You are on page 1of 13

18  Neonatal Seizures

Renée A. Shellhaas, Hannah C. Glass, and Taeun Chang

INTRODUCTION co-transporter becomes dominant, which leads neurons to


display the mature pattern of low intracellular chloride and
Children are at highest risk for seizures during the first hyperpolarization with activation of the GABAA receptor.10–12
month of life.1 Seizures in newborns are usually symptomatic Thus GABAergic medications (such as benzodiazapines and
of an underlying acute injury (such as hypoxic-ischemic injury barbiturates), which are commonly used to treat seizures in
or stroke) and are only rarely manifestations of epilepsy syn- neonates, could, in theory, cause a paradoxical excitatory
dromes. Similar to other critically ill patient populations, response. Nonetheless, the response to phenobarbital as a
most neonatal seizures are subclinical—they have no outward first-line agent is approximately 50%,13 which is similar to the
manifestation and are most accurately diagnosed by electro- effect of first-line agents in the pediatric intensive care unit14
encephalography (EEG). Herein, we review the pathophys­ and suggests that GABA agents are inhibitory in some popula-
iology, epidemiology, diagnosis, treatment, and long-term tions of neurons in the neonatal period. Bumetanide, a potent
implications of seizures in neonates. diuretic that acts to alter the intracellular chloride concentra-
tion in favor of hyperpolarization, has been proposed as an
PATHOPHYSIOLOGY adjunct to improve the efficacy of phenobarbital, but safety
and efficacy data are not yet available.15
The neonatal period presents the highest lifetime risk for sei- Glutamate is the primary excitatory neurotransmitter.
zures.1 Acute symptomatic seizures caused by birth trauma The glutamate receptors are developmentally regulated, and
and hemorrhage, hypoxic-ischemic injury, perinatal infec- lead to enhanced excitability of the immature brain. N-methyl-
tions, and metabolic disturbances account for more than 80% d-aspartate (NMDA) receptors are relatively abundant in new-
of seizures. In addition, but much less commonly, congenital borns, and their subunits are configured with a high level
brain malformations, inborn errors of metabolism, and epi- of the NR2B subunit, which leads to greater excitability
leptic encephalopathies can present in the neonatal period through prolonged current delay and excitatory postsynaptic
with seizures as the first sign of neonatal onset epilepsy. potentials, as well as a relative insensitivity to magnesium
The immature brain is highly susceptible to acute symp- ions.16–18
tomatic seizures because of age-dependent mechanisms that Finally, excitability is enhanced during the neonatal period
lead to excess excitation and reduced inhibition.2,3 Results as a result of physiologic, use-dependent synaptogenesis,
from animal models suggest that acute symptomatic seizures when both synapse and dendritic spine density are at their
are deleterious to the developing brain.4 In humans, seizures peak.19,20
in the newborn period are associated with brain injury and
adverse neurodevelopment.5,6 In addition, among neonates
with hypoxic-ischemic encephalopathy (HIE), seizures are The Effect of Seizures on Early  
associated with higher lactate on magnetic resonance (MR) Brain Development
spectroscopy (indicating injury) and higher rates of adverse
Seizures during early development can lead to developmental
neurodevelopmental outcome that are independent of the
changes that alter neuronal circuitry and may impair learning
severity of brain injury. These data suggest that seizures may
and memory in animal models. Developmental alterations
also be harmful to human newborns with underlying brain
that have been observed in neonatal animal models after
injury.7,8
induced seizures include (1) reduced density of dendritic
spines in hippocampal pyramidal neurons, (2) decreased neu-
Mechanisms of Excitability in the   rogenesis, (3) delayed neuronal loss, and (4) changes in hip-
Developing Brain pocampal plasticity (e.g., decreased capacity for long-term
potentiation, reduced susceptibility to kindling, and enhanced
The developing brain’s propensity to generate seizures is mul- paired-pulse inhibition).21–24 Furthermore, in the setting of
tifactorial.2,3,9 The primary mechanisms of inhibition (through brain injury and hyperthermia, seizures can lead to hippocam-
gamma-amino-butyric acid [GABA]) and excitation (through pal necrosis in animal models.25 In addition, in rodent models,
the glutamatergic system) favor excitability during the neona- neonatal seizures increase the susceptibility to unprovoked
tal period. recurrent seizures (epilepsy) later in the animal’s life.4
GABA is the primary inhibitory mechanism of the adult
brain. In mature neurons, GABAA receptor activation leads to
chloride influx to produce membrane hyperpolarization and EPIDEMIOLOGY
inhibits the neuron’s ability to fire action potentials. In imma-
ture neurons, however, there is a net chloride efflux with
Incidence of Neonatal Seizures
GABAA receptor activation, which leads to membrane depolar- Current estimates of the incidence of seizures in newborns
ization that increases the likelihood of the neuron to fire an range from 1 to 3.5 per 1000 live births, or approximately
action potential. The developmental changes in neuronal 14,000 newborns annually in the United States.26–29 Estimates
chloride gradients are mediated largely by the membrane ion as high as 1 per 20 have been reported for preterm or very low-
transporters NKCC1 and KCC2. birth-weight newborns, with the incidence inversely related to
In early life, a relatively higher expression of NKCC1 birth weight and gestational age26,29–31 (Table 18-1). Males are
leads to a high intracellular chloride concentration, and sub- more often affected than females, and infants of African Amer-
sequent depolarization upon activation of the GABAA receptor. ican origin have a higher incidence of seizures than those of
With increasing age, the expression of the KCC2 chloride other races and ethnicities.27,32

e318

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e319

TABLE 18-1  Estimates of Neonatal Seizure Incidence


18
Overall NBW/
Author Year Type Source Method Incidence* Term <36 wk* <2500 g* VLBW*
Holden et al.34 1959–1966 Prospective National Collaborative — 5.0 — 29.5 31 —
Perinatal Project
Lanska and 1980–1991 Retrospective National Hospital Discharge 2.84 2.4 — 9.4 —
Lanska30 Discharge Survey records
Lanska et al.31 1985–1989 Retrospective Fayette County, Birth certificates 3.5 3.6 — 18.6 57.5
Kentucky Hospital records
Ronen et al.29 1990–1994 Prospective Newfoundland, Training 2.6 2.0 11.1 13.5 —
Canada
Saliba et al.32 1992–1994 Retrospective Harris County, Texas Birth and death 1.8 1.4 4.85 5.6 19
and certificates
prospective
Glass et al.27 1998–2002 Retrospective California Birth certificates 0.95 — — — —
Hospital records
NBW, normal birth weight; VLBW, very low birth weight.
*Per 1000 live births.

Although seizure incidence reported in more recent studies Seizures in the newborn usually occur in the first week of
is lower than that reported in older studies,27,32 differences in life (~70–85%), particularly in the first 2 days.29,32 In the
case ascertainment or study site(s) may be responsible for this National Collaborative Perinatal Project of 1959 through
variation. The true incidence of seizures in the newborn is 1966,34 23% of seizures were diagnosed in the first 12 hours
difficult to determine as population studies have been based of life, 42% in the first 24 hours, and 65% in the first 2 days.
on clinically observed or reported seizure-like activity, without In a more recent study in Newfoundland, Canada,29 36% of
EEG confirmation. More recent data suggest that such case affected infants had seizure onset in the first 24 hours and
ascertainment may include infants whose events were not, in 64% within the first 48 hours. Similar findings were noted in
fact, seizures, while excluding those with exclusively subclini- the more diverse population of Harris County, Texas.32
cal seizures. Most studies use a case definition of seizures in Seizure duration in newborns has not commonly been
infants up to 44 weeks’ postmenstrual age (PMA). examined in these population-based studies as seizures were
Published epidemiologic studies have mainly been retro- defined clinically and often retrospectively. Ronen et al.29 did
spective and based on clinical observation of neonatal sei- capture seizure duration in their questionnaires to the witness-
zures, as documented in birth certificates, death certificates, ing provider. Similar to EEG-based case cohort studies of new-
medical records, discharge records, and/or diagnostic codes. borns with seizures, 85% of seizures lasted less than 5 minutes.
Glass et al.27 included discharge codes from birth admissions Although rare, 5% of newborns were clinically observed to
only. Newborns diagnosed with neonatal seizures after trans- have seizures lasting more than 30 minutes. Emergence of
fer from birth hospital to a tertiary hospital or after discharge continuous prolonged video-EEG recordings with the conver-
would not have been captured. Saliba et al.32 observed that sion from pen-and-paper to digital EEG technology has
infants treated in more tertiary Neonatal Intensive Care Units improved our ability to capture and characterize neonatal sei-
(NICUs) were at higher risk of seizures, reflecting a higher risk zures. Seizures and status epilepticus may be more common
population. A prospective study at a single tertiary children’s than previously realized,35–37 but most neonatal seizures
hospital serving a rural region observed a seizure incidence of remain brief, with 60% lasting less than 90 seconds.38
8.6% of NICU admissions.33
Seizures are up to 10 times more common in preterm than in
term infants with incidence inversely related to birth weight30,31
Risk Factors for Neonatal Seizures
and gestational age.29 The risk for seizures in very low-birth- Risk factors for neonatal seizures are related to their long list
weight (<1500 g) infants is 57.5 per 1000 live births, 4.4 in of underlying etiologies (Table 18-2). In addition to low birth
1500- to 2499-g infants, 2.8 in 2500- to 3999-g infants, and 2.0 weight and gestational age, risks for seizures in a newborn
in infants greater than 4000 g.31 No gender or racial variation include preexisting maternal diabetes, maternal fever or infec-
was found.30,31 These reported rates are higher than in a study by tion, perinatal or postnatal infection, major morbidities
two of the same authors in a nationwide survey of short-stay or surgical interventions in the infant, evidence of fetal dis-
hospitals examining neonatal seizures in infants with less than tress, and postterm delivery.27,28,39,40 Possible risk factors also
30 days of hospitalization. Diagnoses made after transfer or include advanced maternal age, maternal nulliparity, and male
infants discharged beyond 30 days were not captured. gender.27,39
Sheth et al.33 observed a parabolic relationship between
seizure incidence and gestational age, with 30 to 36 weeks’
gestational age infants at lowest risk. This observation may be
ETIOLOGY
related to the two most common causes of seizures, intraven- The causes of seizures in newborns are numerous and diverse
tricular hemorrhage and hypoxic ischemic injury, being more (see Table 18-2); a single newborn can have more than one
common in the younger and older age groups, respectively. Of cause of seizures. Most neonatal seizure etiologies reflect
note, this study involved a single tertiary center with a clinical underlying acute brain injury rather than epilepsy. Our ability
neonatal seizure case definition of only infants that required to determine etiology has improved with enhanced access to
anticonvulsant medications for their seizures. EEG monitoring and brain magnetic resonance imaging

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e320 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

TABLE 18-2  Distribution of Neonatal Seizure Etiologies


Mastrangelo
Loman et al.56 Pisani et al.41 Tekgul et al.43 et al.44 Ronen et al.29
(2002–2009), (1999–2004), (1997–2000), (1990–1998), (1990–1994),
N = 221 N = 106 N = 89 N = 94 N = 89
Hypoxic-ischemic encephalopathy, % 57.5 43.4 40 44.7 40
Metabolic or electrolyte disturbances, % 10.9 6.6 3 3.2 19
Intracranial hemorrhage, % 9.0 23.6 17 4.3 11
Cerebrovascular disorders, % 7.7 — 18 7.4 7
Infections, % 6.3 7.5 3 10.6 20
Congenital central nervous system 3.2 5.7 5 9.6 10
abnormalities, %
Inborn errors of metabolism, % 2.3 6.6 1 7.4 —
Epilepsy syndromes, % 2.3 — — 5.3 6
Intoxications, % 0.5 — — — —
Unknown, % 0.5 6.6 12 1.1 14

(MRI), as well as advances in genetic testing. Determination neonates and medullary vein thrombosis in preterm neonates.
of seizure etiology is critical, as treatment of the underlying The incidence is rare, between 1 and 2.69 per 100,000 new-
cause may be life-saving (e.g., antibiotics for a newborn with borns.54 Venous infarction occurs in approximately 60% of
bacterial meningitis), and combining seizure burden with eti- reported cases. Two thirds of affected neonates present during
ology41,42 can assist in assessing prognosis. the first week of life and with seizures.55 Maternal, fetal, and
Etiologies of neonatal seizures can be divided into three neonatal risk factors for CSVT are similar to those outlined
broad categories: (1) acute symptomatic seizures (by far the earlier for arterial strokes, and many infants have a combina-
most common); (2) developmental brain abnormalities; and tion of predisposing factors that include neonatal sepsis and
(3) genetic or neonatal epilepsy syndromes. The following dehydration.
sections highlight the more common causes. Intracranial hemorrhages can result in neonatal seizures.
Diagnosis typically requires neuroimaging, and head ultraso-
nography and brain MRI are the preferred modalities. Com-
Acute Symptomatic Seizures puted tomography is not preferred for newborns, given the
Hypoxic ischemic encephalopathy (HIE) is the most frequent risk of radiation and lack of benefit relative to ultrasonography
cause and accounts for up to 40% to 45% of neonatal sei- for identification of conditions for which neurosurgical inter-
zures.41,43,44 Therapeutic hypothermia is now standard care for ventions are required. Hemorrhages in term neonates may be
HIE in newborns 36 weeks’ gestation or greater, and EEG caused by trauma, coagulopathy, or vascular malformations.
monitoring is recommended for cooled newborns. Prolonged Subarachnoid hemorrhage (SAH) and subdural hemorrhage
continuous video EEG throughout cooling and rewarming is (SDH) are common in healthy term infants, related to labor
preferred. If continuous EEG is not available, then continuous and delivery, and are usually not significant (i.e., they are not
amplitude-integrated EEG (aEEG), or daily serial routine EEGs necessarily the primary etiology for a newborn who develops
for the first 4 days of life are suggested. Even though cooling seizures) (Chapter 21). Intraventricular hemorrhage (IVH) is
may decrease seizure burden, about half of treated newborns common in preterm infants less than 30 weeks’ gestation but
still have seizures during therapeutic hypothermia.45–49 Seizure on occasion can present in term infants (Chapter 22) usually,
onset continues to be within the first 24 hours of life.43,45,48 in this case, because of sinovenous thrombosis. IVH is the
See Chapter 19 for more details. primary seizure etiology in preterm infants.33 Seizures are
Cerebrovascular events, including arterial and venous infarc- associated with IVH grade 3 or periventricular hemorrhagic
tions and intracranial hemorrhages, are the second most common infarct (IVH grade 4) and typically occur in the first 3 days
cause of neonatal seizures (7%–18%) (Chapter 20). Recogni- of life.
tion and characterization of cerebrovascular events in new- Systemic or central nervous system (CNS) infections are
borns has improved with increased access to brain MRI. the third most common cause of seizures in newborns
Arterial ischemic perinatal strokes (AIS) occur in 1 in 2300 to (3–10%).41,43,44,56 This category includes in utero or post­
5000 live births50 and are typically the result of embolism natal infections, meningitis, and meningoencephalitis. Viral
from the placenta or umbilical cord, carotid artery, or heart. etiologies include herpes simplex virus (HSV), cytomegalovi-
Seizures are the most frequent presenting symptom of AIS.51 rus (CMV), parechovirus, lymphocytic choriomeningitis virus
Almost two thirds involve the left middle cerebral artery terri- (LCMV), disseminated enterovirus, and parvovirus (Chapter
tory. Maternal risk factors for perinatal arterial ischemic strokes 115). Bacterial sources include group B streptococcus (early
include oligohydramnios, clinical or histologic chorioamnio- and late), Escherichia coli, and toxoplasmosis (Chapter 114).
nitis, premature rupture of membranes, preeclampsia, diabe- Lumbar puncture is recommended in all neonates with sus-
tes, and smoking. Risk factors in the neonate include congenital pected infection. Infants too unstable to undergo lumbar
cardiac abnormalities, systemic infections, coagulation disor- puncture should be treated empirically for CNS infection.
ders in the neonate, placental abnormalities, and male Seizures caused by primary metabolic disturbances, or meta-
gender.51–53 bolic abnormalities associated with acute medical illness,
Cerebral sinovenous thrombosis (CSVT) usually involves account for 3% to 7% of EEG-confirmed seizures.41,43,44 Such
the superior sagittal sinus and/or transverse sinus in term disturbances most often include abnormal glucose, calcium,

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e321

or sodium levels. Typically, reversal of these abnormalities encephalopathy and is responsive to oral pyridoxal phosphate
results in resolution of the neonatal seizures. and not to pyridoxine. 18
Hypoglycemia occurs in 1 to 3 out of 1000 live births. The Disorders of energy metabolism include glucose transporter
definition is based on gestational age and day of life. In deficiency (Chapter 76), pyruvate dehydrogenase deficiency
general, hypoglycemia is defined as less than 45 mg/dL in (PDH), pyruvate carboxylase deficiency (PCD), biotinidase
term infants and less than 30 mg/dL in preterm infants. Risk deficiency, respiratory chain disorders (Leigh disease and
factors include maternal diabetes, small for gestational age, in Alpers disease), Menkes disease, fumarase deficiency, sulfite
conjunction with illness, feeding issues, and/or pancreatic oxidase deficiency, purine disorders, and creatine synthesis
insulinoma. Overall the incidence has declined with improved and transporter defects. Glucose transporter type 1 deficiency
neonatal care. Initial treatment is to provide supplemental (GLUT-1) results in decreased transport of glucose at the
glucose. Antiseizure drug administration is necessary only if blood–brain barrier and across glial cell membranes causing
seizures persist despite glucose boluses. Severe hypoglycemia hypoglycorrhachia (CSF glucose less than 40 mg/dL). Seizure
may result in brain parenchymal injury, which is most often onset is usually between 6 and 12 weeks of life and is associ-
localized to the posterior brain quadrants.57 ated with developmental delay. Treatment is with the keto-
Hypocalcemia is the cause of 1% of all neonatal seizures.43 genic diet. Molybdenum cofactor deficiency and isolated sulfite
It is defined based on free ionized calcium levels rather than oxidase deficiency are rare but can mimic HIE in clinical presen-
total serum calcium. Early onset hypocalcemia, within 3 days tation. Affected infants have cystic white matter changes seen
of life, is associated with low-birth-weight infants, maternal on neuroimaging.
diabetes, HIE, and endocrinopathies. Late onset, after 1 week Biosynthetic defects causing brain malformation and cerebral
of life, is now rare but is associated with maternal hyperpara- dysfunction include peroxisomal disorders (Chapter 43),
thyroidism or maternal vitamin D deficiency, or may be a congenital disorders of glycosylation (CDG) (Chapter 40),
manifestation of DiGeorge syndrome. Hypocalcemia caused glycolipid synthesis (GM3 synthase deficiency), cholesterol
by high-phosphorus–containing formulas or use of cow’s milk synthesis (Smith–Lemli–Opitz syndrome) (Chapter 39),
is uncommon in the United States. serine and glutamine deficiency syndromes (rare), methyla-
Hyponatremia or hypernatremia is seen most commonly tion disorders (homocysteine and folate disorders), neuronal
in extremely premature infants. Among term infants, sodium ceroid lipofuscinosis, and lysosomal disorders (Chapter 41).
imbalance can be caused by incorrect mixing, because of Peroxisomal biogenesis disorders include Zellweger syndrome,
incorrect mixing of formula or severe dehydration, or can neonatal adrenoleukodystrophy, and infantile Refsum disease.
be a complication of intracranial injury. Severe dehydration Presentation includes severe encephalopathy, refractory neo-
with hypernatremia and seizures may be associated with natal seizures, and subtle dysmorphic features. The diagnosis
CSVT. is made by measurement of very long-chain fatty acids and
Inborn errors of metabolism (IEM) cause 1% to 7% of neo- phytanic acid.
natal seizures.41,43,44 Prompt diagnosis is necessary to correct
the underlying metabolic disorder and minimize irreversible
brain injury. The presence of seizures is related to the degree
Developmental Brain Abnormalities
of metabolic derangement and resultant brain injury. Seizures Congenital CNS abnormalities are relatively common causes
can be associated with urea cycle defects (hyperammonemia), of neonatal seizures (5–10%). Diagnosis requires neuroimag-
organic acidurias, and aminoacidopathies (metabolic acido- ing, with MRI the preferred imaging modality. Recognition
sis). These are discussed further in Chapter 23. and precise classification have increased with brain MR
Metabolic epileptic encephalopathies are uncommon, but imaging. Affected newborns may have comorbid HIE as they
sometimes treatable causes of seizures in newborns. These may not tolerate labor and delivery. Common developmental
disorders can be divided into three broad categories: (1) dis- brain abnormalities include focal cortical dysplasia, hemi-
orders of neurotransmitter metabolism; (2) disorders of megalencephaly, lissencephaly, heterotopias, schizencephaly,
energy metabolism; and (3) biosynthetic defects causing brain and polymicrogyria (see Chapters 24–32 for details). Neona-
malformation, dysfunction, and degeneration.58 tal seizures in the context of developmental brain abnormali-
Disorders of neurotransmitter metabolism (Chapter 44) ties are associated with intractable seizures, chronic epilepsy,
include nonketotic hyperglycinemia, pyridoxal-5′-phosphate– and poor neurodevelopmental outcome. Some neurocutane-
responsive encephalopathy (Chapter 76), pyridoxine- ous disorders, in particular tuberous sclerosis complex, can
dependent epilepsy (PDE) (Chapter 76), folinic acid–responsive present with seizures in the newborn period.
seizures, mitochondrial glutamate transporter, aromatic
amino acid decarboxylase deficiency (AADC), D-2-
hydroxyglutaric aciduria, and GABA disorders (Chapter 76).
Epilepsy Syndromes
Nonketotic hyperglycinemia (NKH) is a defect in the glycine Recognition of monogenic causes of neonatal or early infan-
cleavage system. Newborns often present with apnea, hypoto- tile epilepsy has increased dramatically with recent advances
nia, encephalopathy, and refractory seizures. Seizures are in genetic testing. Identification of a specific genetic etiology
believed to be the result of overactivation of NMDA excitatory provides insight into the pathophysiology of seizures, may
amino acid receptors. Diagnosis is by detecting an elevated direct treatment options, and can allow for specific discussions
cerebrospinal fluid (CSF)–to–serum glycine ratio. regarding prognosis. Neonatal epilepsy syndromes can range
PDE is caused by a rare autosomal recessive antiquitin from benign (seizures expected to resolve quickly and long-
deficiency (ALDH7A1 gene). The seizures rapidly respond to term neurodevelopment likely to be normal) to severe early
intravenous (IV) pyridoxine, and so a pyridoxine administra- onset epileptic encephalopathies (seizures expected to be
tion trial is warranted for newborns with unexplained treatment-resistant and long-term neurodevelopment likely to
treatment-resistant seizures (see Treatment section later). be abnormal).
Folinic acid–dependent seizures are an allelic phenotype. Severe epilepsy syndromes should be suspected for neo-
Diagnosis is by examining alpha-aminoadipic semialdehyde nates with no obvious acute symptomatic cause of seizures,
levels in urine, and by observation of seizure remittance with especially if the EEG background shows burst suppression.
pyridoxine treatment. Pyridoxine-5′-phosphate deficiency is Initial diagnostic testing should include brain MRI, often with
related to PDE, but often presents with severe epileptic MR spectroscopy.58 MRI findings may help narrow the

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e322 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

differential diagnosis and tailor the subsequent workup. An relationship, or no relationship at all, to seizures on EEG.
IV pyridoxine challenge (as outlined in the Treatment section) Motor automatisms (abnormal mouth or eye movements)
should be performed while serum, urine, CSF, and genetic and myoclonus were often found to lack clear EEG correlate.
studies are collected and processed. These data highlight the difficulty in relying on clinical obser-
Familial epilepsies include benign familial neonatal epi- vation for the diagnosis of epileptic neonatal seizures.
lepsy (BFNE) and benign idiopathic neonatal seizures (BINS, Although it is possible that the events that lacked EEG corre-
previously known as fifth-day fits) (Chapters 64 or 70). BFNE lates truly were epileptic seizures, but with foci too deep to
is an autosomal dominant epilepsy syndrome with 85% pen- record on scalp EEG, it is more likely that many of the abnor-
etrance that is related to KCNQ2 or KCNQ3 voltage-gated mal events diagnosed clinically as seizures were, in fact, not
potassium channel mutations. Repolarization of the cell epileptic seizures.
membrane is impaired, and this leads to neuronal hyperexcit- Additional studies have clearly demonstrated that bedside
ability. Seizure onset is usually in the first week of life and the clinical observation is inaccurate for neonatal seizure detec-
semiology is commonly focal tonic seizures. Seizures resolve tion. In one study, bedside clinicians (nurses and physicians)
spontaneously in early infancy. Infants have a normal neuro- were trained to record any events that were suspicious for
logic examination, normal interictal EEG, and a family history seizures for a sample of high-risk neonates who were undergo-
of neonatal seizures. BINS usually occur between 4 and 6 days ing conventional EEG recording. Only 9% (48 of 526) of all
of life and resolve within 2 weeks, and the infant has no family EEG-confirmed seizures had clinical signs that were noted in
history of neonatal seizures. Seizures can be focal clonic, the bedside logs, whereas 78% (129 of 177) of the abnormal
apneic, and sometimes status epilepticus, but the evaluation paroxysmal events documented by NICU staff had no EEG
for etiology is unrevealing. Some affected individuals have correlate (i.e., the events were not seizures).63 In another study,
been reported to have KCNQ2 mutations. 137 physicians and nurses viewed video recordings of electro-
A more severe phenotype is also associated with KCNQ2 clinical seizures (EEG-confirmed seizures that had clinical
mutations. KCNQ2 encephalopathy presents with frequent signs) and nonseizure events (events that had no correspond-
pharmacoresistant seizures in the first week of life. Seizures ing EEG change).64 Interobserver agreement was poor (multi-
often have a tonic semiology. The interictal EEG is markedly rater kappa 0.21–0.29) and although 66% of clonic seizures
abnormal and often meets criteria for burst suppression. Brain were correctly diagnosed, only 32% of seizures with other
MRI reveals subtle T1 and T2 hyperintensities in the basal semiologies were accurately identified. Conversely, only 29%
ganglia and thalami—findings that may resolve after the neo- to 55% of nonseizure events (such as nonseizure clonus,
natal period. Affected children typically have severe global benign sleep myoclonus, or other nonspecific movements)
neurodevelopmental disabilities.59 were classified correctly.
There are two classic neonatal epileptic encephalopathy Imprecise seizure diagnosis has significant consequences:
syndromes.60 Early myoclonic encephalopathy (EME) has neona- newborns with primarily subclinical seizures may be under-
tal onset with erratic focal myoclonic jerks, often accompanied treated, whereas many infants whose paroxysmal events are,
by focal seizures and occasionally by tonic seizures. The typical in fact, not because of seizures, may be exposed unnecessarily
interictal EEG shows a burst–suppression pattern during to potentially detrimental anticonvulsant medications.
sleep. Later, the EEG evolves into atypical hypsarrhythmia or Although some neonatal seizures have associated clinical
multifocal sharp waves or spikes. EME is typically caused by signs, multiple studies of critically ill newborns have high-
metabolic disorders, such as nonketotic hyperglycinemia, lighted the fact that most neonatal seizures are subclini-
pyridoxine dependency, and propionic aciduria; molybdenum cal.48,65–67 Subclinical seizures have no outward manifestation,
cofactor deficiency; sulfite oxidase deficiency; Menkes disease; and are therefore only detectable with EEG. That most neona-
and Zellweger syndrome. Ohtahara syndrome presents with fre- tal seizures are subclinical should not be surprising. Preverbal
quent tonic spasms in newborns or young infants. It is associ- children cannot communicate their experiences of sensory
ated with a burst–suppression EEG pattern during both the phenomena caused by seizures (e.g., visual changes associated
awake and sleep states, along with severe encephalopathy and with occipital seizures, or déjà vu because of temporal lobe
treatment-resistant epilepsy. Causes typically include struc- seizures), and unless the seizure originates from, or propa-
tural brain abnormalities (cerebral dysgenesis, lissencephaly, gates to, the motor cortex, there will be no abnormal move-
porencephaly, hemimegalencephaly, and Aicardi syndrome) ments. Isolated paroxysmal changes in blood pressure or heart
and monogenic mutations (e.g., ARX, SLC25A22, PLCβ1, and rate, without other associated clinical signs, often raise concern
PNKP). for seizures in high-risk newborns, but these events are rarely
because of seizures.66,68 Even neonates who present with clini-
cally apparent seizure semiologies often experience electro-
DIAGNOSIS clinical dissociation when medication is administered (e.g.,
Traditionally, neonatal seizures were diagnosed by clinical EEG seizures persist even after resolution of the clinical
observation, with the semiology categorized according to a signs).67
system developed by Volpe.61 This schema classifies seizures Because clinical signs of neonatal seizures are uncommon
according to their motor manifestations—focal clonic, multi- and difficult to identify, EEG monitoring is required for precise
focal clonic, generalized tonic, myoclonic, and subtle. The diagnosis of paroxysmal events and for quantification of
“subtle” semiology refers to seizures with signs such as abnor- seizure burden. Neonatal seizures are most properly defined
mal eye movements, lip smacking, swimming or pedaling by their EEG patterns and they need not have a clinical semiol-
movements, or apnea. ogy. A seizure is an abnormal EEG pattern that evolves, is of
Although classic research relied on recognition of clinical greater than 2-µV amplitude, and has a duration of 10 seconds
signs for the diagnosis of neonatal seizures, modern EEG or greater (Figure 18-1).69 Distinct seizures are separated by a
data have demonstrated that not all clinically suspicious 10-second or greater seizure-free interval. It is very uncommon
events are epileptic seizures—indeed, most are not. For for individual neonatal seizures to be more than a few minutes
example, in one seminal study of 349 neonates, clinical signs in duration, but the seizures are often very frequent. Therefore
seen on video were correlated with simultaneously recorded neonatal status epilepticus is defined as seizures that occupy
EEG.62 Of 415 clinical events characterized as seizures on the more than 50% of any 1-hour EEG epoch (e.g., a newborn
basis of the video recording, 296 (71%) had an inconsistent with 30 1-minute seizures in an hour would meet criteria for

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e323

18

Figure 18-1.  This electroencephalography was recorded from a 3-day-old male, born at 38 weeks’ gestation, who presented with apnea
associated with focal tonic stiffening. The arrow indicates an electrographic seizure, which arose from the right temporal region. The star
indicates the respiratory channel, which demonstrated apnea associated with the seizure.

status epilepticus, as would an infant with a single seizure that


lasts 30 minutes or greater).69

Neonatal Electroencephalogram Monitoring Fp1 Fp2


Conventional Video Electroencephalogram
Video EEG monitoring remains the gold standard for neonatal
seizure detection and is indispensible for research studies that Fz
attempt to quantify seizure burden and/or treatment responses.
Standard electrode positions, using the international 10–20
system, modified for neonates are recommended70 (Figure
18-2). In addition to the scalp electrodes, extracerebral leads T3 C3 Cz C4 T4
for respiratory and electrocardiogram recording are required
for accurate evaluation of behavioral state and exclusion of
extracerebral artifacts. Time-locked video monitoring is highly
recommended during the EEG recording to assist in the dif- Pz
ferential diagnosis of abnormal paroxysmal events. Video is
critical to distinguish sources of artifact, such as handling or
patting the infant, or electronic interference from intensive care O1 O2
unit (ICU) equipment. Surface electromyography and extra-
oculogramy leads are often utilized, but these are not manda-
tory. A bedside observer (typically the bedside nurse) is also
important, as this person can press a patient event marker, or Figure 18-2.  The international 10–20 system for electroencepha-
enter information directly onto the digital file through a lography electrode placement, modified for neonates, includes
bedside computer, to alert the neurophysiologist to clinically the electrode positions indicated by the boxes. By convention, the
important events, such as the occurrence of target paroxysmal odd and even numbers designate the left and right sides of the head,
events or administration of neuroactive medications. The respectively, and “z” labels the midline positions. Additional extracere-
minimum standards for neonatal EEG recording have been bral channels, including respiratory and electrocardiogram tracings,
defined by the American Clinical Neurophysiology Society.71 are also necessary to exclude artifact and determine behavioral state.
Extraocular and surface electromyography leads are often useful, but
not always required.
Indications for Electroencephalogram Monitoring
Not every sick newborn requires EEG monitoring. Rather, this Society also provides guidelines on selection of newborns for
resource-intensive monitoring should be targeted at those at EEG monitoring.70
highest risk for seizures. In general, consideration for EEG
monitoring is recommended for neonates with paroxysmal
clinical events that are suspicious for seizures, as well as those
Duration of Electroencephalogram Recording
with proven or suspected acute brain injury and clinical A routine-length, 60-minute, neonatal EEG is inadequate to
encephalopathy. The American Clinical Neurophysiology screen for neonatal seizures.70 High-risk newborns should

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e324 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

ideally be monitored with at least 24 hours of conventional Because conventional EEG monitoring is resource-intensive
EEG if seizures are suspected, because nearly all neonates with and requires specialized equipment, available technologists,
seizures will be identified within 24 hours of monitoring.66,72,73 and trained electroencephalographers, a reduced-montage
If seizures are identified, experts suggest that they be moni- EEG has gained popularity in many Neonatal Intensive Care
tored with EEG until the infant is seizure-free electrographi- Units (NICUs). aEEG is a simplified trend monitor that dis-
cally for at least 24 hours, although there are no published plays one or two channels of time-compressed, processed EEG
data to support this recommendation.70 In some circum- signal on a semilogarithmic scale.76,77 Interpretation of the
stances this duration of monitoring is not feasible, or is not aEEG signal is enhanced by concurrent display of the raw
in the infant’s best interests (e.g., an infant with a severe neo- single or multichannel EEG. aEEG background patterns have
natal epilepsy syndrome might not be expected to remain predictive value for neonates with HIE and other causes of
seizure-free, or monitoring might be appropriately suspended encephalopathy.74,78,79 The use of aEEG for seizure detection is
to allow for neuroimaging). controversial, as this tool is specific but not sensitive in this
A 60-minute routine-length neonatal EEG is typically suf- capacity. There is substantial variability in the reported sensi-
ficient to assess the EEG background, which might be of inter- tivity of aEEG for seizure detection (usually 25–35%, but
est as a marker of prognosis or as an adjunctive tool for occasionally reported 85% sensitivity for individual seizure
estimating gestational age. The suggested 60-minute duration detection).80
is longer than routine-length EEGs for older infants or chil-
dren, because it is imperative that all behavioral states be
recorded. It is not unusual for a newborn to have a relatively Diagnostic Considerations for Neonates  
normal awake EEG background but have clinically significant
abnormalities detected during quiet sleep.
with Seizures
If EEG monitoring is initiated in order to clarify whether a If the diagnosis of neonatal seizures is being entertained, a
newborn’s paroxysmal clinical events are caused by seizures, complete evaluation for the etiology is warranted (Figures
then the EEG should be recorded until several typical episodes 18-3 and 18-4). The great majority of neonatal seizures are
are captured. If the EEG background is stable and the target acute symptomatic manifestations of brain injury and many
events are not seizures, then monitoring for this purpose can require urgent, specific treatment. This diagnostic evaluation
be terminated. should occur in tandem with the treatment of seizures. Glucose
As discussed under the Acute Symptomatic Seizures sub- and electrolyte levels should be measured and any abnormal-
heading, HIE is the most common etiology of neonatal sei- ity corrected emergently. A full sepsis evaluation, including
zures. These newborns are now treated with therapeutic cultures of blood, urine, and CSF, is strongly recommended
hypothermia for neuroprotection. Because at least half of (unless the infant is too unstable to undergo a lumbar
neonates treated with therapeutic hypothermia for HIE have puncture or a definite alternative diagnosis is made). Empiric
EEG-confirmed seizures,45,49 many neonatal neurointensive antibiotics are usually administered until the cultures are
care programs recommend that EEG monitoring continue negative.
throughout cooling and rewarming. EEG background patterns In most hospitals and emergency departments, it is straight-
and their prognostic significance are altered by therapeutic forward to obtain a head ultrasound. Despite its low resolu-
hypothermia. Sleep–wake cycling, for example, emerges later tion, cranial ultrasonography can be used to screen for obvious
in infants who are cooled than those who do not receive hydrocephalus or intracranial hemorrhage. Computed tomog-
therapeutic hypothermia, and not all neonates with a sup- raphy is usually avoided in neonates, because of the concern
pressed EEG background in the first day of life will have a poor about radiation exposure. Rather, MRI is the standard neuro-
prognosis, especially in the setting of sedative anticonvulsant imaging modality for neonates with seizures. If arterial ische-
medication.74,75 Despite therapeutic hypothermia, severe inter- mic stroke or vascular malformations are suspected, an MR
ictal EEG abnormalities, especially burst suppression, remain angiogram is recommended. If a venous sinus thrombosis is
ominous if they persist beyond 24 to 30 hours of life.73 suspected (e.g., in a term neonate with intraventricular or

Suspected clinical seizures

High risk*

Begin evaluation for acute


Initiate or review EEG symptomatic etiology.
Treat as soon as possible.**

EEG-confirmed seizures No EEG seizures

Record 3–4 typical events or


24 hours without seizures

• Rapid medication titration*** until EEG Discontinue EEG and seizure medication
seizures controlled  24 hours
• Continued evaluation for etiology

* High-risk scenarios: known or suspected acute brain injury, encephalopathy, previous EEG seizures, first 72 hours of life
** Reverse glucose or electrolyte abnormalities, and/or administer lorazepam 0.1 mg/kg IV, and/or administer
phenobarbital loading dose.
*** Refer to Figure 18.5

Figure 18-3.  Assessment algorithm for newborns with suspected seizures.

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e325

First-line testing*
Glucose 18
Electrolytes
Sepsis evaluation
Consider lumbar puncture
Consider urgent head ultrasound
Birth and family histories
Check placental pathology

Brain MRI

Suspected HIE Stroke Suspected infection Suspected epilepsy,


or cause unknown

Consider evaluation for Echocardiogram if clinical TORCH titers Pyridoxine trial


secondary seizure etiology suspicion for congenital LP with CSF studies: Genetic testing**
(e.g. infection), depending cardiac malformation. Glucose/protein PAA, UOA, lactate, pyruvate, NH3
on clinical scenario. Consider anticoagulation Cell counts Consider CSF testing (AA, neurotransmitters)
for venous thrombosis. HSV PCR Check newborn screen results
Consider thrombophilia Bacterial culture Ophthalmology exam
evaluation in the nonacute
setting.

Neurology and neurodevelopmental follow-up

**Genetic testing can include: karyotype, chromosomal microarray, epilepsy gene panel, or single gene testing.
Figure 18-4.  Assessment algorithm for newborns with seizures. First-line testing should occur simultaneously with initiation of electroen-
cephalography (and empiric seizure treatment in high-risk clinical scenarios). Most infants should receive neuroimaging, and brain magnetic reso-
nance imaging (MRI) is the preferred neuroimaging modality. Second-line testing depends on the clinical scenario and MRI findings. Newborns
with seizures are at high risk for long-term neurodevelopmental disability and epilepsy, and so they require careful follow-up by appropriate
clinicians.

thalamic hemorrhage), an MR venogram may be diagnostic EEG should be initiated as soon as possible to confirm that
and concurrent MRI may reveal venous strokes. clinical events have an electrographic correlate.70 If infection
Further diagnostic testing depends on the clinical scenario. is suspected, appropriate cultures should be drawn and treat-
If the evaluation for infection is negative and there is no other ment initiated, including broad-spectrum antibiotics for a
immediately obvious cause of seizures after MRI and basic suspected bacterial infection and acyclovir for herpes simplex
metabolic workup, then testing for inborn errors of metabo- virus in the appropriate clinical scenario.
lism is reasonable for newborns with abnormal neurologic The maternal and family history must be reviewed. Mater-
examinations and pharmacoresistant seizures. This testing nal nutritional deficiency can affect the developing brain (see
may include measurement of lactate, pyruvate, ammonia, Chapter 47). Prescription and nonprescription drug with-
plasma amino acids, urine organic acids, and sometime drawal is increasingly common in neonates. Maternal selective
advanced testing of the CSF. If the infant has congenital anom- serotonin reuptake inhibitors (SSRI) use in the third trimester
alies and/or dysmorphic features, or if a neonatal epilepsy is a common cause of abnormal movements during the first
syndrome is suspected, genetic testing and consultation 12 hours of life, but these are usually not seizures.81,82
should be considered. Single-gene testing is occasionally Serotonin–norepinephrine reuptake inhibitors (SNRI) with-
helpful, but clinical availability of early onset epilepsy gene drawal may be associated with electroclinical seizures starting
panels is improving and may be more cost-effective. Chromo- in the first 3 days of life.83,84 Even if withdrawal from prescrip-
somal microarray may also reveal clinically significant tion or illicit drugs is suspected, other serious causes of sei-
abnormalities. zures should be ruled out. Convulsions or other abnormal
movements without electrographic correlate need not be
treated with seizure medications. Family history may reveal
TREATMENT BFNE as the likely cause in an otherwise well-appearing
infant.
Acute Treatment
Because new-onset seizures in a neonate often reflect a serious
underlying neurologic condition, they should be treated as a
Treatment of Acute Symptomatic Seizures
medical emergency. Once the bedside clinician has estab- Treatment with antiseizure medications should be initiated as
lished that the vital signs are stable and taken note of the soon as possible once electrographic seizures are confirmed
clinical manifestations of a suspected seizure, glucose must be (or immediately in a patient with a high-risk condition such
checked immediately and electrolytes (including calcium) as HIE, acute intracranial hemorrhage, or clonic motor hemi-
drawn to rule out rapidly treatable causes of seizures. Video convulsions indicating a likely stroke; see Figure 18-3).

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e326 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

Medication should be administered in adequate bolus doses Midazolam infusion is an alternative or add-on agent in
titrated to abolish electrographic seizures (including electro- refractory cases in the setting of acute brain injury and status
graphic seizures without clinical correlate) as quickly as pos- epilepticus.89–91 Lidocaine infusions can be used for refractory
sible (Figure 18-5). acute symptomatic neonatal seizures.92–94 Note that lidocaine
Few data from clinical trials are available to guide anticon- is contraindicated for neonates with congenital heart disease
vulsant management decisions. A suggested treatment algo- and for those who have previously been treated with
rithm is presented in Figure 18-5. According to international phenytoin/fosphenytoin, because of the risk of arrhythmia.
surveys, phenobarbital remains the most commonly used Additionally, lidocaine dosing must be adjusted for neonates
agent for first-line treatment of seizures in newborn infants.85,86 treated with therapeutic hypothermia (phenobarbital dosing
Seizures are controlled in roughly half of patients after a single does not need to be adjusted).93,95 Several excellent reviews
phenobarbital loading dose of 20 mg/kg.13 Phenytoin (or, provide detailed data about seizure treatment options.45,96,97
preferably, IV fosphenytoin in the acute setting) has similar To date, data suggest that the available seizure medications
efficacy to phenobarbital when used at a loading dose of have similar efficacy for acute symptomatic neonatal seizures.
20 mg/kg,13 but maintenance dosing—particularly with oral Therefore multiple potential treatment algorithms may be jus-
administration—is challenging in neonates because of less tifiable. However, establishing a consensus between the neo-
predictable absorption and pharmacokinetics than phenobar- natology and neurology services regarding medication choice,
bital. Levetiracetam is increasingly used in spite of limited dose and timing, can help to facilitate rapid administration
safety and efficacy data. Two studies provide pharmacokinetic of medication, which probably does affect seizure burden,
data, which suggest use of a levetiracetam loading dose of at because infants with fewer seizures are more easily treated.13
least 40 mg/kg and maintenance dosing of at least 10 mg/kg/ Maintenance anticonvulsant medication dosing should be
dose administered every 8 hours can be considered.87,88 initiated for neonates who have confirmed electrographic sei-
If seizures are not controlled after repeated loading doses zures. For newborns with clinical events without proven elec-
of standard medications, infusions are sometimes indicated. trographic seizures, maintenance dosing may be discontinued

Phenobarbital loading dose


20–30 mg/kg

Seizures stop?

Yes No

• Continue electroencephalogram • Continue EEG monitoring


(EEG) monitoring until  24 hours • Repeat 10–20 mg/kg
seizure-free phenobarbital boluses until seizures
• Begin maintenance stop, cumulative dose 40–60 mg/kg,
phenobarbital 4–6 mg/kg or level  40 g/mL
per day in 2 divided doses

No Seizures stop?

Select second-line agent, depending on clinical scenario*

Infrequent seizures, Cardiac abnormality Status epilepticus and Status epilepticus,


no cardiac or hepatic and/or hepatic disorder, no cardiac abnormalities airway secure, and no
abnormalities, and no or and/or cardiovascular and never received hypotension
minimal cardiovascular instability. phenytoin or
instability. fosphenytoin.

Fosphenytoin 20 mg/kg bolus Levetiracetam 40 mg/kg bolus Lidocaine infusion Midazolam (if bolus,
(may repeat 10 mg/kg bolus), (may repeat 20 mg/kg) then (2 mg/kg bolus, then 0.15 mg/kg IV, then 1 g/kg/
then 8–10 mg/kg per day in IV 40–60 mg/kg per day in 6 mg/kg/hour and decrease min infusion, titrating upward
in 2 or 3 divided doses; check 2 or 3 divided doses. by 2 mg/kg/hour every to effect). Wean gradually,
free and total blood levels 12 hours). Maximum usually after 24 hours of
infusion time is 48 hours seizure freedom.
due to arrhythmia risk.

*If the infant has acute symptomatic seizures, select from these options, based on co-morbidities and seizure severity.
If the newborn has epilepsy (e.g. lissencephaly, tuberous sclerosis), consider levetiracetam, topiramate,
or oxcarbazepine as second- or third-line treatments.
Figure 18-5.  Suggested treatment algorithm for neonatal seizures. The rapidity of medication administration will depend on local guidelines
and resources. Frequent assessment of treatment response is recommended.

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e327

and the EEG reviewed frequently to ensure that the neonate “favorable” (survival without impairment) or “unfavorable”
does not subsequently develop electrographic seizures. (death, or intellectual disability, and/or cerebral palsy, and/or 18
epilepsy). The risk for unfavorable outcomes related to neo-
natal seizures is difficult to isolate from the underlying cause
Discontinuation of Medication for Acute of the seizures. Additionally, because many infants with neo-
Symptomatic Seizures natal seizures are critically ill, they often have multiple comor-
bidities that affect long-term outcomes. As an example, a
Acute symptomatic seizures typically arise within the first 24 newborn whose seizures are caused by HIE is at risk for
to 48 hours of life, after which there is a short period of high impaired neurodevelopment because of the acute brain injury,
seizure burden, followed by a longer period of lower seizure as well as the seizures, but might also have neonatal sepsis or
burden.98,99 Overall duration of acute symptomatic seizures is feeding problems that lead to suboptimal nutrition, all of
typically 48 to 96 hours. Knowing the seizure tempo and which may elevate the risk for adverse outcomes.
careful use of video-EEG monitoring to determine when sei-
zures arise and resolve can help guide duration of treatment
with seizure medications. Because the recurrence risk in the Mortality After Neonatal Seizures
neonatal period is low, many neonates can be safely discon- Despite modern neonatal intensive care, 10% to 30% of new-
tinued from medications after resolution of acute symptom- borns with seizures die during the neonatal period.5 The risk
atic seizures.100 There is no need to wean medications that have of death is higher in certain subgroups (e.g., preterm infants).
been used for only a short duration (less than 1 week). If the Geographic variation in reported mortality may reflect cultural
child is to be maintained on medications, the regimen should differences regarding decisions to withdraw intensive care for
be simplified and a plan put in place to reassess the infant neonates with extremely poor prognosis. Among survivors of
after discharge from hospital so that anticonvulsants may be neonatal seizures, those with abnormal neurodevelopment
discontinued during the first few months of life. EEG monitor- remain at elevated risk of death throughout childhood.
ing after the resolution of seizures has a low yield for seizure
detection and rarely adds to the prognostic impression; centers
with limited resources should consider prolonged, continuous Cognitive Outcomes After Neonatal Seizures
video EEG during the acute phase of the admission rather than The longest-term follow-up studies for neonatal seizures
weekly brief intermittent monitoring. included infants with clinically diagnosed seizures.5,104 Among
survivors, 40% to 50% have global developmental delays, and
preterm infants are at higher risk than full-term neo-
Treatment of Early Onset Epilepsy Syndromes nates.5,8,104,105 Among neonates with HIE, a diagnosis of neo-
If the diagnostic evaluation rules out an acute symptomatic natal seizures increases the risk for developmental delays and
cause for neonatal seizures, early onset epilepsy should be intellectual disability, after controlling for the degree of
suspected. The treatment of epilepsy in the neonatal period is encephalopathy.106 Severe seizure burden increases this risk.
different from the approach for acute symptomatic seizures. One study reported that HIE survivors with severe clinical
First, rapid treatment of seizures probably does not affect seizure burden, compared with those with HIE who remained
seizure burden or long-term outcome, and so medications seizure-free, had on average a 30-point (2 standard deviations)
should be carefully titrated to maximally tolerated doses to lower full-scale IQ at age 4 years, even after adjusting for MRI-
ensure efficacy or failure. Second, neonates with epilepsy must detected brain injury. On average, HIE survivors with a lower
continue medications after discharge home, even if seizures seizure burden still scored one full standard deviation lower
have been controlled with medical management. than their seizure-free peers.8
Vitamin-responsive inborn errors of metabolism may
present with neonatal encephalopathy and refractory seizures,
and so pharmacologic doses of pyridoxine, folinic acid, and
Cerebral Palsy After Neonatal Seizures
pyridoxal 5′-phosphate are indicated in this setting.101,102 Neo- About 25% to 35% of long-term neonatal seizure survivors
nates with suspected PDE should be administered a trial of IV develop cerebral palsy.104,105 Most of those with severe cerebral
pyridoxine (100 mg IV while EEG is recording). The infant palsy have comorbid global developmental delays, including
must be monitored during this trial, as apnea and bradycardia intellectual disability. Among survivors of neonatal seizures,
can be provoked. If there is a clear response, test urine alpha- having cerebral palsy is also a significant risk factor for post-
AASA and/or plasma pipecolic acid levels and consider con- neonatal epilepsy.107,108
firmation with ALDH7A1 mutation analysis. If there is no
response to pyridoxine, a trial of pyridoxal 5′-phostphate
(60 mg/kg/day divided 3 times/day for 2–3 days) and then
Postneonatal Epilepsy
folinic acid (2.5 mg IV) may be attempted. Postneonatal epilepsy—in which the acute symptomatic
Carbamazepine and oxcarbazepine are effective and well seizures subside, but recurrent unprovoked seizures develop
tolerated for neonates with seizures caused by KCNQ2/3 later in life—affects 20% to 30% of survivors of neonatal
mutations (either BFNE or KCNQ2 encephalopathy).103 If car- seizures.41,105,109,110 Most often, epilepsy develops in the first few
bamazepine or oxcarbazepine is not effective, a trial of retiga- years of life. Children can have a range of epilepsy syndromes,
bine, which selectively affects the K+ channel, may be depending on the etiology of the neonatal seizures. Most
worthwhile.103 studies have not been powered to describe details of the epi-
lepsy syndromes, but several groups have reported a 10% to
16% incidence of West syndrome after clinical neonatal
OUTCOMES AFTER NEONATAL SEIZURES seizures.104,105,109
Neonatal seizures are associated with an array of adverse out- There are many known risk factors for postneonatal epi-
comes, including intellectual disability, cerebral palsy, post- lepsy. Clinical risk factors include requirement of more than
neonatal epilepsy, and death. Most often in the literature, one medication to control the acute neonatal seizures, moder-
cognitive and behavioral outcomes are not reported separately ate or severe neonatal encephalopathy, abnormal neuroimag-
from cerebral palsy. Instead, outcomes are grouped into ing, and low birth weight. EEG risk factors include status

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e328 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

10. Khazipov R, Khalilov I, Tyzio R, et al. Developmental changes in


BOX 18-1  Neonatal Seizure Pearls GABAergic actions and seizure susceptibility in the rat hippo-
campus. Eur J Neurosci 2004;19:590–600.
• Clinical observation is unreliable for neonatal seizure detection; 11. Dzhala VI, Staley KJ. Excitatory actions of endogenously released
EEG is the gold standard for neonatal seizure diagnosis. GABA contribute to initiation of ictal epileptiform activity in the
• Newborns typically have a high seizure burden—it is rare to developing hippocampus. J Neurosci 2003;23:1840–6.
have only one or two seizures. 12. Dzhala VI, Talos DM, Sdrulla DA, et al. NKCC1 transporter facili-
• Most neonatal seizures have an acute symptomatic cause. tates seizures in the developing brain. Nat Med 2005;11:
• If seizures persist beyond 96 hours, they are probably not 1205–13.
caused by an acute symptomatic cause. 13. Painter MJ, Scher MS, Stein AD, et al. Phenobarbital compared
with phenytoin for the treatment of neonatal seizures. N Engl J
• Seizure treatment should occur in tandem with an expedited
Med 1999;341:485–9.
evaluation for the seizures’ etiology. 14. Abend NS, Sanchez SM, Berg RA, et al. Treatment of electro-
• Electroclinical seizures can become subclinical after treatment graphic seizures and status epilepticus in critically ill children: a
is initiated. single center experience. Seizure 2013;22:467–71.
• Duration of treatment depends on the etiology. 15. Cleary RT, Sun H, Huyhn T, et al. Bumetanide enhances pheno-
barbital efficacy in a rat model of hypoxic neonatal seizures.
PLoS ONE 2013;8:e57148.
16. Dunah AM, Yasuda RP, Wang YH, et al. Regional and ontogenic
expression of the NMDA receptor subunit NR2D protein in rat
brain using a subunit-specific antibody. J Neurochem 1996;67:
epilepticus, persistently abnormal interictal EEG background, 2335–45.
multifocal (vs. focal) seizures, and ictal spread to the contra- 17. Rakhade SN, Jensen FE. Epileptogenesis in the immature brain:
lateral hemisphere.5 emerging mechanisms. Nat Rev Neurol 2009;5:380–91.
Among people with chronic epilepsy, a history of clinical 18. Sanchez RM, Jensen FE. Maturational aspects of epilepsy mecha-
nisms and consequences for the immature brain. Epilepsia
neonatal seizures is a definite risk factor for long-term non-
2001;42:577–85.
remission of the epilepsy.111 By extension, because treatment- 19. Huttenlocher PR, de Courten C, Garey LJ, et al. Synaptogenesis
resistant epilepsy is associated with cognitive and behavioral in human visual cortex—evidence for synapse elimination
challenges, early life seizures may predispose to long-term during normal development. Neurosci Lett 1982;33:247–52.
intellectual and behavioral difficulties. 20. Takashima S, Chan F, Becker LE, et al. Morphology of the devel-
oping visual cortex of the human infant: a quantitative and
qualitative Golgi study. J Neuropathol Exp Neurol 1980;39:
CONCLUSIONS 487–501.
The immature brain is predisposed to seizures. Neonatal sei- 21. Jiang M, Lee CL, Smith KL, et al. Spine loss and other persistent
alterations of hippocampal pyramidal cell dendrites in a model
zures usually reflect acute underlying brain injury, rather than
of early-onset epilepsy. J Neurosci 1998;18:8356–8.
epilepsy. Thus evaluations to confirm the diagnosis of seizures 22. McCabe BK, Silveira DC, Cilio MR, et al. Reduced neurogenesis
(with EEG) and to determine and treat their etiology must after neonatal seizures. J Neurosci 2001;21:2094–103.
occur simultaneously. There are very few data from clinical 23. Montgomery EM, Bardgett ME, Lall B, et al. Delayed neuronal
trials to guide treatment decisions. Phenobarbital is the most loss after administration of intracerebrocentricular kainic
commonly prescribed first-line agent. Controversy remains acid to preweanling rats. Brain Res Dev Brain Res 1999;112:
regarding the ideal duration of treatment. However, it is clear 107–16.
that neonatal seizures are associated with substantial risk of 24. Lynch M, Sayin U, Bownds J, et al. Long-term consequences of
mortality and of long-term neurodevelopmental disability early postnatal seizures on hippocampal learning and plasticity.
Eur J Neurosci 2000;12:2252–64.
and epilepsy. High-quality research is urgently needed in order
25. Yager JY, Armstrong EA, Jaharus C, et al. Preventing hyperther-
to determine the best treatment algorithms and, ultimately, to mia decreases brain damage following neonatal hypoxic-
improve long-term outcomes. ischemic seizures. Brain Res 2004;1011:48–57.
26. Vasudevan C, Levene M. Epidemiology and aetiology of neonatal
REFERENCES seizures. Semin Fetal Neonatal Med 2013;18:185–91.
1. Annegers JF, Hauser WA, Lee JR, et al. Incidence of acute symp- 27. Glass HC, Pham TN, Danielsen B, et al. Antenatal and intrapar-
tomatic seizures in Rochester, Minnesota, 1935–1984. Epilepsia tum risk factors for seizures in term newborns: a population-
1995;36:327–33. based study, California 1998–2002. J Pediatr 2009;154:24–8.
2. Jensen FE. Neonatal seizures: an update on mechanisms and 28. Hall DA, Wadwa RP, Goldenberg NA, et al. Maternal risk factors
management. Clin Perinatol 2009;36:881–900. for term neonatal seizures: population-based study in Colorado,
3. Nardou R, Ferrari DC, Ben-Ari Y. Mechanisms and effects of 1989–2003. J Child Neurol 2006;21:795–8.
seizures in the immature brain. Semi Fetal Neonatal Med 29. Ronen GM, Penney S, Andrews W. The epidemiology of clinical
2013;18:175–84. neonatal seizures in Newfoundland: a population-based study.
4. Holmes GL. The Long-term effects of neonatal seizures. Clin J Pediatr 1999;134:71–5.
Perinatol 2009;36:901–14. 30. Lanska MJ, Lanska DJ. Neonatal seizures in the United States:
5. Uria-Avellanal C, Marlow N, Rennie JM. Outcome following results of the National Hospital Discharge Survey, 1980–1991.
neonatal seizures. Semin Fetal Neonatal Med 2013;18: Neuroepidemiology 1996;15:117–25.
224–32. 31. Lanska MJ, Lanska DJ, Baumann RJ, et al. A population-based
6. Glass HC, Nash KB, Bonifacio SL, et al. Seizures and magnetic study of neonatal seizures in Fayette County, Kentucky. Neurol-
resonance imaging-detected brain injury in newborns cooled ogy 1995;45:724–32.
for hypoxic-ischemic encephalopathy. J Pediatr 2011;159: 32. Saliba RM, Annegers FJ, Waller DK, et al. Incidence of neonatal
731–5. seizures in Harris County, Texas, 1992–1994. Am J Epidemiol
7. Miller SP, Weiss J, Barnwell A, et al. Seizure-associated brain 1999;150:763–9.
injury in term newborns with perinatal asphyxia. Neurology 33. Sheth RD, Hobbs GR, Mullett M. Neonatal seizures: incidence,
2002;58:542–8. onset, and etiology by gestational age. J Perinatol 1999;19:
8. Glass HC, Glidden D, Jeremy RJ, et al. Clinical neonatal seizures 40–3.
are independently associated with outcome in infants at risk for 34. Holden KR, Mellits ED, Freeman JM. Correlation of prenatal and
hypoxic-ischemic brain injury. J Pediatr 2009;155:318–23. perinatal events with outcomes. Pediatrics 1982;70:165–76.
9. Dulac O, Milh M, Holmes GL. Brain maturation and epilepsy. 35. Wusthoff CJ. Diagnosing neonatal seizures and status epilepti-
Handb Clin Neurol 2013;111:441–6. cus. J Clin Neurophysiol 2013;30:115–21.

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Neonatal Seizures e329

36. Pavlidis E, Spagnoli C, Pelosi A, et al. Neonatal status epilepti- 59. Weckhuysen S, Mandelstam S, Suls A, et al. KCNQ2 encepha-
cus: differences between preterm and term newborns. Eur J lopathy: emerging phenotype of a neonatal epileptic encepha- 18
Pediatr Neurol 2015;19:314–19. lopathy. Ann Neurol 2012;71:15–25.
37. Payne ET, Hahn CD. Continuous electroencephalography for 60. Beal JC, Cherian K, Moshé SL. Early-onset epileptic encephalopa-
seizures and status epilepticus. Curr Opin Pediatr 2014;26: thies: Ohtahara syndrome and early myoclonic encephalopathy.
675–81. Pediatr Neurol 2012;47:317–23.
38. Shellhaas RA, Clancy RR. Characterization of neonatal seizures 61. Volpe JJ. Chaper 5: neonatal seizures. Neurology of the newborn.
by conventional and single channel EEG. Clin Neurophysiol 5th ed. Philadephia, PA: Saunders Elsevier; 2008.
2007;118:2156–61. 62. Mizrahi EM, Kellaway P. Characterization and classification of
39. Saliba RM, Annegers FJ, Waller DK, et al. Risk factors for neona- neonatal seizures. Neurology 1987;37:1837–44.
tal seizures: a population-based study, Harris County, Texas, 63. Murray DM, Boylan GB, Ali I, et al. Defining the gap between
1992–1994. Am J Epidemiol 2001;154:14–20. electrographic seizure burden, clinical expression and staff rec-
40. Kohelet D, Shochat R, Lusky A, et al. Risk factors for seizures in ognition of neonatal seizures. Arch Dis Child Fetal Neonatal Ed
very low birthweight infants with periventricular leukomalacia. 2008;93:F187–91.
J Child Neurol 2006;21:965–70. 64. Malone A, Ryan CA, Fitzgerald A, et al. Interobserver
41. Pisani F, Cerminara C, Fusco C, et al. Neonatal status epilepticus agreement in neonatal seizure identification. Epilepsia 2009;
vs. recurrent neonatal seizures: clinical findings and outcome. 50:2097–101.
Neurology 2007;69:2177–85. 65. Clancy RR, Legido ADL. Occult neonatal seizures. Epilepsia
42. Pisani F, Copioli C, Di Gioia C, et al. Neonatal seizures: 1988;29:256–61.
relation of ictal video-electroencephalography (EEG) findings 66. Clancy RR, Sharif U, Ichord R, et al. Electrographic neonatal
with neurodevelopmental outcome. J Child Neurol 2008;23: seizures after infant heart surgery. Epilepsia 2005;46:84–
394–8. 90.
43. Tekgul H, Gauvreau K, Soul J, et al. The current etiologic profile 67. Scher MS, Alvin J, Gaus L, et al. Uncoupling of EEG-clinical
and neurodevelopmental outcome of seizures in term newborn neonatal seizures after antiepileptic drug use. Pediatr Neurol
infants. Pediatrics 2006;117:1270–80. 2003;28:277–80.
44. Mastrangelo M, van Lierde A, Bray M, et al. Epileptic seizures, 68. Cherian PJ, Deburchgraeve W, Swarte RM, et al. Validation of a
epilepsy, and epileptic syndromes in newborns: a nosological new automated neonatal seizure detection system: a clinician’s
approach to 94 new cases by the 2001 proposed diagnostic perpective. Clin Neurophysiol 2011;122:1490–9.
scheme for people with epileptic seizures and with epilepsy. 69. Tsuchida TN, Wusthoff CJ, Shellhaas RA, et al. American clinical
Seizure 2005;14:304–11. neurophysiology society standardized EEG terminology and cat-
45. Glass HC, Wusthoff CJ, Shellhaas RA, et al. Risk factors for EEG egorization for the description of continuous EEG monitoring
seizures in neonates treated with hypothermia: a multi-center in neonates: report of the American Clinical Neurophysiology
cohort study. Neurology 2014;82:1–6. Society critical care monitoring committee. J Clin Neurophysiol
46. Low E, Boylan GB, Mathieson SR, et al. Cooling and seizure 2013;30:161–73.
burden in term neonates: an observational study. Arch Dis Child 70. Shellhaas RA, Chang T, Tsuchida T, et al. The American Clinical
Fetal Neonatal Ed 2012;97:F267–72. Neurophysiology Society’s guideline on continuous electroen-
47. Shah PS. Hypothermia: a systematic review and meta-analysis cephalography monitoring in neonates. J Clin Neurophysiol
of clinical trials. Semin Fetal Neonatal Med 2010;15: 2011;28:611–17.
238–46. 71. Epstein CM. Guidelines two: minimum technical standards for
48. Wusthoff CJ, Dlugos DJ, Gutierrez-Colina A, et al. Electrographic pediatric electroencephalography. J Clin Neurophysiol 2006;23:
seizures during therapeutic hypothermia for neonatal hypoxic- 92–6.
ischemic encephalopathy. J Child Neurol 2011;26:724–8. 72. Laroia N, Guillet R, Burchfiel J, et al. EEG background as predic-
49. Boylan GB, Kharoshankaya L, Wusthoff CJ. Seizures and hypo- tor of electrographic seizures in high-risk neonates. Epilepsia
thermia: importance of electroencephalographic monitoring 1998;39:545–51.
and considerations for treatment. Semin Fetal Neonatal Med 73. Nash KB, Bonifacio SL, Glass HC, et al. Video-EEG monitoring
2015;20:103–9. in newborns with hypoxic-ischemic encelphalopathy treated
50. Raju TN, Nelson KB, Ferriero DM, et al. NICHD-NINDS Perina- with hypothermia. Neurology 2011;76:556–62.
tal Stroke Workshop Participants. Ischemic perinatal stroke: 74. Osredkar D, Toet MC, van Rooij LGM, et al. Sleep-wake cycling
summary of a workshop sponsored by the National Institute of on amplitude-integrated electroencephalography in term new-
Child Health and Human Development and the National Insti- borns with hpoxic-ischemic encephalopathy. Pediatrics 2005;
tute of Neurological Disorders and Stroke. Pediatrics 2007;120: 115:327–32.
609–16. 75. Thoresen M, Hellström-Westas L, Liu X, et al. Effect of hypother-
51. Kirton A, Armstrong-Wells J, Chang T, et al. Symptomatic neo- mia on amplitude-integrated electroencephalogram in infants
natal arterial ischemic stroke: the International Pediatric Stroke with asphyxia. Pediatrics 2010;126:e131–9.
Study. Pediatrics 2011;128:e1402–10. 76. Hellström-Westas L, de Vries LS, Rosen I. Atlas of amplitude-
52. Harteman JC, Groenendaal F, Kwee A, et al. Risk factors for integrated EEGs in the newborn. 2nd ed. London: Informa
perinatal arterial ischemic stroke in full-term infants: a case- Healthcare; 2008.
control study. Arch Dis Child Fetal Neonatal Ed 2012;97: 77. Tao JD, Mathur AM. Using amplitude-integrated EEG in neona-
F411–16. tal intensive care. J Perinatol 2010;30:S73–81.
53. Darmency-Stamboul V, Chantegret C, Ferdynus C, et al. Antena- 78. Hallberg B, Grossmann K, Bartocci M, et al. The prognostic value
tal factors associated with perinatal arterial ischemic stroke. of early aEEG in asphyxiated infants undergoing systemic hypo-
Stroke 2012;43:2307–12. thermia treatment. Acta Paediatr 2010;99:531–6.
54. Berfelo FJ, Kersbergen KJ, van Ommen CH, et al. Neonatal cere- 79. Massaro AN, Tsuchida T, Kadom N, et al. aEEG evolution during
bral sinovenous thrombosis from symptom to outcome. Stroke therapeutic hypothermia and prediction of NICU outcome
2010;41:1382–8. in encephalopathic neonates. Neonatology 2012;102:197–
55. Jordan LC, Rafay MF, Smith SE, et al. Antithrombotic treatment 202.
in neonatal cerebral sinovenous thrombosis: results of the Inter- 80. Glass HC, Wusthoff CJ, Shellhaas RA. Amplitude integrated EEG:
national Pediatric Stroke Study. J Pediatr 2010;156:704–10. the child neurologist’s perspective. J Child Neurol 2013;28:
56. Loman AM, ter Horst HJ, Lambrechtsen FA, et al. Neonatal sei- 1342–50.
zures: aetiology by means of a standardized work-up. Eur J Pae- 81. Hayes RM, Wu P, Shelton RC, et al. Maternal antidepressant use
diatr Neurol 2014;18:360–7. and adverse outcomes: a cohort study of 228,876 pregnancies.
57. Boardman JP, Wusthoff CJ, Cowan FM. Hypoglycaemia and neo- Am J Obstet Gynecol 2012;207:e1–9.
natal brain injury. Arch Dis Child Educ Pract Ed 2013;98:2–6. 82. Moses-Kolko EL, Bogen D, Perel J, et al. Neonatal signs after late
58. Von Hove JL, Lohr NJ. Metabolic and monogenic causes of sei- in utero exposure to serotonin reuptake inhibitors: literature
zures in neonates and young infants. Mol Genet Metab 2011; review and implications for clinical applications. JAMA 2005;
104:214–30. 293:2372–83.

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
e330 PART IV  Perinatal Acquired and Congenital Neurologic Disorders

83. Hoppenbrouwers CJ, Bosma J, Wennink HJ, et al. Neonatal sei- 97. Pressler RM, Mangum B. Newly emerging therapies for neonatal
zures on EEG after in utero exposure to venlafaxine. Br J Clin seizures. Semin Fetal Neonatal Med 2013;18:216–23.
Pharmacol 2010;70:454–6. 98. Lynch NE, Stevenson NJ, Livingstone V, et al. The temporal evo-
84. Haukland LU, Kutzsche S, Hovden IA, et al. Neonatal seizures lution of electrographic seizure burden in neonatal hypoxic
with reversible EEG changes after antenatal venlafaxine expo- ischemic encephalopathy. Epilepsia 2012;53:549–57.
sure. Acta Paediatr 2013;102:e524–6. 99. Abend NS, Wusthoff CJ, Goldberg EM, et al. Electrographic sei-
85. Bartha AI, Shen J, Katz KH, et al. Neonatal seizures: multicenter zures and status epilepticus in critically ill children and neonates
variability in current treatment practices. Pediatr Neurol with encephalopathy. Lancet Neurol 2013;12:1170–9.
2007;37:85–90. 100. Guillet R, Kwon J. Seizure recurrence and developmental dis-
86. Glass HC, Kan J, Bonifacio SL, et al. Neonatal seizures: treatment abilities after neonatal seizures: outcomes are unrelated to use
practices among term and preterm infants. Pediatr Neurol of Phenobarbital prophylaxis. J Child Neurol 2007;22:
2012;26:111–15. 389–95.
87. Merhar SL, Schibler KR, Sherwin CM, et al. Pharmacokinetics of 101. Stockler S, Plecko B, Gospe SM Jr, et al. Pyridoxine dependent
levetiracetam in neonates with seizures. J Pediatr 2011;159: epilepsy and antiquitin deficiency: clinical and molecular char-
152–4. acteristics and recommendations for diagnosis, treatment and
88. Sharpe CM, Capparelli EV, Mower A, et al. A seven-day study of follow-up. Mol Genet Metab 2011;104:48–60.
the pharmacokinetics of intravenous levetiracetam in neonates: 102. Gospe SM Jr. Neonatal vitamin-responsive epileptic encepha-
marked changes in pharmacokinetics occur during the first week lopathies. Chang Gung Med J 2010;33:1–12.
of life. Pediatr Res 2012;72:43–9. 103. Numis AL, Angriman M, Sullivan JE, et al. KCNQ2 encephalopa-
89. Castro Conde JR, Hernandez Borges AA, Domenech Martinez E, thy: delineation of the electroclinical phenotype and treatment
et al. Midazolam in neonatal seizures with no response to phe- response. Neurology 2014;82:368–70.
nobarbital. Neurology 2005;64:876–9. 104. Ronen GM, Buckley D, Penney S, et al. Long-term prognosis in
90. Shany E, Benzaqen O, Watemberg N. Comparison of continuous children with neonatal seizures: a population based study. Neu-
drip of midazolam or lidocaine in the treatment of intractable rology 2007;69:1816–22.
neonatal seizures. J Child Neurol 2007;22:255–9. 105. Garfinkle J, Shevell MI. Cerebral palsy, developmental delay,
91. Yamamoto H, Aihara M, Niijima S, et al. Treatments with mid- and epilepsy after neonatal seizures. Pediatr Neurol 2011;44:
azolam and lidocaine for status epilepticus in neonates. Brain 88–96.
Dev 2007;29:559–64. 106. Wyatt JS, Gluckman PD, Liu PY, et al. Determinants of outcomes
92. Malingre MM, VanRooij LGM, Rademaker CMA, et al. Develop- after head cooling for neonatal encephalopathy. Pediatrics
ment of an optimal lidocaine infusion strategy for neonatal 2007;119:912–21.
seizures. Eur J Pediatr 2006;165:598–604. 107. Mert GG, Incecik F, Altunbasak S, et al. Factors affecting epilepsy
93. van den Broek MPH, Rademaker CMA, van Straaten HLM, et al. development and epilepsy prognosis in cerebral palsy. Pediatr
Anticonvulsant treatment of asphyxiated newborns under hypo- Neurol 2011;45:89–94.
thermia with lidocaine: efficacy, safety, and dosing. Arch Dis 108. Yildiz EP, Tatli B, Ekici B, et al. Evaluation of etiologic and prog-
Child Fetal Neonatal Ed 2013;98:F341–5. nostic factors in neonatal convulsions. Pediatr Neurol 2012;47:
94. Lundqvist M, Agren J, Hellström-Westas L, et al. Efficacy and 186–92.
safety of lidocaine for treatment of neonatal seizures. Acta Pae- 109. Nunes ML, Martins MA, Barea BM, et al. Neurological outcomes
diatr 2013;102:863–7. of newborns with neonatal seizures. Arq Neuropsiquiatr
95. Shellhaas RA, Ng CM, Dillon CH, et al. Population pharmaco- 2008;66:168–74.
kinetics of phenobarbital in infants with neonatal encephalopa- 110. Painter MJ, Sun Q, Scher MS, et al. Neonates with seizures: what
thy treated with therapeutic hypothermia. Pediatr Crit Care Med predicts development? J Child Neurol 2012;27:1022–6.
2013;14:194–202. 111. Sillanpää M, Camfield P, Camfield C. Predicting long-term
96. Hellström-Westas L, Boylan GB, Agren J. Systematic review of outcome of childhood epilepsy in Nova Scotia, Canada, and
neonatal seizure management strategies provides guidance on Turku, Finland. Validation of a simple scoring system. Arch
anti-epileptic treatment. Acta Paediatr 2015;104:123–9. Neurol 1995;52:589–92.

Downloaded for FK Universitas Sam Ratulangi (mahasiswafkunsrat19@gmail.com) at Sam Ratulangi University from ClinicalKey.com by
Elsevier on April 22, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.

You might also like