Bravo 2021

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Drugs (2021) 81:749–770

https://doi.org/10.1007/s40265-021-01502-4

REVIEW ARTICLE

Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive


Status Epilepticus
Pablo Bravo1 · Aparna Vaddiparti1 · Lawrence J. Hirsch1

Accepted: 13 March 2021 / Published online: 8 April 2021


© The Author(s), under exclusive licence to Springer Nature Switzerland AG 2021

Abstract
Most seizures in critically ill patients are nonconvulsive. A significant number of neurological and medical conditions can be
complicated by nonconvulsive seizures (NCSs) and nonconvulsive status epilepticus (NCSE), with brain infections, hemor-
rhages, global hypoxia, sepsis, and recent neurosurgery being the most prominent etiologies. Prolonged NCSs and NCSE
can lead to adverse neurological outcomes. Early recognition requires a high degree of suspicion and rapid and appropriate
duration of continuous electroencephalogram (cEEG) monitoring. Although high quality research evaluating treatment with
antiseizure medications and long-term outcome is still lacking, it is probable that expeditious pharmacological management
of NCSs and NCSE may prevent refractoriness and further neurological injury. There is limited evidence on pharmaco-
therapy for NCSs and NCSE, although a few clinical trials encompassing both convulsive and NCSE have demonstrated
similar efficacy of different intravenous (IV) antiseizure medications (ASMs), including levetiracetam, valproate, lacosamide
and fosphenytoin. The choice of specific ASMs lies on tolerability and safety since critically ill patients frequently have
impaired renal and/or hepatic function as well as hematological/hemodynamic lability. Treatment frequently requires more
than one ASM and occasionally escalation to IV anesthetic drugs. When multiple ASMs are required, combining different
mechanisms of action should be considered. There are several enteral ASMs that could be used when IV ASM options have
been exhausted. Refractory NCSE is not uncommon, and its treatment requires a very judicious selection of ASMs aiming
at reducing seizure burden along with management of the underlying condition.

1 Introduction
Key Points
Status epilepticus (SE), as defined by the International
League Against Epilepsy (ILAE), is “a condition result- Nonconvulsive seizures and nonconvulsive status epilep-
ing either from the failure of the mechanisms responsible ticus are common complications of acute neurological
for seizure termination or from the initiation of mecha- brain injury and are associated with worse neurological
nisms which lead to abnormally prolonged seizures (after outcomes, including later epilepsy, functional status, and
time point t1). It is a condition that can have long-term cognition.
consequences (after time point t2), including neuronal Early clinical suspicion, rapid diagnosis, and appropriate
death, neuronal injury, and alteration of neuronal net- length of monitoring with continuous EEG is funda-
works, depending on the type and duration of seizures” mental in the diagnosis and treatment of nonconvulsive
[1]. Further details in the classification of SE in regard to seizures and nonconvulsive status epilepticus.
semiology, etiology, EEG correlates, and age can be found
in the Report of the ILAE Task Force on Classification of Seizure burden reduction while avoiding iatrogenicity is
Status Epilepticus [1]. the mainstay of treatment of nonconvulsive seizures and
nonconvulsive status epilepticus.

* Lawrence J. Hirsch
lawrence.hirsch@yale.edu
1
Comprehensive Epilepsy Center, Department of Neurology,
Yale University School of Medicine, New Haven, CT, USA

Vol.:(0123456789)
750 P. Bravo et al.

T1 and t2 have been clearly defined by the ILAE for appropriate monitoring with continuous electroencephalo-
generalized convulsive SE as well as for focal SE with gram (cEEG), as well as limited pharmacotherapy evidence
impairment of consciousness (5 and 30 min for the former, specific to the treatment of NCSs and NCSE. We also pro-
10 and 60 min for the latter), in part as a result of evidence pose a stepwise description of our approach to treating NCSs
in animal experiments [2–6] and clinical research [7–10]. and NCSE that clinicians may find practical, even if based
There is still limited evidence for the exact time threshold on limited evidence. Finally, we highlight areas that need
for long-term consequences in any type of SE other than further research.
convulsive. This threshold is even less clear for frequent
nonconvulsive seizures (NCSs) not qualifying as SE. It
is also likely that t2 is variable, being shorter in patients 2 The Quest to Define t2 in NCSs and NCSE
with acute brain injury compared to patients with chronic
epilepsy [11]. Several animal models have demonstrated that repeated
Since nonconvulsive status epilepticus (NCSE) is, by NCSs and NCSE can cause behavioral deficits [27, 28], epi-
definition, simply SE without prominent motor symptoms lepsy [29, 30], and neuronal death [3, 31]. In a rodent model
[1], the signs and symptoms of NCSE (and NCSs) are of NCSs elicited by irreversible occlusion of the middle cere-
broad and often subtle, including, although not limited bral artery, not only did NCSs correlate with increased infarct
to, inattention, disorientation, confusion, abulia, abnormal size and mortality but also their treatment with antiseizure
eye movements (e.g. gaze deviation or nystagmus), subtle medications reduced mortality [32]. In another rodent model
repetitive facial or distal movements of extremities, and of temporal onset complex partial NCSE [33], brain pathol-
in more severe cases, stupor and coma [12–14]. Electro- ogy 1-week post-NCSE showed active inflammation with
graphically, NCSs may range from focal to generalized, fewer synaptic proteins and excessive microglial and astro-
brief and single, to frequent and multiple, including cyclic, cytic activation; moreover, 4-week post-NCSE brain pathol-
as well as prolonged and continuous [15, 16]. ogy showed continued immune response with neuronal loss.
NCSs and NCSE may occur in virtually any condition Understandably, the search for evidence of neuronal
that may affect, directly or indirectly, the homeostasis of injury and adverse outcomes in humans with NCSs and
the supratentorial brain. This includes cerebral anoxia, NCSE in acute brain injury is a very complex problem given
infarcts, intracranial hemorrhage (intraparenchymal/sub- the significant heterogeneity in the multiple underlying eti-
dural/subarachnoid), neoplasm, infection, traumatic brain ologies that on their own pose a significant risk for detri-
injury, sepsis, prior history of epilepsy, metabolic abnor- mental neurological outcome. Whether NCSs and NCSE
malities, medication overdose/toxicity (e.g., cefepime), can cause neuronal injury and/or increase mortality versus
and neurodegenerative disorders, among others [1]. being just a surrogate of increased severity of the underlying
Previous studies have attempted to define the incidence, acute brain injury has long been a topic of significant debate
prevalence, and mortality of SE and NCSE [11, 16–22]. [34, 35]. Many studies on different etiologies of acute brain
However, given the evolution in the classification of SE in injury in all ages have demonstrated an independent and
the last few decades (from > 30 min to > 5 min threshold “dose-related” (proportional to seizure burden) association
for t1 for convulsive SE), previous estimates of its epide- of worse outcome with NCSs/NCSE [36–49].
miology may not accurately reflect the current understand- Some studies have more clearly demonstrated a correla-
ing [23]. In a recent prospective population-based study tion between length of NCSs/NCSE and worse outcome after
evaluating the incidence of SE based on the latest ILAE controlling for other risk factors. A retrospective study on
classification, although applying a pragmatic > 10-min 49 critically ill patients with NCSs and NCSE, demonstrated
time period for SE for all seizure types, NCSE comprised that patients with NCSs/NCSE had an increased mortality
18% of all cases, and 30-day mortality for SE of all seizure with multivariate logistic regression showing that of all fac-
types was 4.6% [24]. tors studied, only seizure duration and delay to diagnosis
Regardless of the differences in reported epidemiology of were associated with increased mortality [36]. A prospec-
SE over the past several decades, the substantial morbidity tive observational study evaluating 259 infants and children
and mortality of SE, as well as its direct economic burden in admitted to the intensive care unit (ICU), adjusting for diag-
the healthcare system are undeniable [25, 26]. Therefore, a nosis and illness severity, concluded that a seizure burden
clear understanding on the diagnosis and treatment of NCSs threshold > 20% per hour (12 min) was independently asso-
and NCSE is vital. ciated with a greater probability and magnitude of neurolog-
In this review, we discuss the current knowledge on ical decline across all diagnostic categories, although seizure
treatment for NCSs and NCSE, concentrating on acutely ill burden was not associated with mortality [43].
patients including available evidence on secondary neuro- Another prospective observational study evaluating
logical injury due to NCSs and NCSE, rationale for early and outcome on 60 critically ill children (median of 2.7 years
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 751

follow-up) demonstrated that patients with NCSE, but not important to remember that after convulsive SE is con-
NCSs, were associated with an increased risk of unfavorable trolled, nearly half of the patients may develop NCSs in the
global outcome, lower health-related quality of life scores, following 24 h [16]. Therefore, appropriate identification
and an increased risk of subsequently diagnosed epilepsy of the population at risk [58], recognition of possible sub-
even after adjusting for neurologic disorder category, EEG tle symptoms and signs [1], early and appropriate length of
background category, and age [44]. EEG monitoring [55, 59–61] are all paramount in the diag-
Similarly, in a prospective study on 402 consecutive adult nosis and ultimate treatment of NCSs and NCSE. Moreover,
patients with subarachnoid hemorrhage (SAH), after adjust- accurate determination of electrographic patterns and cri-
ing for established predictors, patients with seizures (12% teria for NCSs and NCSE is fundamental [58, 62–65]. The
of all patients, median seizure burden 6 h) had more than most widely accepted criteria for the diagnosis of NCSE
3 times higher odds of disability or death. Seizure burden are the Salzburg criteria [66, 67]; a modified version of the
was associated with worse functional outcome: the higher Salzburg criteria and all other related EEG patterns has been
the seizure burden, the worse the outcome with every hour incorporated into the American Clinical Neurophysiology
of seizure on cEEG associated with 10% higher odds of dis- Society (ACNS)’s Standardized Critical Care EEG Termi-
ability and mortality at 3 months [45]. nology, 2021 version [68].
Finally, delay in treatment can lead to further refractori- In a study evaluating 4772 hospitalized patients under-
ness [50–52]. A prospective study on encephalopathic criti- going cEEG with a subsequent validation study of 1407
cally ill children investigated treatment efficacy before and patients from a different population compared to the foun-
after implementing a pathway to expedite EEG monitoring dational study, it was determined that when different elec-
and seizure management. Data were collected on 41 patients trographic patterns of interictal activity as well as history
before and 21 patients after pathway implementation. The of recent or remote seizures were taken into account, the
median duration from seizure onset to antiseizure medica- length of EEG monitoring necessary to identify subsequent
tion administration was shorter for patients treated with the seizures could be predicted with accuracy after one hour of
pathway compared to the baseline group (64 min vs 139 min, EEG monitoring [59, 61, 65]. This risk stratification was fur-
p = 0.0006). Importantly, seizure termination was much ther validated in a recent study on acute brain injury patients
more likely to occur following initial antiseizure medica- [69]. These risk factors and the length of EEG monitoring
tion administration after implementing the pathway than in necessary to decrease the risk of missing NCSs are depicted
the baseline group (67% vs 27%, p = 0.002) [53] . Also, a in Fig. 1.
retrospective observational study on 625 neonates and pedi- It is worth noting that while coma (and other risk factors
atric patients, in whom NCSs occurred in 28.2% and SE in evaluated in the studies mentioned above, including acute
11.4%, after controlling for gender and age, showed that both structural brain injury) did not have a statistically signifi-
SE and time from ICU admission to cEEG initiation were cant association with subsequent seizures, cardiac arrest
independent predictors of mortality in both neonates and patients were not included in the validation study. There-
pediatric patients [54]. fore, such criteria cannot be used to determine length of
Based on the available evidence to date, it is reasonable monitoring when evaluating cardiac arrest patients. This is
to conclude that the longer NCSs and NCSE last, the worse evident in a retrospective study on 570 critically ill patients
the outcome. While we cannot conclusively define a t2 at [55] also evaluating the required length of cEEG to detect
this time, it is prudent to take a > 12 min per hour seizure seizures, where cardiac arrest patients were not excluded
burden [43] as a measure of concern for potential neuronal and where coma did play a significant role in determining
injury and that every extra hour of seizure burden might the length required to capture the first seizure. Patients with
worsen neurological outcome [45]. Prospective randomized coma required greater than 48 h of cEEG without seizures to
clinical trials evaluating long-term outcomes after treatment reach a > 93% certainty of not having seizures with further
of NCSs/NCSE with antiseizure medications have not been monitoring.
performed; thus, aggressive treatment methods remain A comparison of routine EEG and continuous EEG moni-
unclear. toring to identify seizures in the ICU was investigated in a
recent meta-analysis involving 16,707 patients. The preva-
lence of either NCSs or NCSE was significantly higher with
3 Early Recognition of NCSs and NCSE continuous EEG (15.6%; odds ratio [OR] = 2.57) than with
routine EEG (6.3%) [70]. This was also evident in a recent
Since most seizures in critically ill patients are nonconvul- multicenter randomized “pragmatic” clinical trial in Europe
sive [19, 55], the diagnosis of NCSs and NCSE requires comparing serial routine EEGs to continuous EEG in 364
a high degree of suspicion given their heterogenous and patients who had never had a clinical seizure, including
usually subtle or undetectable semiology [56, 57]. It is also those with hypoxic-ischemic encephalopathy, with Glasgow
752 P. Bravo et al.

Fig. 1  Duration of cEEG without seizures required to avoid miss- cEEG continuous electroencephalogram, LPD lateralized periodic
ing seizures with a certainty of 95 and 98% based on history of any discharges, LRDA lateralized rhythmic delta activity. Plus features:
previous seizures and electrographic patterns (2HELPS2B score: R superimposed rhythmic, S superimposed sharp waves or spikes, F
see box on right side) [59, 61]. BIPD bilateral independent peri- superimposed fast activity
odic discharges, BIRDS brief potentially ictal rhythmic discharges,

Coma Scale < 11 and follow-up of 6 months [71]. While large database study including > 40,000 patients, use of con-
there were more seizures detected in the cEEG arm than tinuous EEG rather than routine EEG was associated with
the routine EEG arm, there was no significant difference lower mortality and no difference in cost [73].
in mortality, despite potential modifications in pharmaco-
therapy for seizures based on the cEEG results. However,
there were many methodological limitations in this study, 4 Treatment of NCSs and NCSE
including a marked delay from admission to initial cEEG
hookup (mean 60 h) and no EEG review overnight, leading 4.1 Evidence on Pharmacotherapy for NCSs
to marked delay in treatment of detected seizures (timing of and NCSE
treatment not provided). Furthermore, only 21% of patients
undergoing cEEG had any change in treatment (vs 11% in To date, there has only been one prospective, randomized,
those with routine EEG), leading to a markedly underpow- blinded, controlled trial evaluating the efficacy and safety of
ered study; thus, it is not surprising that no effect was found antiseizure medications (ASMs) in the treatment of NCSs.
on mortality. Lastly, treating NCSE early is done mainly to The TRENdS trial (Treatment of Recurrent Electrographic
preserve neurons to improve overall neurological function, Nonconvulsive Seizures) was a noninferiority, prospective,
cognition and to prevent later epilepsy; these were not part multicenter, randomized treatment trial of patients diag-
of the primary or secondary outcome measures in that trial, nosed with NCSs by cEEG [74]. Inclusion criteria included
although there was no effect of cEEG on 6-month functional patients with electrographic seizures with or without clini-
outcome in a tertiary outcome measure. It remains unclear cal correlate lasting > 10 s and < 30 min, and refractory to
whether a larger study, where post-cardiac arrest patients can at least 2 ASMs. Treatment was randomized to intravenous
be separately studied or excluded, would have demonstrated (IV) lacosamide 400 mg or IV fosphenytoin 20 mg/kg. The
different results, or if more rapid cEEG, more timely inter- primary endpoint was absence of seizures for 24 h deter-
pretation, and earlier treatment would lead to better cogni- mined by a blinded EEG reviewer. Seizures were controlled
tive and functional outcomes, and lower rates of epilepsy. in 19 of 30 (63.3%) patients in the lacosamide arm and 16
A retrospective study surveying over 7 million US adult of 32 (50%) subjects in the fosphenytoin arm. This trial
ventilated ICU patients (excluding cardiac arrest), of whom demonstrated noninferiority of lacosamide to fosphenytoin
22,728 received cEEG, showed that the use of cEEG was (p = 0.02 for noninferiority) with similar treatment-related
associated with reduced in-hospital mortality after adjust- adverse events (AEs).
ment for patient and hospital characteristics [72]. In a similar
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 753

The most pivotal randomized clinical trial for treatment signs including nystagmoid or rhythmic eye movements
of convulsive SE conducted in 16 Veterans Affairs medical (i.e., had transitioned to nonconvulsive SE). The primary
centers [8] also shed some light on the efficacy of different outcome was cessation of status epilepticus and improve-
ASMs for subtle SE, a prior term that is now considered a ment in the level of consciousness at 60 min. The primary
type of NCSE [1]. In this trial, 384 patients with overt con- safety outcome was life-threatening hypotension or cardiac
vulsive SE and 134 patients with subtle convulsive SE were arrhythmia within 60 min after the start of trial drug dura-
included. Subtle generalized convulsive SE was considered tion. There was no statistically significant difference in the
present when the patient had coma and ictal discharges on success rate or AEs with nearly identical rates of success
the EEG with or without subtle convulsive movements. Four (levetiracetam 47%, phenytoin 45%, valproate 46%). After
IV regimens were used: diazepam (0.15 mg/kg) followed by extending enrollment in children, for a total of 478 patients,
phenytoin (18 mg/kg), lorazepam (0.1 mg/kg), phenobarbi- the authors then described similar results when comparing
tal (15 mg/kg) and phenytoin (18 mg/kg). Treatment was three populations of patients (< 18 years vs 18–65 years vs
considered successful if all clinical and electrical evidence > 65 years) [76].
of seizure activity stopped within 20 min after the start of Two recent multicenter, randomized, intention-to-treat,
the infusion and did not recur during the period from 20 to clinical trials of pediatric benzodiazepine-refractory con-
60 min after the start of treatment. While lorazepam was vulsive status epilepticus, ConSEPT and EcLiPSE, assessed
more effective than phenytoin in patients with overt convul- efficacy of levetiracetam versus phenytoin. In ConSEPT
sive SE, among the 134 patients with a verified diagnosis [77], 233 children aged 3 months to 16 years were ran-
of subtle SE, no statistically significant differences among domly assigned to phenytoin (20 mg/kg, maximum 1000)
the treatments were detected; response rates were consist- or levetiracetam (40 mg/kg, maximum 3000); 14 patients
ently low in this subgroup (phenytoin 7.7%, diazepam + (12%) in each arm had focal onset status epilepticus. Clinical
phenytoin 8.3%, lorazepam 17.9%, phenobarbital 24.2%). seizure cessation measured 5 min after completion of infu-
The rates of success with different ASMs for patients with sion of the trial drug occurred in 68 (60%) patients in the
subtle SE were much lower than those for patients treated phenytoin group and 60 (50%) patients in the levetiracetam
for overt convulsive SE (range 43.6% to 64.9%). Recurrence group (p = 0·16). In this trial, levetiracetam was not superior
of SE during the 12-hour study period was also higher in to phenytoin and no major ASM-related AEs occurred in
the subtle SE group than in the overt convulsive SE group either group. In EcLiPSE [78], 404 children aged 6 months
(20% vs 11%). Mortality within 30 days after SE was much to < 18 years were randomly assigned to phenytoin (20 mg/
higher in patients with subtle SE (65%) compared to overt kg, maximum 2000) or levetiracetam (40 mg/kg, maximum
convulsive SE (27%). An explanation for the difference in 2500); 22 patients (16%) in the phenytoin group and 33
success rate as well as mortality rate could be drawn from patients (22%) in the levetiracetam group had focal clonic
the dramatically different baseline characteristics in these seizures as initial presentation. Cessation of all visible signs
two types of SE (i.e. cardiopulmonary arrest: subtle SE 38% of convulsive activity was observed in 106 (70%) children
vs overt convulsive SE 6%; life-threatening medical condi- within 35 min of randomization in the levetiracetam group
tion: subtle SE 56% vs overt convulsive SE 32%) as well as and in 86 (64%) within 45 min of randomization in the phe-
the median duration of SE at enrollment (subtle SE: 5.8 h vs nytoin group (p = 0.20). In this trial, levetiracetam was not
overt convulsive SE 2.8 h). superior to phenytoin; one patient in the phenytoin group
The more recent Established Status Epilepticus Treat- had serious AEs.
ment Trial (ESETT), was a multicenter, randomized, Apart from the three landmark randomized clinical tri-
double-blind, adaptive clinical trial evaluating the efficacy als in adults and the two recent clinical trials in children
and safety of levetiracetam (60 mg/kg, maximum 4500), mentioned above, of which only one was restricted to non-
phenytoin (20 mg/kg, maximum 1500) and valproate (40 convulsive seizures, there is only low level of evidence for
mg/kg, maximum 3000) in patients with benzodiazepine- treatment with ASMs for NCS and NCSE, mostly derived
refractory convulsive SE [75]. While NCSs and NCSE were from subgroup analyses from larger studies of SE. A few
not specifically included in this trial, it is, nevertheless, fun- recent and pertinent studies for each of the more commonly
damental to discuss these results since these three ASMs used ASMs are summarized in Table 1: only studies with at
are widely used in the treatment of NCSs and NCSE. A least 10 patients (or 10 episodes) with NCSE were included
total of 384 patients, including children aged > 2 years and [79–88].
adults, were enrolled. While inclusion criteria required a In analyzing efficacy and tolerability data for individ-
clinically apparent seizure, it is worth noting that besides the ual ASMs in NCS and NCSE, lacosamide remains one of
more obvious focal or generalized tonic-clonic movements, the better-studied drugs. In addition to the TRENdS trial
and generalized or segmental myoclonus, the trial also discussed above providing class I data, an observational
included patients who had evolved to having more subtle study [80] demonstrated similar efficacy of lacosamide in
754 P. Bravo et al.

NCSE relative to convulsive SE both in terms of seizure For brivaracetam, based on a subgroup analysis of
termination and improvement in EEG epileptiform burden. NCSE (n = 19/43) patients in a retrospective multicenter
In an efficacy and tolerability observational study [81] of study [79], overall responder rate of 53% was noted with
lacosamide for the treatment of NCSs and NCSE in an response within 6 h of a loading dose (median 100 mg).
elderly population (median age 77 years) with post-stroke In a systematic review of brivaracetam in SE, there was
epilepsy, 50% responder rate (n = 7/14) was noted with no a 50% responder rate correlating with higher mean dose
side effects even with an initial loading dose of 400 mg. 3.3 mg/kg versus 1.5 mg/kg. No response was seen below
Cardiac side effects with lacosamide include dose-depend- 1.9 mg/kg and no correlation was seen with plasma level
ent prolongation of PR interval to 1.4 ms (LCM 200 mg/ [90]. Another retrospective study showed similar results
day), 4.4 ms (LCM 400 mg/day), and 6.6 ms (LCM 600 with higher response rate with doses > 1.82 mg/kg [79].
mg/day) as well as first-degree AV block [89].

Table 1  Summary of retrospective case series of ASMs in NCSs and NCSE


ASM Study Total n with Initial dose, Median time Median ASM Response rate Adverse effects (n
n NCS/ median from SE onset trials prior to or%)
NCSE to oral ASM oral ASM
initiation

Brivaracetam Santamarina 43 19 IV 100 mg N/A N/A 53% Somnolence (n = 5)


2019 [79] (25–400 mg) Worsening seizure
(n = 1)
Lacosamide Moreno 53 30 IV 390.6 mg N/A N/A 90% No major AE
Morales
2015 [80]
Belcastro 14 14 IV 400 mg N/A N/A 50% No major AE
2013 [81]
Perampanel Strzelczyk 52 13 PO 6 mg/day 10 days 5 (0–13) 36% Dizziness @ 12 mg/
2019 [82] (2–10 mg/day) day (n = 1) Somno-
lence @ 8 mg/day
(n = 1)
Rohrachter 30 21 Two arms: 2.3 days 4 (2–8) 17% No major AE (ele-
2018 [83] standard dose: (0.5–18.3) 3/16 standard vated liver enzymes
4 mg (2–12) dose without clinical
n 16 2/14 high dose symptoms in 7/30,
High dose: 32 6/7 were also on
mg (16–32) phenytoin)
n 14
Topiramate Fechner 106 17 PO 100 mg 12 days 5 27% Hyperammonemia
2019 [84] (25–400 mg) (35% but most on
concurrent VPA/
PB)
Pancreatitis (0.9%)
Hyperchloremic
acidosis (0.9%)
Madzar 17 12 PO 100 mg (in 29 days (15–66) 11 (7–14) 100% DNP
2016 [85] 6/17, 400 mg)
Pregabalin Swisher 21 21 PO 342 mg/ 1–3 days DNP 52% Dizziness, sedation
2013 [86] day* (n = 2)
Clobazam Madzar 2016 24 16 PO 20 mg/day+ 2 (3–6) days 3 (2–5) 25% DNP
[87]
Sivakumar 17 15 PO 10 mg/day 4 (1–61) days 3 (2–6) 76% Sedation (33%)
2015 [88] PO 20 mg/day+

ASMs antiseizure medications, DNP data not provided, IV intravenous, N/A not applicable, NCSs nonconvulsive seizures, NCSE nonconvulsive
status epilepticus, PO oral
*Average daily dose; + maintenance dose
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 755

Head-to-head comparison of ASMs for treatment of to pharmacotherapy of NCSs and NCSE, as exemplified by
NCSs and NCSE is limited. In a five-year observational the limited number of high-quality clinical trials that address
study [91] comparing IV levetiracetam, lacosamide, val- this topic. In the last decade, a few guidelines in the treat-
proate and phenytoin, no significant difference was seen ment of SE have become available [108–110]. Although
in overall resolution rate of SE; however, in the subgroup these guidelines primarily focus on management of con-
of NCSE episodes (74%, n = 207/277), valproate was vulsive SE, two of them did address a general approach on
significantly more effective than levetiracetam (86% vs the management of NCSE. The European guidelines from
62%, p < 0.002). Of three recent metanalyses compar- 2010 considered NCSE as > 10 min and the Neurocritical
ing levetiracetam and phenytoin for status epilepticus, two Care Society guidelines from 2012 as > 5 min. Despite these
[92, 93] showed no difference in efficacy while one [94] guidelines, how aggressively NCSs and NCSE should be
showed a higher rate of clinical seizure cessation with lev- treated and which ASMs should be used are still topics of
etiracetam (75.2% vs 67.8%, p = 0.003). In a retrospec- significant controversy [111, 112]. As clinicians embark on
tive single-center observational study [95] for treatment of the treatment of NCSs/NCSE with the well imprinted man-
refractory SE episodes, in the subgroup of episodes with tra of primum non nocere (first do no harm) as their com-
NCSE without coma, no difference was noted between lev- pass, their journey often encounters seemingly unnavigable
etiracetam (34%), valproate (34%) and lacosamide (33%) waters with patients suffering from very complex acute and
for cessation of SE. Phenytoin was ineffective; however, chronic neurological and medical conditions; clinicians are
it was used at significantly lower doses than proven effec- left with the decision to treat potentially harmful seizures
tive in other trials. Overall, in terms of efficacy among IV with yet potentially toxic pharmacological options in very
ASMs, newer ASMs such as levetiracetam, brivaracetam sick patients. There is, therefore, a need for a rigorous phar-
and lacosamide are similar to traditional ASMs such as macotherapy workflow in which both benefits and risks are
phenytoin and valproate. always to be balanced.
For enteral ASMs, all data are retrospective with no Considering the limited evidence in the specific phar-
head-to-head studies. Pregabalin is the only ASM studied macotherapy for NCSs/NCSE, and supported by available
specifically for NCSs/NCSE [86]. Data for other enteral guidelines, it is reasonable to differentiate the treatment of
ASMs such as perampanel, topiramate and clobazam have NCSs and NCSE based on three distinct forms:
derived from subgroup analyses of NCSs and NCSE from
retrospective studies on SE. In a retrospective single- 1. NCSE that follows convulsive SE.
center study of NCSs and NCSE [86], 21 critically ill 2. NCSE with coma or stupor.
patients received pregabalin at an average initial dose of 3. NCSE without coma or stupor.
342 mg/day to assess for termination of seizures within
24 h without addition of other ASMs. In this study, over- NCSE after convulsive SE usually warrants rapid and
all response rate was 52% (11 patients), with higher effi- aggressive escalation to IV anesthetic drugs if second-line
cacy in terminating NCSs (9 patients, 82%) than NCSE (2 pharmacotherapy options fail to control seizures. In this
patients, 18%). It must be noted that most studies evaluat- case, a convulsive SE protocol can be followed. NCSE with
ing oral ASMs may not reflect true efficacy and are limited coma (not preceded by convulsive SE), likely requires a judi-
by many factors including smaller studies with electroclin- cious and rapid stepwise pharmacotherapy approach with
ical heterogeneity, concurrent use of other ASMs and IV first- and second-line ASMs using full IV loads and rapid
anesthetic drugs, publication bias, and most importantly, assessment of response; anesthetic levels of medications
long median times to initiation of therapy from onset of should only be attempted after multiple ASM trials have
SE. failed to control seizures and the patient does not demon-
strate any improvement in the level of consciousness. NCSE
4.2 Primum non nocere without coma also requires a rapid and stepwise approach
with ASMs; however, it is unclear if or when use of anes-
There are several reviews available on the treatment of the thetic levels of medications outweigh the risks in this health-
broader spectrum of SE [96–105]. Very detailed descrip- ier population of patients.
tions of mechanisms of action, efficacy, safety and dosing
for multiple ASMs are available in some of these reviews 4.3 Seizure Burden Reduction as the Mainstay
[100–106]. Few specifically explore the treatment of NCSE Target of Pharmacotherapy
[14, 57] and NCSs [56, 106]. It is widely accepted in the
medical community that treatment of convulsive SE should With the premise that every hour matters, parenteral ASMs
include rapid and aggressive escalation of pharmacotherapy should always be attempted first. A list of parenteral ASMs
[107]. However, much less is known regarding the approach with suggested boluses and maintenance doses can be found
756 P. Bravo et al.

in Table 2. While some authors advocate for benzodiaz- can modify the treatment accordingly. A seizure burden of
epines as first-line therapy even for NCSs and NCSE [57], 30% in 12 h may sound significant, and indeed it is, but if
it is unclear at this point if benzodiazepines are superior contrasted with a previous seizure burden of 80% in the prior
to non-benzodiazepine ASMs in NCSs/NCSE as partially 12-h epoch, the management may be to continue the current
demonstrated in the Veterans Affairs trial for the subgroup treatment, especially if the patient’s overall status is stable, if
without overt convulsive SE [8]. After exhausting several there are limitations due to liver/renal failure, and more so if
parenteral options without improvement in the seizure bur- there has been neurological improvement. On the other hand,
den, enteral ASMs and/or general anesthetic drips (in select a burden of 30% when the prior burden was < 10% would
cases only) should be considered (Tables 3, 4). suggest an urgent need for a change in treatment.
In the first several hours, as long as there is no deterio- Counting seizures can also be misleading. In one 12-h
ration in neurological status or development of overt con- epoch, one could encounter 10 seizures, and in the next
vulsive SE, a judicious trial of parenteral ASMs should be epoch only 5, but if the length of seizures is longer on the
implemented. After one ASM has been administered at an second epoch, the seizure burden is unchanged. Therefore,
appropriate loading dose, and possibly a second bolus, with- the clinician and neurophysiologist should make every effort
out any objective seizure burden reduction on cEEG, this to clarify this burden. A patient with 30 seizures in 12 h, but
ASM should be discontinued, followed by initiation of the each lasting 20 s, has a seizure burden of < 2% (10 min out
next best option, based on side effect profile, underlying eti- of 720 min = 0.014) whereas a patient with 10 seizures in
ology when known, such as primary generalized versus focal 12 h but each lasting 10 min will have a seizure burden of
epilepsy, as well as active comorbidities (i.e. liver failure, 14% (100 min out of 720 = 0.14). It is understandable that
renal failure, hypotension, PR prolongation, thrombocyto- an on-call provider that is not well aware of an improvement
penia, etc.). If an ASM lowers seizure burden but some sei- in the overall seizure burden, especially when cross-covering
zure activity persists, the next ASM can be added on while multiple patients, may interpret 30 seizures in the last 12 h
continuing the first. This process can be repeated for a few as a patient that most certainly requires an anesthetic drip,
ASMs over a few hours (not days). When choosing different whereas the same provider with the appropriate information
ASMs, different mechanisms of action [100–106, 113–116] will unlikely proceed with such an aggressive escalation of
should be considered as depicted in Fig. 2. treatment.
An often neglected, yet important, factor to consider is The choice of a specific first ASM is not as important
previously effective ASMs. It is not uncommon for NCSs as is the appropriate dosing and rapid administration. A
and NCSE to recur, especially if the underlying etiology stepwise approach on the selection of different ASMs and
remains active or decompensation of chronic conditions IV anesthetic drugs can be found in Fig. 3. All attempts
occurs. An exhaustive review of the patient’s chart may should be made at loading a patient with a full dose of an
demonstrate that multiple ASMs have been tried in the past ASM. Subsequent dosing can be tailored based on renal or
and that certain ones were more effective than others. This hepatic clearance, ASM blood levels, and development of
would save hours, if not days, of unnecessary trial and error. side effects or AEs. It is important to remember that renal
A medical history review as well as gathering collateral and hepatic failure are irrelevant when loading a medication;
information with the patient’s family or even previous treat- those only affect elimination rates, not how much is needed
ing clinicians may also demonstrate that some patients may to reach a therapeutic amount to start. It is unclear how long
develop NCSs/NCSE after recurrence of specific conditions to wait after loading a medication and what the right rate of
such as systemic infections, electrolyte imbalances, acute on reduction of seizure burden is considered acceptable. It is up
chronic renal failure, hypertensive encephalopathy, hyper- to the clinician’s discretion to take the totality of the clinical
ammonemia from decompensated cirrhosis, hypoglycemia circumstances into consideration to decide which such rate
in insulin-dependent diabetic patients, among others. Such should be targeted on each individual case. Examples of this
conditions should be treated promptly, and early recogni- include patients whose seizures are slowly improving and
tion is key. have multiple acute medical issues but are not intubated.
Accurate reporting of the seizure burden during the Waiting several hours or even days for seizures to gradually
pharmacotherapy of NCSs and NCSE is crucial. Given that reduce in frequency may be less problematic than loading
patients with NCSs and NCSE are usually cared for by a more medications. Another example is a patient with NCSE
multidisciplinary team of providers, it is paramount for all with partially retained consciousness with severe obstruc-
involved to maintain a very close and effective venue of tive sleep apnea or advanced age with risk for aspiration
communication. The seizure burden should be expressed in pneumonia. If the seizure burden is already improving in
an approximate percentage per epoch, i.e. seizure burden % those cases, and more so if the neurological status is improv-
in past 12 h. More importantly, this seizure burden should be ing—even if slowly—it may be preferable to observe further
contrasted with the previous epoch so that treating clinicians
Table 2  Parenteral antiseizure medications used for NCSs and NCSE [128]
IV (non-drip) Dosing Clinically relevant pharma- Approx. half-life Dose adjustment in renal Dose adjustment in hepatic Comments
antiseizure medi- LD = loading dose cokinetic interactions with (h) in non-critically impairment (maintenance impairment (maintenance
cations MD = maintenance dose other antiseizure medica- ill doses) doses)
tions HD = hemodialysis

Brivaracetam LD: 100–400 mg IV ↑ Concentrations of carba- 9 None Consider dose reduction May obtain brivaracetam
MD: 50–600 mg/day mazepine and phenytoin. drug level
divided q12h or q8h
Fosphenytoin LD: 15–20 mg PE/kg IV See phenytoin. Conversion half-life to phenytoin ~15 to 30 min. May be administered IM if
(up to 150 mg/min); max Note: fosphenytoin is dosed in phenytoin equivalents (PE). no IV access (up to 99%
2000 mg. If still seizing, absorption after IM admin-
give additional 5 mg/kg istration).
IV (max 500 mg) Compatible in saline, dex-
MD: Use phenytoin trose, and lactated Ringers
solution. Non-toxic diluent;
↓cutaneous reactions with
extravasation.
May cause hypotension,
arrhythmias.
Obtain peak phenytoin level
(free and total) 2–3 h post
IV dose or 4 h post IM
dose.
Lacosamide LD: 5–10 mg/kg IV over 13 Reduce dose in severe renal Consider dose reduction May prolong PR interval or
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus

5–10 min (max 400 mg). impairment (CrCl < 30 induce tachyarrhythmias,
If still seizing, give an mL/min); max 300 mg/ including atrial fibrillation.
additional 5 mg/kg over 5 day. HD: 50% removed; Max IV push dose is 400 mg
min (max 200 mg IV) lower dose based on CrCl, administered at a rate of 80
MD: 200–600 mg/day divide Q12h and add 50% mg/min.
divided q12h–q6h of am dose to pm dose May obtain lacosamide drug
post HD. CRRT​: lower level.
dose based on CrCl, then
increase total daily dose
by 50% and divide Q8h.
Levetiracetam LD: 40–60 mg/kg IV over 6 Reduce dose based on May cause behavioral distur-
15 min (max 4500 mg). CrCl. HD: 50% removed; bances.
Max IV push dose of 1500 lower dose based on CrCl, May obtain levetiracetam
mg administered at a rate divide Q12h and add 50% STAT drug level.
of 500 mg/min of AM dose to PM dose No added therapeutic benefit
MD: 1500–4500 mg/day post HD. CRRT​: lower when co-administered with
divided q12h–q6h dose based on CrCl, then brivaracetam.
increase total daily dose
by 50% and divide Q6h.
757
Table 2  (continued)
758

IV (non-drip) Dosing Clinically relevant pharma- Approx. half-life Dose adjustment in renal Dose adjustment in hepatic Comments
antiseizure medi- LD = loading dose cokinetic interactions with (h) in non-critically impairment (maintenance impairment (maintenance
cations MD = maintenance dose other antiseizure medica- ill doses) doses)
tions HD = hemodialysis

Lorazepam LD: 0.5–2 mg IV over 2 12 Not applicable. Not applicable Rapid redistribution. IV
min; repeat every 5 min formulation contains 80%
until seizures stop, up to propylene glycol; may
3 doses cause metabolic acidosis.
MD: not applicable Do not administer IM or SC
(IM midazolam preferred if
IV access not available).
Phenobarbital LD: 5–15 mg/kg IV (up to Strong inducer of UGT, 80 Consider dose reduction. Consider dose reduction Prolonged half-life (up to
60 mg/min); max dose CYP 3A4, 2B6, 2C9, HD: give full daily dose 140 h).
1500 mg. If still seizing, 2A6, 1A2. Dose adjust- in evening after hemodi- May cause hypotension.
give an additional 5–10 ments of ASMs including alysis. IV formulation contains 70%
mg/kg phenytoin and valproate propylene glycol; may
MD: 1–3 mg/kg/day given might be necessary. cause metabolic acidosis.
q day or divided q12h or May obtain phenobarbital
q8h drug level.
Phenytoin LD: 15–20 mg/kg IV (up to Induces CYP 1A2, 2B6, 2C, 15 None. Consider dose reduction May cause rash, fever, hypo-
50 mg/min; 25 mg/min in 3A3/4. Generally avoid tension, or arrhythmias.
elderly and patients with use with most CYP3A4 IV formulation contains
preexisting cardiovascular substrates. Coadminis- 40% propylene glycol; may
conditions) tration with valproate cause metabolic acidosis.
MD: 200–600 mg/day displaces phenytoin from Only compatible in saline
divided q12h or q8h protein binding sites. (unlike fosphenytoin).
Induces metabolism of Incompatibilities include
valproate. D5W, potassium, insulin,
heparin, norepinephrine,
cephalosporins, dobu-
tamine.
Severe tissue injury may
occur with extravasation,
including rare purple glove
syndrome.
Obtain peak phenytoin level
(free and total) 2 h post IV
loading dose.
P. Bravo et al.
Table 2  (continued)
IV (non-drip) Dosing Clinically relevant pharma- Approx. half-life Dose adjustment in renal Dose adjustment in hepatic Comments
antiseizure medi- LD = loading dose cokinetic interactions with (h) in non-critically impairment (maintenance impairment (maintenance
cations MD = maintenance dose other antiseizure medica- ill doses) doses)
tions HD = hemodialysis
Valproate LD: 20–40 mg/kg IV (over Phenytoin and valproate 12 None. Caution in hepatic impair- Highly plasma protein
5–10 min); max 4000 mg. may displace each other ment bound (up to 90%). May
If still seizing, give addi- from protein binding sites. cause hyperammonemic
tional 20 mg/kg IV (max Valproate markedly inhib- encephalopathy (treated
2000 mg) over 5 min its lamotrigine metabo- with L-carnitine supple-
MD: 2000–6000 mg lism ↑↑ lamotrigine levels mentation), hepatotoxicity,
divided q8h–q6h and risk of side effects thrombocytopenia, and
including rash. platelet dysfunction.
Concurrent use with carbap-
enems (meropenem, doripe-
nem, imipenem, ertapenem)
may result in markedly
decreased valproic acid
plasma concentrations.
May obtain valproate total
level 2 h post IV loading
dose.
Highly teratogenic and asso-
ciated with other adverse
fetal effects.
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus

Adapted from the Yale New Haven Hospital status epilepticus protocol 2020 with permission of Abdalla Ammar, PharmD; Lawrence J. Hirsch, MD; Emily J. Gilmore, MD; the Yale New
Haven Hospital Department of Pharmacy Services; and the Yale University Dept of Neurology [128]
CRRT​ continuous renal replacement therapy, HD hemodialysis, IM intramuscular, IV intravenous, LD loading dose, MD maintenance dose, NCSs nonconvulsive seizures, NCSE nonconvulsive
status epilepticus, SC subcutaneous
759
760

Table 3  Intravenous anesthetic drugs used for refractory NCSs and NCSE [128]
IV anesthetic drugs Dosing Clinically relevant pharma- Approximate- Dose adjustment in Dose adjustment in Comments
LD = loading dose cokinetic interactions with half life (h) in renal impairment hepatic impairment (main-
MD = maintenance dose other antiseizure medications non-critically (maintenance doses) tenance doses)
ill HD = hemodialysis

Ketamine LD: 1.5 mg/kg IV push over 2.5 None. Consider dose reduction. NMDA antagonist; provides an
3–5 min (max 150 mg); infusion with a different mecha-
repeat until seizures stop; nism of action (non-GABA). May
max total load of 4.5 mg/kg have sympathomimetic properties
MD: initial 1.2 mg/kg/h, range but can also cause hypotension
0.3–7.5 mg/kg/h; titrate to when HR/SBP ≥0.9
seizure suppression. Simultaneous benzodiazepine infu-
sion
recommended for synergism.
May ↑ICP (conflicting evidence).
Midazolam LD: 0.2 mg/kg IV (push over 7 Consider dose Consider dose reduction. Rapid redistribution. Active
1–2 min); max 20 mg Repeat reduction: risk of metabolites. May be administered
0.2–0.4 mg/kg boluses (max active metabolite via alternate routes: 0.2 mg/kg (up
40 mg per bolus) q5min until accumulation. to 10 mg) IM, intranasal, or buccal
seizures stop; max total load routes; all well absorbed rapidly.
of 2 mg/kg
MD: 0.1–2.9 mg/kg/h; titrate to
seizure suppression
Pentobarbital LD: 5 mg/kg IV (up to 50 mg/ 22 None. Consider dose reduction. Prolonged half-life (up to 50 h;
min); max 500 mg. Repeat dose dependent). May cause
until seizures stop; max total hypotension, ileus, myocardial
load of 25 mg/kg suppression, immunosuppression,
MD: 0.5–10 mg/kg/h; titrate to thrombocytopenia. IV formulation
seizure suppression contains 40% propylene glycol;
may cause metabolic acidosis.
P. Bravo et al.
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 761

rather than adding a sedating agent to a patient who could

Adapted from the Yale New Haven Hospital status epilepticus protocol 2020 with permission of Abdalla Ammar, PharmD; Lawrence J. Hirsch, MD; Emily J. Gilmore, MD; the Yale New
nate, triglycerides, creatine kinase,
with hypersensitivity to egg or soy

(> 48 h) or high doses (> 80 mcg/


arrest, rhabdomyolysis, renal fail-
demia, pancreatitis, and propofol
May cause respiratory depression,
end up losing their airway as a result of this intervention.

ure). Contraindicated in patients

products. Monitor pH, bicarbo-


infusion syndrome (metabolic

lipase with prolonged therapy


acidosis, bradycardia, cardiac
hypotension, hypertriglyceri-
As some cases of NCSs and NCSE remain refractory to
multiple trials of different ASMs, it is possible that some

kg/min or 5 mg/kg/h).
of those ASMs may need to be re-introduced or main-
tained, especially those that have demonstrated at least
a modest effect in the seizure burden. At this point, it is
paramount for clinicians to document which ASMs have
Comments

made a more significant impact. Once seizures are con-


trolled, and only after IV drips are successfully weaned
to off for > 24 h and seizure free > 48 h, at that point it
hepatic impairment (main-

is reasonable to consider weaning those ASMs that were


the least effective, especially when multiple ASMs had
Dose adjustment in

to be introduced. Most of the time, this weaning process


requires further intermittent monitoring with cEEG. The
tenance doses)

decision to wean any ASM at this point should always be


contrasted with the risk of further seizures. Occasionally,
None.

it is not possible to wean any ASM in the acute phase, but


it will still be very important for the treating clinicians to
IV intravenous, LD loading dose, MD maintenance dose, NCSs nonconvulsive seizures, NCSE nonconvulsive status epilepticus
(maintenance doses)

document the pattern of success with different ASMs so


HD = hemodialysis
Dose adjustment in
renal impairment

that weaning can be attempted in a rational order as an out-


patient once the underlying acute condition has resolved.
None.

4.4 Refractory SE, Super‑refractory SE, New‑onset


Refractory Status Epilepticus (NORSE)
and Beyond
Approximate-
half life (h) in

0.6 (extended
non-critically

longed use)

Haven Hospital Department of Pharmacy Services; and the Yale University Dept of Neurology [128]
with pro-

These topics have been reviewed in great detail [103,


117–120] including in a recent comprehensive review by the
ill

American Epilepsy Society Treatments Committee [121].


Definitions and available evidence in management of refrac-
other antiseizure medications
Clinically relevant pharma-
cokinetic interactions with

tory SE in such reviews encompass a broad spectrum of SE,


including convulsive SE, and therefore a lengthy discussion
in the topic is out of the scope of this review. Neverthe-
less, as NCSE with coma often evolves into refractory and
super-refractory SE, it is important to discuss a few studies
relevant to the use of IV anesthetic drugs.
In a retrospective study evaluating 171 ICU patients
with SE, excluding post-cardiac arrest patients, 37% were
LD:1–2 mg/kg IV over 5 min;

treated with IV anesthetic drugs. Patients treated with IV


seizures stop up to total LD

(1.8–12 mg/kg/h); titrate to


max 200 mg. Repeat until

anesthetic drugs had more infections and a 2.9-fold rela-


MD: 30–200 mcg/kg/min
MD = maintenance dose

tive risk of death, independent of possible confounders and


seizure suppression
LD = loading dose

without significant effect modification by different grades


of SE severity and etiologies [122]. However, after adjust-
of 10 mg/kg

ing for the refractoriness of seizures, there were no longer


any significant findings. Thus, it is most likely that patients
IV anesthetic drugs Dosing

with more severe seizures simply needed more aggressive


treatment with IV anesthetic drugs, and that these patients
Table 3  (continued)

had worse outcomes. Furthermore, barbiturates were used


to induce an isoelectric EEG in that series, an unusually
aggressive approach [123].
Propofol

In another retrospective study evaluating 467 consecu-


tive adult cases with SE lasting 30 min or longer, 35%
762

Table 4  Enteral antiseizure medications used for refractory NCSs and NCSE [128]
Enteral antisei- Dosing Clinically relevant pharma- Approximate-half life (h) Dose adjustment in renal Dose adjustment in hepatic Comments
zure medica- LD = cokinetic interactions with in non-critically ill impairment (maintenance impairment (maintenance
tions loading dose other antiseizure medica- doses) doses)
MD = maintenance dose tions HD = hemodialysis

Cannabidiol LD: not usually given CYP3A4 and CYP2C19 58 None. Consider dose reduction. Concomitant use of higher
MD: 2.5–20 mg/kg/day substrate. Phenytoin and doses of cannabidiol and
divided q12h other CYP3A4 induc- valproate increases the risk
ers ↓↓levels. Valproic of transaminase elevations
acid and other CYP3A4 and hepatocellular injury.
inhibitors ↑↑ levels. Consider discontinuation
or dose adjustment of can-
nabidiol and/or valproate
if liver enzyme elevations
occur. AST and/or ALT
> 3 times ULN and total
bilirubin > 2 times ULN,
discontinue treatment. Sus-
tained AST and/or ALT >
5 times ULN, discontinue
treatment.
Carbamazepine LD: 400–800 mg Major CYP3A4 substrate; 24 Consider dose reduction in Consider dose reduction: Strong association between
MD: 400–1600 mg/day major CYP2C19/3A4 8 (with prolonged use due severe renal impairment undergoes extensive the risk of develop-
divided q12h inducer. Phenytoin and to auto-induction; 2–4 (CrCl < 10 ml/min): hepatic metabolism. ing Stevens-Johnson
other CYP3A4 induc- weeks) reduce dose by 25%. syndrome/TEN and the
ers ↓↓levels. Valproic presence of HLA-B*1502
acid and other CYP3A4 allele (documented mostly
inhibitors ↑↑ levels. in Asian decent).
Dose-dependent hypona-
tremia; decreased
incidence compared to
oxcarbazepine.
May obtain carbamazepine
drug level.
Clobazam LD: 20–40 mg Felbamate: ↑plasma con- Clobazam: 16, Caution in severe renal Consider dose reduction: Decreased sedation com-
MD: 20–60 mg/day centrations of N-desmethylclobazam: 39 impairment (CrCl < 30 undergoes extensive pared to other benzodiaz-
divided q12h N-desmethylclobazam. ml/min). hepatic metabolism. epines.
May obtain clobazam
and active metabolite
(N-desmethylclobazam)
drug level.
Clonazepam LD: 1–2 mg. Major CYP3A4 sub- 30 Caution in severe renal Consider dose reduction:
MD: 1.5–3 mg/day divided strate. Phenytoin and impairment (CrCl < 30 undergoes extensive
q12h or q8h other CYP3A4 inducers ml/min). hepatic metabolism.
↓↓levels.
P. Bravo et al.
Table 4  (continued)
Enteral antisei- Dosing Clinically relevant pharma- Approximate-half life (h) Dose adjustment in renal Dose adjustment in hepatic Comments
zure medica- LD = cokinetic interactions with in non-critically ill impairment (maintenance impairment (maintenance
tions loading dose other antiseizure medica- doses) doses)
MD = maintenance dose tions HD = hemodialysis

Gabapentin LD: 1200–3600 mg 6 Reduce dose based on None. Occasional peripheral


MD: 2400–4800 mg CrCl. edema.
divided q8h–q6h HD: dose based on CrCl,
administer supplemental
dose post HD.
Oxcarbazepine LD: 600–1200 mg. ↑ concentrations of pheno- 5 Consider 50% dose reduc- ER formulation not recom- Dose-dependent hypona-
MD: 600–2400 mg/day barbital and phenytoin. tion in severe renal mended. tremia; more common in
divided q12h–q6h impairment. HD: IR elderly.
formulations preferred. May obtain oxcarbazepine
drug level.
Perampanel LD: 6–12 mg. 105 Use not recommended in Consider dose reduction in May cause behavioral issues/
MD: 12 mg/day severe renal impairment mild to moderate hepatic agitation.
(CrCl < 30 ml/min). impairment. Use not
recommended in severe
hepatic impairment.
Pregabalin LD: 150–300 mg 6 Reduce dose. None. Occasional peripheral
MD:150–600 mg/day HD: dose based on CrCl, edema.
divided q8h–q6h administer supplemental
dose post HD.
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus

Topiramate LD: 200–400 mg. Use with zonisamide and 21 Reduce dose by ~50%. Consider dose reduction. May cause metabolic acido-
MD: 200–600 (reports up other carbonic anhydrase HD: supplemental dose sis; caution with propofol,
to 1600) mg/day divided inhibitors may worsen may be necessary. acetazolamide, zonisamide
q12h to q6h metabolic acidosis. and metformin. May cause
renal stones. May be asso-
ciated with oligohidrosis,
with risk of hyperther-
mia, mainly in pediatric
patients.
May obtain topiramate drug
level.
763
Table 4  (continued)
764

Enteral antisei- Dosing Clinically relevant pharma- Approximate-half life (h) Dose adjustment in renal Dose adjustment in hepatic Comments
zure medica- LD = cokinetic interactions with in non-critically ill impairment (maintenance impairment (maintenance
tions loading dose other antiseizure medica- doses) doses)
MD = maintenance dose tions HD = hemodialysis
Vigabatrin LD: 1500 mg 10 (but sustained effect for Reduce dose based on None. Can only be ordered by
MD: 1000–3000 mg/day days) CrCl. vigabatrin REMS Program
divided q12h certified prescribers; addi-
tional information can be
found at www.​vigab​atrin​
rems.​com.
Potential progressive
permanent peripheral
vision loss after months
to years of use; regular
ophthalmology examina-
tions recommended with
prolonged use.
May markedly reduce liver
function test (ALT/AST)
in patients with docu-
mented liver disease. It is
not recommended to use
plasma liver function test
activity as an index of liver
cell damage.

Adapted from the Yale New Haven Hospital status epilepticus protocol 2020 with permission of Abdalla Ammar, PharmD; Lawrence J. Hirsch, MD; Emily J. Gilmore, MD; the Yale New
Haven Hospital Department of Pharmacy Services; and the Yale University Dept of Neurology [128]
HD hemodialysis, LD loading dose, MD maintenance dose, NCSs nonconvulsive seizures, NCSE nonconvulsive status epilepticus
P. Bravo et al.
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 765

Fig. 2  Mechanisms of action of most commonly used antiseizure proate have multiple mechanisms of action with varying degrees of
medications and intravenous anesthetic drugs for nonconvulsive sei- potency. Red dots: inhibitory neurotransmitter (GABA); Green dots:
zures and nonconvulsive status epilepticus. *Topiramate and val- excitatory neurotransmitter (glutamate)

developed new disability and 14% died. After accounting institutions (in the USA) and 10% in the other institution
for etiology, severity and comorbidities, therapeutic coma (Switzerland), there was no difference in mortality and
was highly associated with worse outcome at hospital dis- therapeutic coma associated with increased hospital length
charge, including new disability and mortality. This effect of stay [125].
was most marked for patients with milder seizure types Another retrospective study evaluating duration and depth
(complex partial SE rather than generalized convulsive SE of therapeutic coma, predominantly with propofol for refrac-
or nonconvulsive SE in coma); thus, the authors concluded tory SE in 182 patients treated at two academic institutions,
that therapeutic coma should be used cautiously in complex demonstrated that shorter but deeper therapeutic coma for
partial SE but that it was “fully justified” in those with gen- refractory SE may be more effective and safer [126]. Longer
eralized convulsive SE or NCSE in coma [124]. duration of the first trial of IV anesthetic drugs was indepen-
In a “natural” experiment, outcome of refractory SE dently associated with higher chance of seizure recurrence
was compared (retrospectively) in two different countries following the first wean, although without poor functional
where use of therapeutic coma differed. A total of 236 neurological outcome upon discharge or increased mortal-
patients including all etiologies of SE, except post-anoxic, ity. Furthermore, higher doses during the first trial were
were included. Both institutions had similar demograph- independently associated with fewer in-hospital complica-
ics, co-morbidities, SE characteristic and SE severity score. tions and associated with a shorter duration of mechanical
Although therapeutic coma was used in 25% in one of the ventilation and total length of stay. The best cut-off time
766 P. Bravo et al.

Fig. 3  Suggested stepwise approach for treatment of nonconvul- ness/airway status. This choice of ASMs, especially if effective in
sive seizures and nonconvulsive status epilepticus with and with- SE cessation, extends beyond the acute treatment phase; long-term
out coma/stupor. A seizure burden guided algorithm for treatment tolerability based on concurrent co-morbidities must be taken into
of NCSE with categorization of parental ASMs, enteral ASMs, and account (medication choices based on the Yale-New Haven Hospital
IV anesthetic drugs into different tiers based on efficacy and toler- status epilepticus protocol; used with permission of Abdalla Ammar,
ability. The choice of ASM should also be based on mechanism of PharmD; Lawrence J. Hirsch, MD; Emily J. Gilmore, MD; the Yale
action (see Fig. 2), renal versus hepatic metabolism, hemodynamic/ New Haven Hospital Department of Pharmacy Services; and the Yale
hematologic/cardiac status (see Tables 2, 3, 4) and level of conscious- University Dept of Neurology [128]). ASMs antiseizure medications

for duration of IV anesthetic drugs was 35 h (shorter being suppression or isoelectric EEG and was observed in 39.3%
better). of patients; 62.3% of these underwent > 48-h induced coma.
Finally, long-term prognosis related to deep sedation The remainder of the patients (60.7%) had seizure suppres-
with IV anesthetic drugs was further evaluated in a recent sion pattern on EEG, defined as all other EEG patterns with-
longitudinal observational study evaluating 61 patients with out evidence of seizure activity and not meeting criteria for
refractory SE [127]. Deep sedation was defined as burst deep sedation. Multiple regression analysis demonstrated
that deep sedation was associated with worse long-term
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 767

prognosis (1 and 2 years, p < 0.05) including lower survival References


(p < 0.05).
In summary, there is a lack of randomized clinical trials 1. Trinka E, Cock H, Hesdorffer D, Rossetti AO, Scheffer IE, Shin-
in the use of IV anesthetic drugs for NCSE/NCSs. Their use nar S, et al. A definition and classification of status epilepti-
cus—report of the ILAE Task Force on Classification of Status
should be cautious and likely tailored towards decreasing/ Epilepticus. Epilepsia. 2015;56(10):1515–23.
suppressing seizure burden rather than suppressing brain 2. Meldrum BS, Horton RW. Physiology of status epilepticus in
activity. primates. Arch Neurol. 1973;28(1):1–9.
3. Meldrum BS, Brierley JB. Prolonged epileptic seizures in pri-
mates. Ischemic cell change and its relation to ictal physiological
events. Arch Neurol. 1973;28(1):10–7.
5 Future Directions 4. Meldrum BS, Vigouroux RA, Brierley JB. Systemic factors and
epileptic brain damage. Prolonged seizures in paralyzed, artifi-
cially ventilated baboons. Arch Neurol. 1973;29(2):82–7.
Based on both animal and human studies, it is evident that 5. Holmes GL, Thompson JL, Marchi T, Feldman DS. Behavioral
NCSs and NCSE are associated with worse outcomes, espe- effects of kainic acid administration on the immature brain. Epi-
cially when prolonged or frequent (high seizure burden), lepsia. 1988;29(6):721–30.
6. Milgram NW, Isen DA, Mandel D, Palantzas H, Pepkowski MJ.
including long-term outcomes such as function, cogni- Deficits in spontaneous behavior and cognitive function follow-
tion, and epilepsy. However, prospective randomized trials ing systemic administration of kainic acid. Neurotoxicology.
addressing the relationship between appropriate pharma- 1988;9(4):611–24.
cotherapy and improvement in outcome are still lacking. 7. Treatment of Convulsive Status Epilepticus. Recommendations
of the Epilepsy Foundation of America’s Working Group on sta-
Earlier and faster diagnosis and treatment lead to more tus epilepticus. JAMA. 1993;270(7):854–9.
rapid cessation of seizures and likely better outcomes. The 8. Treiman DM, Meyers PD, Walton NY, Collins JF, Colling C,
substantial heterogeneity in the different forms of NCSE as Rowan AJ, et al. A comparison of four treatments for generalized
well as all the multiple underlying etiologies will continue to convulsive status epilepticus. Veterans Affairs Status Epilepticus
Cooperative Study Group. N Engl J Med. 1998;339(12):792–8.
challenge the development of appropriate studies. With the 9. DeLorenzo RJ, Garnett LK, Towne AR, Waterhouse EJ, Boggs
available evidence, clinicians are urged to continue to main- JG, Morton L, et al. Comparison of status epilepticus with pro-
tain a high level of suspicion for NCSs and NCSE in acutely longed seizure episodes lasting from 10 to 29 minutes. Epilepsia.
ill neurological and medical patients, identify promptly, treat 1999;40(2):164–9.
10. Shinnar S, Berg AT, Moshe SL, Shinnar R. How long do new-
quickly with full loads of medications, monitor appropri- onset seizures in children last? Ann Neurol. 2001;49(5):659–64.
ately, and continue to treat wisely, always balancing benefits 11. Logroscino G, Hesdorffer DC, Cascino GD, Annegers JF, Bag-
and risks of aggressive therapy. iella E, Hauser WA. Long-term mortality after a first episode of
status epilepticus. Neurology. 2002;58(4):537–41.
12. Husain AM, Horn GJ, Jacobson MP. Non-convulsive status epi-
Declarations lepticus: usefulness of clinical features in selecting patients for
urgent EEG. J Neurol Neurosurg Psychiatry. 2003;74(2):189–91.
Funding None. 13. Kaplan PW. The clinical features, diagnosis, and prog-
nosis of nonconvulsive status epilepticus. Neurologist.
Conflict of interest/competing interests Dr. Bravo and Dr. Vad- 2005;11(6):348–61.
diparti have no conflicts of interest/competing interests. Dr. Hirsch 14. Meierkord H, Holtkamp M. Non-convulsive status epilepti-
has received consultation fees for advising from Accure, Aquestive, cus in adults: clinical forms and treatment. Lancet Neurol.
Ceribell, Marinus, Medtronic, Neuropace and UCB; royalties from 2007;6(4):329–39.
Wolters-Kluwer for authoring chapters for UpToDate-Neurology, and 15. Pinto LF, Gilmore EJ, Petroff OA, Sivaraju A, Rampal N,
from Wiley for co-authoring the book “Atlas of EEG in Critical Care”, Hirsch LJ, et al. Cyclic seizures in critically ill patients: Clini-
by Hirsch and Brenner; and honoraria for speaking from Neuropace cal correlates, DC recordings and outcomes. Clin Neurophysiol.
and Natus. 2017;128(6):1083–90.
16. DeLorenzo RJ, Waterhouse EJ, Towne AR, Boggs JG, Ko D,
Ethics approval Not applicable. DeLorenzo GA, et al. Persistent nonconvulsive status epilepti-
cus after the control of convulsive status epilepticus. Epilepsia.
Consent to participate Not applicable. 1998;39(8):833–40.
17. DeLorenzo RJ, Hauser WA, Towne AR, Boggs JG, Pellock JM,
Availability of data and material Not applicable. Penberthy L, et al. A prospective, population-based epidemio-
logic study of status epilepticus in Richmond, Virginia. Neurol-
Author contributions Dr. PB contributed to the draft of the entire ogy. 1996;46(4):1029–35.
manuscript and design of all figures; Dr. AV contributed to the draft of 18. Hesdorffer DC, Logroscino G, Cascino G, Annegers JF, Hauser
Sect. 4.1 (Evidence on pharmacotherapy for NCSs and NCSE) includ- WA. Incidence of status epilepticus in Rochester, Minnesota,
ing Table 1 and design of Fig. 3; Dr. LJH contributed to the draft and 1965–1984. Neurology. 1998;50(3):735–41.
revision of the entire manuscript as well as design of all figures and 19. Towne AR, Waterhouse EJ, Boggs JG, Garnett LK, Brown AJ,
tables. All authors have reviewed and read the final version of this Smith JR Jr, et al. Prevalence of nonconvulsive status epilepticus
manuscript. in comatose patients. Neurology. 2000;54(2):340–5.
768 P. Bravo et al.

20. Logroscino G, Hesdorffer DC, Cascino G, Annegers JF, Hauser 40. Vespa PM, Miller C, McArthur D, Eliseo M, Etchepare M,
WA. Time trends in incidence, mortality, and case-fatality after Hirt D, et al. Nonconvulsive electrographic seizures after
first episode of status epilepticus. Epilepsia. 2001;42(8):1031–5. traumatic brain injury result in a delayed, prolonged increase
21. Logroscino G, Hesdorffer DC, Cascino G, Hauser WA, Coey- in intracranial pressure and metabolic crisis. Crit Care Med.
taux A, Galobardes B, et al. Mortality after a first episode of 2007;35(12):2830–6.
status epilepticus in the United States and Europe. Epilepsia. 41. Vespa PM, McArthur DL, Xu Y, Eliseo M, Etchepare M,
2005;46(Suppl 11):46–8. Dinov I, et al. Nonconvulsive seizures after traumatic brain
22. Dham BS, Hunter K, Rincon F. The epidemiology of status epi- injury are associated with hippocampal atrophy. Neurology.
lepticus in the United States. Neurocrit Care. 2014;20(3):476–83. 2010;75(9):792–8.
23. Gilmore EJ, Hirsch LJ. Epilepsy: Status epilepticus epi- 42. Abend NS, Arndt DH, Carpenter JL, Chapman KE, Cornett KM,
demiology–tracking a moving target. Nat Rev Neurol. Gallentine WB, et al. Electrographic seizures in pediatric ICU
2015;11(7):377–8. patients: cohort study of risk factors and mortality. Neurology.
24. Bergin PS, Brockington A, Jayabal J, Scott S, Litchfield R, Rob- 2013;81(4):383–91.
erts L, et al. Status epilepticus in Auckland, New Zealand: Inci- 43. Payne ET, Zhao XY, Frndova H, McBain K, Sharma R, Hutch-
dence, etiology, and outcomes. Epilepsia. 2019;60(8):1552–64. ison JS, et al. Seizure burden is independently associated with
25. Penberthy LT, Towne A, Garnett LK, Perlin JB, DeLorenzo short term outcome in critically ill children. Brain. 2014;137(Pt
RJ. Estimating the economic burden of status epilepticus to the 5):1429–38.
health care system. Seizure. 2005;14(1):46–51. 44. Wagenman KL, Blake TP, Sanchez SM, Schultheis MT, Rad-
26. Strzelczyk A, Knake S, Oertel WH, Rosenow F, Hamer HM. cliffe J, Berg RA, et al. Electrographic status epilepticus
Inpatient treatment costs of status epilepticus in adults in Ger- and long-term outcome in critically ill children. Neurology.
many. Seizure. 2013;22(10):882–5. 2014;82(5):396–404.
27. Liu Z, Gatt A, Werner SJ, Mikati MA, Holmes GL. Long-term 45. De Marchis GM, Pugin D, Meyers E, Velasquez A, Suwatcha-
behavioral deficits following pilocarpine seizures in immature rangkoon S, Park S, et al. Seizure burden in subarachnoid hemor-
rats. Epilepsy Res. 1994;19(3):191–204. rhage associated with functional and cognitive outcome. Neurol-
28. Krsek P, Mikulecka A, Druga R, Kubova H, Hlinak Z, ogy. 2016;86(3):253–60.
Suchomelova L, et al. Long-term behavioral and morphological 46. Vespa P, Tubi M, Claassen J, Buitrago-Blanco M, McArthur
consequences of nonconvulsive status epilepticus in rats. Epi- D, Velazquez AG, et al. Metabolic crisis occurs with sei-
lepsy Behav. 2004;5(2):180–91. zures and periodic discharges after brain trauma. Ann Neurol.
29. Turski L, Cavalheiro EA, Czuczwar SJ, Turski WA, Kleinrok 2016;79(4):579–90.
Z. The seizures induced by pilocarpine: behavioral, electroen- 47. Witsch J, Frey HP, Schmidt JM, Velazquez A, Falo CM, Reznik
cephalographic and neuropathological studies in rodents. Pol J M, et al. Electroencephalographic periodic discharges and fre-
Pharmacol Pharm. 1987;39(5):545–55. quency-dependent brain tissue hypoxia in acute brain injury.
30. Mello LE, Cavalheiro EA, Tan AM, Kupfer WR, Pretorius JK, JAMA Neurol. 2017;74(3):301–9.
Babb TL, et al. Circuit mechanisms of seizures in the pilocarpine 48. Zafar SF, Postma EN, Biswal S, Boyle EJ, Bechek S, O’Connor
model of chronic epilepsy: cell loss and mossy fiber sprouting. K, et al. Effect of epileptiform abnormality burden on neurologic
Epilepsia. 1993;34(6):985–95. outcome and antiepileptic drug management after subarachnoid
31. Nevander G, Ingvar M, Auer R, Siesjo BK. Status epilepticus hemorrhage. Clin Neurophysiol. 2018;129(11):2219–27.
in well-oxygenated rats causes neuronal necrosis. Ann Neurol. 49. Tabaeizadeh M, Aboul Nour H, Shoukat M, Sun H, Jin J, Javed
1985;18(3):281–90. F, et al. Burden of epileptiform activity predicts discharge neuro-
32. Williams AJ, Tortella FC, Lu XM, Moreton JE, Hartings JA. logic outcomes in severe acute ischemic stroke. Neurocrit Care.
Antiepileptic drug treatment of nonconvulsive seizures induced 2020;32(3):697–706.
by experimental focal brain ischemia. J Pharmacol Exp Ther. 50. Kapur J, Macdonald RL. Rapid seizure-induced reduction of ben-
2004;311(1):220–7. zodiazepine and Z ­ n2+ sensitivity of hippocampal dentate granule
33. Avdic U, Ahl M, Chugh D, Ali I, Chary K, Sierra A, et al. Non- cell GABAA receptors. J Neurosci. 1997;17(19):7532–40.
convulsive status epilepticus in rats leads to brain pathology. 51. Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich
Epilepsia. 2018;59(5):945–58. S, et al. A comparison of lorazepam, diazepam, and placebo for
34. Aminoff MJ. Do nonconvulsive seizures damage the brain?—No. the treatment of out-of-hospital status epilepticus. N Engl J Med.
Arch Neurol. 1998;55(1):119–20. 2001;345(9):631–7.
35. Young GB, Jordan KG. Do nonconvulsive seizures damage the 52. Hillman J, Lehtimaki K, Peltola J, Liimatainen S. Clinical signifi-
brain?—Yes. Arch Neurol. 1998;55(1):117–9. cance of treatment delay in status epilepticus. Int J Emerg Med.
36. Young GB, Jordan KG, Doig GS. An assessment of nonconvul- 2013;6(1):6.
sive seizures in the intensive care unit using continuous EEG 53. Williams RP, Banwell B, Berg RA, Dlugos DJ, Donnelly M,
monitoring: an investigation of variables associated with mortal- Ichord R, et al. Impact of an ICU EEG monitoring pathway on
ity. Neurology. 1996;47(1):83–9. timeliness of therapeutic intervention and electrographic seizure
37. Waterhouse EJ, Vaughan JK, Barnes TY, Boggs JG, Towne termination. Epilepsia. 2016;57(5):786–95.
AR, Kopec-Garnett L, et al. Synergistic effect of status epi- 54. Sanchez Fernandez I, Sansevere AJ, Guerriero RM, Buraniqi
lepticus and ischemic brain injury on mortality. Epilepsy Res. E, Pearl PL, Tasker RC, et al. Time to electroencephalography
1998;29(3):175–83. is independently associated with outcome in critically ill neo-
38. DeGiorgio CM, Heck CN, Rabinowicz AL, Gott PS, Smith T, nates and children. Epilepsia. 2017;58(3):420–8.
Correale J. Serum neuron-specific enolase in the major subtypes 55. Claassen J, Mayer SA, Kowalski RG, Emerson RG, Hirsch
of status epilepticus. Neurology. 1999;52(4):746–9. LJ. Detection of electrographic seizures with continu-
39. Vespa PM, O’Phelan K, Shah M, Mirabelli J, Starkman S, Kid- ous EEG monitoring in critically ill patients. Neurology.
well C, et al. Acute seizures after intracerebral hemorrhage: a 2004;62(10):1743–8.
factor in progressive midline shift and outcome. Neurology. 56. Jirsch J, Hirsch LJ. Nonconvulsive seizures: developing a rational
2003;60(9):1441–6. approach to the diagnosis and management in the critically ill
population. Clin Neurophysiol. 2007;118(8):1660–70.
Pharmacotherapy for Nonconvulsive Seizures and Nonconvulsive Status Epilepticus 769

57. Kinney MO, Craig JJ, Kaplan PW. Hidden in plain sight: non- 74. Husain AM, Lee JW, Kolls BJ, Hirsch LJ, Halford JJ, Gupta PK,
convulsive status epilepticus—recognition and management. et al. Randomized trial of lacosamide versus fosphenytoin for
Acta Neurol Scand. 2017;136(4):280–92. nonconvulsive seizures. Ann Neurol. 2018;83(6):1174–85.
58. Herman ST, Abend NS, Bleck TP, Chapman KE, Drislane FW, 75. Kapur J, Elm J, Chamberlain JM, Barsan W, Cloyd J, Lowenstein
Emerson RG, et al. Consensus statement on continuous EEG in D, et al. Randomized trial of three anticonvulsant medications
critically ill adults and children, part I: indications. J Clin Neu- for status epilepticus. N Engl J Med. 2019;381(22):2103–13.
rophysiol. 2015;32(2):87–95. 76. Chamberlain JM, Kapur J, Shinnar S, Elm J, Holsti M, Babcock
59. Struck AF, Ustun B, Ruiz AR, Lee JW, LaRoche SM, Hirsch LJ, L, et al. Efficacy of levetiracetam, fosphenytoin, and valproate for
et al. Association of an electroencephalography-based risk score established status epilepticus by age group (ESETT): a double-
with seizure probability in hospitalized patients. JAMA Neurol. blind, responsive-adaptive, randomised controlled trial. Lancet.
2017;74(12):1419–24. 2020;395(10231):1217–24.
60. Struck AF, Osman G, Rampal N, Biswal S, Legros B, Hirsch 77. Dalziel SR, Borland ML, Furyk J, Bonisch M, Neutze J, Donath
LJ, et al. Time-dependent risk of seizures in critically ill S, et al. Levetiracetam versus phenytoin for second-line treat-
patients on continuous electroencephalogram. Ann Neurol. ment of convulsive status epilepticus in children (ConSEPT):
2017;82(2):177–85. an open-label, multicentre, randomised controlled trial. Lancet.
61. Struck AF, Tabaeizadeh M, Schmitt SE, Ruiz AR, Swisher 2019;393(10186):2135–45.
CB, Subramaniam T, et al. Assessment of the validity of the 78. Lyttle MD, Rainford NEA, Gamble C, Messahel S, Hum-
2HELPS2B score for inpatient seizure risk prediction. JAMA phreys A, Hickey H, et al. Levetiracetam versus phenytoin for
Neurol. 2020;77(4):500–7. second-line treatment of paediatric convulsive status epilepticus
62. Hirsch LJ, LaRoche SM, Gaspard N, Gerard E, Svoronos A, (EcLiPSE): a multicentre, open-label, randomised trial. Lancet.
Herman ST, et al. American Clinical Neurophysiology Society’s 2019;393(10186):2125–34.
standardized critical care EEG terminology: 2012 version. J Clin 79. Santamarina E, Parejo Carbonell B, Sala J, Gutiérrez-Viedma
Neurophysiol. 2013;30(1):1–27. Á, Miró J, Asensio M, et al. Use of intravenous brivar-
63. Yoo JY, Rampal N, Petroff OA, Hirsch LJ, Gaspard N. Brief acetam in status epilepticus: a multicenter registry. Epilepsia.
potentially ictal rhythmic discharges in critically ill adults. JAMA 2019;60(8):1593–601.
Neurol. 2014;71(4):454–62. 80. Moreno Morales EY, Fernandez Peleteiro M, Bondy Peña EC,
64. Gaspard N, Hirsch LJ, LaRoche SM, Hahn CD, Westover MB, Domínguez Lorenzo JM, Pardellas Santiago E, Fernández A.
Critical Care EEGMRC. Interrater agreement for Critical Care Observational study of intravenous lacosamide in patients with
EEG Terminology. Epilepsia. 2014;55(9):1366–73. convulsive versus non-convulsive status epilepticus. Clin Drug
65. Rodriguez Ruiz A, Vlachy J, Lee JW, Gilmore EJ, Ayer T, Haider Investig. 2015;35(7):463–9.
HA, et al. Association of periodic and rhythmic electroencepha- 81. Belcastro V, Vidale S, Pierguidi L, Sironi L, Tancredi L, Striano
lographic patterns with seizures in critically ill patients. JAMA P, et al. Intravenous lacosamide as treatment option in post-stroke
Neurol. 2017;74(2):181–8. non convulsive status epilepticus in the elderly: a proof-of-con-
66. Beniczky S, Hirsch LJ, Kaplan PW, Pressler R, Bauer G, Aurlien cept, observational study. Seizure. 2013;22(10):905–7.
H, et al. Unified EEG terminology and criteria for nonconvulsive 82. Strzelczyk A, Knake S, Kalviainen R, Santamarina E, Toledo
status epilepticus. Epilepsia. 2013;54(Suppl 6):28–9. M, Willig S, et al. Perampanel for treatment of status epilepticus
67. Leitinger M, Trinka E, Gardella E, Rohracher A, Kalss G, in Austria, Finland, Germany, and Spain. Acta Neurol Scand.
Qerama E, et al. Diagnostic accuracy of the Salzburg EEG cri- 2019;139(4):369–76.
teria for non-convulsive status epilepticus: a retrospective study. 83. Rohracher A, Kalss G, Neuray C, Höfler J, Dobesberger J,
Lancet Neurol. 2016;15(10):1054–62. Kuchukhidze G, et al. Perampanel in patients with refractory
68. Hirsch LJ, Fong MWK, Leitinger M, LaRoche SM, Beniczky S, and super-refractory status epilepticus in a neurological inten-
Abend NS, et al. American Clinical Neurophysiology Society’s sive care unit: a single-center audit of 30 patients. Epilepsia.
standardized critical care EEG terminology: 2021 version. J Clin 2018;59(Suppl 2):234–42.
Neurophysiol. 2021;38(1):1–29. 84. Fechner A, Hubert K, Jahnke K, Knake S, Konczalla J, Menzler
69. Moffet EW, Subramaniam T, Hirsch LJ, Gilmore EJ, Lee JW, K, et al. Treatment of refractory and superrefractory status epi-
Rodriguez-Ruiz AA, et al. Validation of the 2HELPS2B sei- lepticus with topiramate: a cohort study of 106 patients and a
zure risk score in acute brain injury patients. Neurocrit Care. review of the literature. Epilepsia. 2019;60(12):2448–58.
2020;33(3):701–7. 85. Madžar D, Kuramatsu JB, Gerner ST, Huttner HB. Assessing
70. Limotai C, Ingsathit A, Thadanipon K, McEvoy M, Attia J, the value of topiramate in refractory status epilepticus. Seizure.
Thakkinstian A. How and whom to monitor for seizures in an 2016;38:7–10.
ICU: a systematic review and meta-analysis. Crit Care Med. 86. Swisher CB, Doreswamy M, Husain AM. Use of pregabalin for
2019;47(4):e366–73. nonconvulsive seizures and nonconvulsive status epilepticus.
71. Rossetti AO, Schindler K, Sutter R, Ruegg S, Zubler F, Novy Seizure. 2013;22(2):116–8.
J, et al. Continuous vs routine electroencephalogram in criti- 87. Madžar D, Geyer A, Knappe RU, Gollwitzer S, Kuramatsu JB,
cally ill adults with altered consciousness and no recent sei- Gerner ST, et al. Effects of clobazam for treatment of refractory
zure: a multicenter randomized clinical trial. JAMA Neurol. status epilepticus. BMC Neurol. 2016;16(1):202.
2020;77(10):1225–32. 88. Sivakumar S, Ibrahim M, Parker D Jr, Norris G, Shah A,
72. Hill CE, Blank LJ, Thibault D, Davis KA, Dahodwala N, Litt Mohamed W. Clobazam: an effective add-on therapy in refrac-
B, et al. Continuous EEG is associated with favorable hos- tory status epilepticus. Epilepsia. 2015;56(6):e83–9.
pitalization outcomes for critically ill patients. Neurology. 89. Rudd GD, Haverkamp W, Mason JW, Wenger T, Jay G,
2019;92(1):e9–18. Hebert D, et al. Lacosamide cardiac safety: clinical trials
73. Ney JP, van der Goes DN, Nuwer MR, Nelson L, Eccher in patients with partial-onset seizures. Acta Neurol Scand.
MA. Continuous and routine EEG in intensive care: utiliza- 2015;132(5):355–63.
tion and outcomes, United States 2005–2009. Neurology. 90. Aicua-Rapun I, André P, Rossetti AO, Decosterd LA, Buc-
2013;81(23):2002–8. lin T, Novy J. Intravenous brivaracetam in status epilepticus:
770 P. Bravo et al.

correlation between loading dose, plasma levels and clinical critically ill patients: a survey of neurologists. Neurocrit Care.
response. Epilepsy Res. 2019;149:88–91. 2010;12(3):382–9.
91. Orlandi N, Giovannini G, Rossi J, Cioclu MC, Meletti S. Clini- 112. Rossetti AO, Hirsch LJ, Drislane FW. Nonconvulsive seizures
cal outcomes and treatments effectiveness in status epilepticus and nonconvulsive status epilepticus in the neuro ICU should or
resolved by antiepileptic drugs: a five-year observational study. should not be treated aggressively: a debate. Clin Neurophysiol
Epilepsia Open. 2020;5(2):166–75. Pract. 2019;4:170–7.
92. Li L, Zhang Y, Jia L, Jia D, Faramand A, Chong W, et al. Leveti- 113. Rogawski MA, Loscher W. The neurobiology of antiepileptic
racetam versus phenytoin for the treatment of established status drugs. Nat Rev Neurosci. 2004;5(7):553–64.
epilepticus: A systematic review and meta-analysis of rand- 114. White HS, Smith MD, Wilcox KS. Mechanisms of action of
omized controlled trials. Seizure. 2020;78:43–8. antiepileptic drugs. Int Rev Neurobiol. 2007;81:85–110.
93. DeMott JM, Slocum GW, Gottlieb M, Peksa GD. Leveti- 115. Morano A, Iannone L, Palleria C, Fanella M, Giallonardo AT, De
racetam vs. phenytoin as 2nd-line treatment for status epilep- Sarro G, et al. Pharmacology of new and developing intravenous
ticus: A systematic review and meta-analysis. Epilepsy Behav. therapies for the management of seizures and epilepsy. Expert
2020;111:107286. Opin Pharmacother. 2019;20(1):25–39.
94. Xue T, Wei L, Shen X, Wang Z, Chen Z, Wang Z. Leveti- 116. Abou-Khalil BW. Update on antiepileptic drugs 2019. Continuum
racetam versus phenytoin for the pharmacotherapy of benzodi- (Minneap Minn). 2019;25(2):508–36.
azepine-refractory status epilepticus: a systematic review and 117. Gaspard N, Foreman BP, Alvarez V, Cabrera Kang C, Probasco
meta-analysis of randomized controlled trials. CNS Drugs. JC, Jongeling AC, et al. New-onset refractory status epilep-
2020;34:1205–15. ticus: etiology, clinical features, and outcome. Neurology.
95. Redecker J, Wittstock M, Rösche J. The efficacy of different 2015;85(18):1604–13.
kinds of intravenously applied antiepileptic drugs in the treat- 118. Hirsch LJ, Gaspard N, van Baalen A, Nabbout R, Demeret S,
ment of status epilepticus. How can it be determined? Epilepsy Loddenkemper T, et al. Proposed consensus definitions for new-
Behav. 2017;71(Pt A):35–8. onset refractory status epilepticus (NORSE), febrile infection-
96. Foreman B, Hirsch LJ. Epilepsy emergencies: diagnosis and related epilepsy syndrome (FIRES), and related conditions.
management. Neurol Clin. 2012;30(1):11–41, vii. Epilepsia. 2018;59(4):739–44.
97. Hirsch LJ, Gaspard N. Status epilepticus. Continuum (Minneap 119. Gaspard N, Hirsch LJ, Sculier C, Loddenkemper T, van
Minn). 2013;19(3 Epilepsy):767–94. Baalen A, Lancrenon J, et al. New-onset refractory status epi-
98. Abend NS, Bearden D, Helbig I, McGuire J, Narula S, Panzer JA, lepticus (NORSE) and febrile infection-related epilepsy syn-
et al. Status epilepticus and refractory status epilepticus manage- drome (FIRES): state of the art and perspectives. Epilepsia.
ment. Semin Pediatr Neurol. 2014;21(4):263–74. 2018;59(4):745–52.
99. Betjemann JP, Lowenstein DH. Status epilepticus in adults. Lan- 120. Gainza-Lein M, Sanchez Fernandez I, Jackson M, Abend NS,
cet Neurol. 2015;14(6):615–24. Arya R, Brenton JN, et al. Association of time to treatment with
100. Trinka E, Hofler J, Leitinger M, Brigo F. Pharmacotherapy for short-term outcomes for pediatric patients with refractory con-
status epilepticus. Drugs. 2015;75(13):1499–521. vulsive status epilepticus. JAMA Neurol. 2018;75(4):410–8.
101. Grover EH, Nazzal Y, Hirsch LJ. Treatment of convulsive status 121. Vossler DG, Bainbridge JL, Boggs JG, Novotny EJ, Lodden-
epilepticus. Curr Treat Options Neurol. 2016;18(3):11. kemper T, Faught E, et al. Treatment of Refractory Convulsive
102. Falco-Walter JJ, Bleck T. Treatment of established status epilep- Status Epilepticus: A Comprehensive Review by the Ameri-
ticus. J Clin Med. 2016;5(5):49:1–8. can Epilepsy Society Treatments Committee. Epilepsy Curr.
103. Holtkamp M. Pharmacotherapy for refractory and super-refrac- 2020;20(5):245–264
tory status epilepticus in adults. Drugs. 2018;78(3):307–26. 122. Sutter R, Marsch S, Fuhr P, Kaplan PW, Ruegg S. Anesthetic
104. Farrokh S, Bon J, Erdman M, Tesoro E. Use of newer anticon- drugs in status epilepticus: risk or rescue? A 6-year cohort study.
vulsants for the treatment of status epilepticus. Pharmacotherapy. Neurology. 2014;82(8):656–64.
2019;39(3):297–316. 123. Hirsch LJ. Finding the lesser of two evils: treating refractory
105. Singh A, Stredny CM, Loddenkemper T. Pharmacother- status epilepticus. Epilepsy Curr. 2015;15(6):313–6.
apy for pediatric convulsive status epilepticus. CNS Drugs. 124. Marchi NA, Novy J, Faouzi M, Stahli C, Burnand B, Rossetti
2020;34(1):47–63. AO. Status epilepticus: impact of therapeutic coma on outcome.
106. Wasim M, Husain AM. Nonconvulsive seizure control in the Crit Care Med. 2015;43(5):1003–9.
intensive care unit. Curr Treat Options Neurol. 2015;17(3):340. 125. Alvarez V, Lee JW, Westover MB, Drislane FW, Novy J,
107. Riviello JJ Jr, Claassen J, LaRoche SM, Sperling MR, Alldredge Faouzi M, et al. Therapeutic coma for status epilepticus: dif-
B, Bleck TP, et al. Treatment of status epilepticus: an interna- fering practices in a prospective multicenter study. Neurology.
tional survey of experts. Neurocrit Care. 2013;18(2):193–200. 2016;87(16):1650–9.
108. Meierkord H, Boon P, Engelsen B, Gocke K, Shorvon S, Tinuper 126. Muhlhofer WG, Layfield S, Lowenstein D, Lin CP, Johnson
P, et al. EFNS guideline on the management of status epilepticus RD, Saini S, et al. Duration of therapeutic coma and outcome of
in adults. Eur J Neurol. 2010;17(3):348–55. refractory status epilepticus. Epilepsia. 2019;60(5):921–34.
109. Brophy GM, Bell R, Claassen J, Alldredge B, Bleck TP, Glauser 127. Caronna E, Vilaseca A, Maria Gracia Gozalo R, Sanchez Cor-
T, et al. Guidelines for the evaluation and management of status ral A, Santafe M, Sueiras M, et al. Long-term prognosis related
epilepticus. Neurocrit Care. 2012;17(1):3–23. to deep sedation in refractory status Epilepticus. Acta Neurol
110. Glauser T, Shinnar S, Gloss D, Alldredge B, Arya R, Bainbridge Scand. 2020;142(6):555–62.
J, et al. Evidence-based guideline: treatment of convulsive sta- 128. Yale New Haven Hospital Treatment Algorithm for Generalized
tus epilepticus in children and adults: report of the guideline Convulsive Status Epilepticus in adults: NORSE Institute.; 2020.
Committee of the American Epilepsy Society. Epilepsy Curr. http://w
​ ww.n​ orsei​ nstit​ ute.o​ rg/d​ efini​ tions. Accessed 28 Feb 2021.
2016;16(1):48–61.
111. Abend NS, Dlugos DJ, Hahn CD, Hirsch LJ, Herman ST. Use of
EEG monitoring and management of non-convulsive seizures in

You might also like