonatal neu- 2020;24:117–122.
rocritical care service is associated with decreased administration 166. Wolff M, Johannesen KM, Hedrich UBS, et al. Genetic and
of seizure medication. J Child Neurol. 2015;30:1135–1141. phenotypic heterogeneity suggest therapeutic implications in
143. Cornet MC, Sands TT, Cilio MR. Neonatal epilepsies: Clinical SCN2A-related disorders. Brain. 2017;140:1316–1336.
management. Semin Fetal Neonatal Med. 2018;23:204–212. 167. McTague A, Appleton R, Avula S, et al. Migrating partial sei-
144. Kaur S, Pappas K. Genetic Etiologies of Neonatal Seizures. Neo- zures of infancy: Expansion of the electroclinical, radiological
reviews. 2020;21:e663-e672. and pathological disease spectrum. Brain. 2013;136:1578–1591.
145. Moavero R, Kotulska K, Lagae L, et al. Is autism driven by epi- 168. Milligan CJ, Li M, Gazina EV, et al. KCNT1 gain of function
lepsy in infants with Tuberous Sclerosis Complex? Ann Clin in 2 epilepsy phenotypes is reversed by quinidine. Ann Neurol.
Transl Neurol. 2020;7:1371–1381. 2014;75:581-590.
146. Tye C, Mcewen FS, Liang H, et al. Long-term cognitive out- 169. Mikati MA, Jiang YH, Carboni M, et al. Quinidine in the treatment
comes in tuberous sclerosis complex. Dev Med Child Neurol. of KCNT1-positive epilepsies. Ann Neurol. 2015;78:995–999.
2020;62:322–329. 170. Bearden D, Strong A, Ehnot J, et al. Targeted treatment of migrat-
147. Parrini E, Conti V, Dobyns WB, et al. Genetic basis of brain ing partial seizures of infancy with quinidine. Ann Neurol.
malformations. Mol Syndromol. 2016;7:220–233. 2014;76:457–461.
148. Saffari A, Brösse I, Wiemer-Kruel A, et al. Safety and efficacy 171. Borlot F, Abushama A, Morrison‐Levy N, et al. KCNT1‐related
of mTOR inhibitor treatment in patients with tuberous sclerosis epilepsy: An international multicenter cohort of 27 pediatric
complex under 2 years of age - A multicenter retrospective study. cases. Epilepsia. 2020;61:679–692.
Orphanet J Rare Dis. 2019;14:1–13. 172. Fitzgerald MP, Fiannacca M, Smith DM, et  al. Treatment
149. Wilmshurst JM, Gaillard WD, Vinayan KP, et al. Summary of Responsiveness in KCNT1-Related Epilepsy. Neurotherapeutics.
recommendations for the management of infantile seizures: Task 2019;16:848–857.
Force Report for the ILAE Commission of Pediatrics. Epilepsia. 173. Rizzo F, Ambrosino P, Guacci A, et al. Characterization of two
2015;56:1185–1197. de novo KCNT1 mutations in children with malignant migrating
150. Pearl PL. Amenable Treatable Severe Pediatric Epilepsies. Semin partial seizures in infancy. Mol Cell Neurosci. 2016;72:54–63.
Pediatr Neurol. 2016;23:158–166. 174. Pressler RM, Lagae L. Why we urgently need improved seizure
151. Schwahn BC, Van Spronsen FJ, Belaidi AA, et al. Efficacy and and epilepsy therapies for children and neonates. Neuropharma-
safety of cyclic pyranopterin monophosphate substitution in cology. 2020;170:107854.
severe molybdenum cofactor deficiency type A: A prospective 175. Grinspan ZM, Patel AD, Shellhaas RA, et al. Design and imple-
cohort study. Lancet. 2015;386:1955–1963. mentation of electronic health record common data elements for
152. Reiss J. Molybdenum cofactor deficiency type B knock-in mouse pediatric epilepsy: Foundations for a learning health care system.
models carrying patient-identical mutations and their rescue by Epilepsia. 2021;62:198-216.
singular AAV injections. Hum Genet. 2019;138:355–361. 176. Wusthoff CJ, Sullivan J, Glass HC, et al. Interrater agreement in
153. Pena IA, MacKenzie A, Van Karnebeek CDM. Current knowl- the interpretation of neonatal electroencephalography in hypoxic-
edge for pyridoxine-dependent epilepsy: a 2016 update. Expert ischemic encephalopathy. Epilepsia. 2017;58:429–435.
Rev Endocrinol Metab. 2017;12:5–20.
154. Pérez B, Gutiérrez-Solana LG, Verdú A, et al. Clinical, biochem- Publisher’s Note Springer Nature remains neutral with regard to
ical, and molecular studies in pyridoxine-dependent epilepsy. jurisdictional claims in published maps and institutional affiliations.
Antisense therapy as possible new therapeutic option. Epilepsia.
2013;54:239–248.

13

You might also like