Skip to main content
Erschienen in: Zeitschrift für Epileptologie 4/2022

Open Access 10.11.2022 | Leitthema

Management and prognosis of pediatric status epilepticus

verfasst von: Eroshini Swarnalingam, Kristine Woodward, Micheal Esser, Julia Jacobs

Erschienen in: Clinical Epileptology | Ausgabe 4/2022

Abstract

Background

Pediatric status epilepticus is a neurological emergency with the potential for severe developmental and neurological consequences. Prompt diagnosis and management are necessary.

Objectives

To outline the existing best available evidence for managing pediatric and neonatal status epilepticus, in the light of emerging randomized controlled studies. We also focus on short and long-term prognoses.

Materials and methods

This is a systematic overview of the existing literature.

Results

Status epilepticus, its treatment, and prognosis are usually based on the continuation of seizure activity at 5 and 30 min. Refractory and super-refractory status epilepticus further complicates management and requires continuous EEG monitoring with regular reassessment and adjustment of therapy. Benzodiazepines have been accepted as the first line of treatment on the basis of reasonable evidence. Emerging randomized controlled trials demonstrate equal efficacy for parenterally administered phenytoin, levetiracetam, and valproic acid as second-line agents. Beyond this, the evidence for third-line options is sparse. However, encouraging evidence for midazolam and ketamine exists with further data required for immunological, dietary, and surgical interventions.

Conclusion

Our overview of the management of pediatric and neonatal status epilepticus based on available evidence emphasizes the need for evidence-based guidelines to manage status epilepticus that fails to respond to second-line treatment.
Hinweise
Scan QR code & read article online
Status epilepticus (SE) generally has a bimodal distribution of incidence with the first peak observed during the first decade of life (14.3/100,000/year) and a second peak observed after the age of 60 years [1]. Pediatric SE is the most commonly observed pediatric neurological emergency worldwide, with a high rate of morbidity and a considerable rate of mortality [2, 3]. Each year, 20 per 100,000 children are affected by SE with an overall mortality of 3% [4]. In comparison with its adult counterpart, the mortality of pediatric SE is comparatively low (3% in children vs. 30% in adults), but the burden of short- and long-term morbidity that it carries is considerably higher [57].
The new diagnostic framework for classification of SE through different axes includes an age-group-based classification, i.e., neonatal, infancy, childhood and adolescence, adulthood and older age. Nevertheless, the quality and quantity of evidence available to guide management of pediatric and neonatal SE remains sparse in comparison with the abundant evidence available for its adult counterpart [8].
This is an important distinction because pediatric SE can be considered a separate entity on its own for a myriad of different reasons.
There is a wide heterogeneity in the underlying etiologies and mechanisms driving adult and pediatric SE that differ widely between these two groups. Population-based studies show that the commonest underlying etiology for pediatric status epileptics is non-CNS infection-related fever, commonly febrile SE, while in adults, SE occurs most often in the context of remote symptomatic etiologies with cerebrovascular accidents accounting for the majority of cases [1, 2, 9]. Relatively rare but distinct entities such as febrile infection-related epilepsy syndrome (FIRES) and new-onset refractory status epilepticus (NORSE) show a variable incidence between the two age groups, with FIRES commonly observed within the pediatric age range but NORSE commonly occurring during early to mid-adulthood [10, 11]. Furthermore, certain treatment modalities such as propofol and thiopental, although widely used in the management of adult SE, are known to cause harm in children.
Taking all these factors into consideration, the available abundant data from the adult population cannot be safety extrapolated to the pediatric population.
In this article, we primarily aim to review the available evidence for management of pediatric and neonatal SE. However, investigating the underlying pathophysiology of pediatric SE, including analyzing the associated cellular and molecular disturbances in SE, is beyond the scope of this article.

Definitions

The definition of SE and its subtypes has evolved significantly over time. The International League Against Epilepsy (ILAE) classification and terminology task force has proposed a new framework of pathophysiology-based definitions (Fig. 1).
Status epilepticus is now defined conceptually as “a condition resulting either from the failure of the mechanisms responsible for seizure termination or from the initiation of the mechanisms responsible, which lead to abnormally, prolonged seizures (after time point t1). It is a condition which can have long-term consequences (after time point t2), including neuronal death, neuronal injury, and alteration of neuronal networks, depending on the type and duration of seizures.”
The two operational dimensions or the time points stated above are defined as t1: the length of duration of seizure after which occurrence of “continuous seizure activity” is considered and treatment should be initiated and t2 or time point 2 as the time of seizure activity beyond which the risk of long-term consequences arise. Based on animal research, in convulsive SE, t1 and t2 are 5 min and 30 min, respectively, with no data available for timelines of the other forms of SE.
But both animal and human studies now confirm that the more prolonged the seizures are, the more likely that they are to become refractory leading to worse outcomes, which emphasizes the need for early treatment initiation and termination of seizures.

Prognosis

Although the overall prognosis ultimately depends on the underlying etiology, delay in treatment will not only prolong the duration of seizures and its refractoriness to treatment but it is also known to be associated with increased morbidity and mortality and poor long-term outcomes, independent of the underlying etiology [1216].
Acute complications of SE are numerous and may include cerebral edema, hemodynamic compromise and cardiac arrhythmia, non-cardiogenic pulmonary edema, excessive convulsive motor activity leading to hyperthermia as well as myoglobinuria, which may progress to acute renal tubular necrosis and renal failure, life-threatening electrolyte disturbances such as hyperkalemia and hyponatremia, as well as metabolic disturbances including hypoglycemia and metabolic acidosis, coagulopathy, gastrointestinal paresis s, intensive care neuropathy, and myopathy [17, 126].
The incidence of learning difficulties, neuro-disability, and de novo and drug-resistant epilepsy is also high in survivors.
Unfortunately, despite all the available evidence to the contrary, treatment delay is still a universal observation in the management of pediatric status epilepticus [18].

Management

Pharmacological management of SE can be approached as first, second, and third tiers of treatment options or as management of early or prehospital versus established SE. The first-line treatment approach for convulsive SE is usually with single or repeated doses of a benzodiazepines followed by a tier-two medication choice, either fosphenytoin or phenobarbital.

First-line treatment

Parenteral benzodiazepines remain the first-line treatment of choice in status epileptics (Table 1). First-line treatment with benzodiazepines alone will successfully terminate 40–60% of convulsive SE [18]. However, establishing venous access in an actively convulsing child needs expertise and can be quite challenging particularly in a prehospital setting. Despite the emphasis on importance of initiating prehospital treatment for early SE, pediatric-specific data are lacking to compare the efficacy of different formulations and routes of benzodiazepine administration in an out-of-hospital setting.
Table 1
Pediatric treatment trials: first-line anti-seizure medication for status epilepticus
Study, year (reference)
Study type
Number of pediatric cases
Medication compared
Outcome
Chamberlain et al. 2014 [19]
Class I RCT
273
IV diazepam vs. IV lorazepam
Equipoise in both meds at terminating seizures in 10 min (72%)
RAMPART [20]
Class I RCT
145
IV lorazepam vs. IM midazolam
No subgroup analysis. Overall, noninferiority of IM midazolam in comparison with IV lorazepam
RCT randomized controlled trial, IV intravenous, IM intramuscular
A class I randomized control study enrolled 273 children to first-line treatment with either intravenously administered (IV) diazepam or IV lorazepam and derived equipoise in the efficacy of seizure cessation within 10 min between IV diazepam (101/140, 72.1%) and IV lorazepam (97/133, 72.9%) with similar side effect profiles in the two treatment arms [19].
The Rapid Anticonvulsant Medication Prior to Arrival (RAMPART) trial included 145 patients between the ages of 0 and 20 years alongside 748 participants above this age range, who were then randomized to prehospital treatment with either intramuscular (IM) midazolam or IV lorazepam upon presentation with convulsive seizures lasting longer than 5 min. Although an age-based subgroup analysis was not carried out, overall non-inferiority of IM midazolam was established in comparison with IV lorazepam in prehospital termination of status epileptics with similar adverse effect profiles for both these medications [20].

Second-line treatment

Under familiar circumstances, second-line treatment is necessitated when convulsive SE persists after two doses of benzodiazepines [21]. An infusion of phenytoin is the most commonly utilized second-line approach in established SE with approximately 60% chance of seizure termination [22].
Phenytoin is known to cause severe adverse events when given as an infusion, including life-threatening cardiac arrhythmias and hypotension as well as severe extravasation injuries (purple glove syndrome) and liver toxicity. Fosphenytoin is of higher cost and is not universally available; however, it is preferred over phenytoin due to its better tolerability at higher infusion rates and relatively lower rates of adverse effects including fewer chances of cardiac arrhythmias and less extravasation-related harm. However, the benefit of higher rates of infusion of fosphenytoin is counteracted by the time necessary to transform the pro-drug into the active substance state.
Fortunately, randomized clinical trial-based evidence is emerging for other potential candidates as a second tier of management of pediatric SE (Table 2).
Table 2
Pediatric treatment trials: second-line antiseizure medication for status epilepticus
Study, year (reference)
Study type
Number of pediatric cases
Medication compared
Outcome
EcLIPSE, 2019 [23]
Open-label RCT
286
IV levetiracetam 40 mg/kg vs. IV phenytoin 20 mg/kg
Equipoise:
IV LEV – 70% seizure termination on average 35 min
IV PHT – 65% seizure termination average 45 min
ConSEPT, 2019 [24]
Open-label RCT
233
IV or IO phenytoin 20 mg/kg infusion over 20 min
vs.
IV or IO levetiracetam 40 mg/kg infusion over 5 min
Cessation of seizures at 5 min was equivocal
LEV – 50%
PHT – 60%
ESETT, 2019 [25]
Class I RCT
225
Infusions of,
levetiracetam 60 mg/kg
vs.
fosphenytoin 20 mg/kg
vs.
valproate 40 mg/kg
Seizure termination at 60 min was equivocal:
LEV – 52%
Fosphenytoin – 49%
Valproic acid – 52%
RCT randomized controlled trial, IO intraosseous, IV Intravenous, LEV levetiracetam, PHT phenytoin
Three treatment trials were published in 2019 assessing the treatment response to several different second-line antiseizure medication. The “Levetiracetam versus phenytoin for second-line treatment of pediatric convulsive SE” (EcLiPSE) trial was an open-label randomized clinical trial that was undertaken in United Kingdom. This study showed that out of 286 randomized participants aged between 6 months to under 18 years, receiving levetiracetam (40 mg/kg over 5 min) versus phenytoin (20 mg/kg over at least 20 min), convulsive SE was terminated in 106 of 152 (70%) children in the levetiracetam arm and in 86 of 134 (64%) in the phenytoin arm [23]. The median time from randomization to cessation of convulsive SE was 35 min in the levetiracetam group and 45 min in the phenytoin group (hazard ratio: 1.20, 95% CI: 0.91–1.60; p = 0.20).
Another open-label, multicenter, randomized controlled trial conducted in 13 emergency departments in Australia and New Zealand, the ConSEPT trial, randomly assigned 233 children between the ages of 3 months and 16 years to receive either 20 mg/kg phenytoin (IV or intraosseous [IO] infusion over 20 min) or 40 mg/kg levetiracetam (IV or IO infusion over 5 min) [24]. The study showed that 68 of 114 (60%) patients in the phenytoin group and 60 of 119 (50%) patients in the levetiracetam group had cessation of SE within 5 min of cessation of the infusion (risk difference −9.2% [95% CI: −21.9–3·5]; p = 0.16), with no difference in the rates of adverse events occurring within 2 h of receiving the study drug or subsequently during admission. Although both of these studies did not establish any superiority of one medication over the other, the findings show that levetiracetam is as an appropriate alternative to phenytoin as a second-line agent.
The established SE treatment trial (or ESETT trial) is a multicenter double-blinded, responsive-adaptive, randomized controlled trial carried out in the United States. Overall, 400 patient enrollments (384 individual patients) were included, and were stratified according to age, i.e., 39% were children and adolescents (up to 17 years of age), 48% were younger adults (18–65 years of age), and 13% were older adults (> 65 years of age). They were randomized to receive levetiracetam 60 mg/kg (maximum 4500 mg), fosphenytoin 20 mg/kg (maximum 1500 mg), or valproate 40 mg/kg (maximum 3000 mg) infused over 10 min [25]. Each of the three study medications showed approximately 50% effectiveness in aborting SE within 1 h in all of the age groups. Analyzing the results of the pediatric age group separately, the responses to levetiracetam, fosphenytoin, and valproic acid were 52%, 49%, and 52%, respectively. There was no difference in achieving seizure freedom at 60 min from the onset of study drug infusion and this was without the use of an additional antiseizure medication. The safety outcomes did not differ by treatment group in any of the age groups.

Third-line treatment

Failure of second-line treatment should lead to initiation of the third-line therapy phase when the seizure duration reaches 40 min, at which point the conservative practice is initiation of anesthesia. This requires endotracheal intubation with rapid sequence induction (RSI) to prevent the accompanying airway compromise [21]. All conventional anesthetics delivered through infusion require endotracheal intubation, which is an individual negative prognostic factor of morbidity and mortality in SE [26].
Although first- and second-line treatment for pediatric SE is based on high-grade evidence derived from randomized controlled trials (RCTs) and meta-analyses with even better quality evidence on emergence, unfortunately the evidence for third-line agents remains controversial and is based on observational studies and expert opinion alone ([27], Table 3).
Table 3
Best available evidence: management of pediatric RSE and SRSE
Study, year (reference)
Study type
Number of pediatric cases
Treatment
Outcome
Wilkes and Tasker (2014) [33]
Systematic review
521
IV midazolam
Sz control – 76%
Average time for Sz cessation – 271 min
Breakthrough Sz – 52%
Sz recurrence – 12%
Wilkes and Tasker (2014) [33]
Systematic review
95
Barbiturate infusion
65% who failed midazolam respond to barbiturate
Sz control or BS in 22.6 h
Breakthrough Sz – 67%
Sz recurrence – 22%
a. Invento et al., 2015 [62]
b. Rosati et al., 2012 [121]
Case series
a. 11
b. 9
Ketamine
1. SE resolved – 14/19 episodes
2. 66% Sz control
Kofke et al., 1989 [67]
Case series
5
Isoflurane
Sz cessation or BS – 5/5 (100%)
Mean duration of treatment – 7 days
Guilliams et al., 2013 [99]
Case series
5
Hypothermia (30–35.3 0C)
Sz control – 5/5 (100%)
Relapses – 0%
Schoeler et al., 2021 [89]
Systematic review
147
Ketogenic diet
Ketosis in – 96%
SRSE resolution – 60%
Average time to Sz resolution – 6.3 days
Uberall et al., 2000 [56]
Case series
41
IV valproic acid as an adjunct to a 2nd-line agent
RSE termination in – 78%
IV intravenous, Sz seizure, BS burst suppression, SE status epilepticus, SRSE super-refractory status epilepticus, RSE refractory status epilepticus

Midazolam

Most adult SE treatment protocols include IV midazolam, short-acting barbiturates such as pentobarbital, or thiopentone infusions or propofol as the three most widely utilized options, with a less clear understanding of their effectiveness in comparison [2831]. An even lower number of evidence-driven options are available for the pediatric population.
A prospective observational study carried out by the Pediatric Status Epilepticus Research Group (pSERG) between 2011 and 2013 identified 111 patients with refractory status epilepticus (RSE; median age: 3.7 years) of whom 54 patients (49%) required a continuous infusion of an anesthetic agent. A median of five bolus doses of antiseizure medication (interquartile range: 4–7) had been given with no effect prior to the start of the infusion medication. Midazolam was the most frequently administered first-choice anesthetic agent (78%) followed by pentobarbital. Overall, 94% patients achieved seizure termination with either of these two therapies. Other agents that were used as third-line medications were propofol, ketamine, valproate, and isoflurane [32].
A systematic literature search and review by Tasker and colleagues identified that when midazolam was used as the initial agent for RSE, there was a 76% clinical seizure control achieved within 41 min on average after starting the infusion. However, when two studies with continuous EEG monitoring were included, the weighted mean time from beginning of the midazolam protocol to seizure control increased to 271 min, suggesting the possibility of ongoing subclinical seizures despite cessation of clinically evident seizures. Breakthrough seizures occurred in 52% of patients treated with midazolam infusion and seizures recurred in 12% [33].
The mechanism of action of midazolam is via positive allosteric modulation of gamma aminobutyric acid type A (GABAA) receptors suppressing the neuronal excitability. Its rare but well-described side effects include hypotension, respiratory suppression, and hepatotoxicity as well as development of tolerance or tachyphylaxis with increasing doses [34]. Cumulative evidence suggests that an initial bolus of 0.1–0.5 mg/kg followed by increments of 1–2 μg/kg/min up to 30 μg/kg/min is efficient in controlling SE in the pediatric population [28].

Barbiturates

In those patients in whom midazolam has failed, barbiturate infusion was effective in terminating seizures in 65% of cases [33]. On average, a barbiturate infusion was initiated 66 h after the onset of RSE. Burst suppression, which remains the ultimate electroencephalographic goal, was achieved 22.6 h later. However, 67% of patients had breakthrough seizures and recurrence of seizures after weaning from barbiturates occurred in 22%.
Pentobarbital is a metabolite of thiopental, a barbiturate that depresses neuronal excitability by enhancing GABA-associated inhibitory responses. Compared with phenobarbital, pentobarbital readily penetrates the blood–brain barrier allowing for rapid onset of action and seizure control with a shorter half-life thereby enabling faster recovery from coma and awakening upon weaning. It may accumulate in tissue with prolonged administration due to its lipid solubility and has a considerable side effect profile including significant severe respiratory suppression, myocardial depression, profound hypotension, immune suppression, and risk of multiorgan failure, among others.
A retrospective data analysis of 26 children between the ages of 1 day and 13 years with RSE showed a response rate of 52% when a loading dose of 5 mg/kg of pentobarbital followed by an infusion of 1–3 mg/kg/h was used [35]. Seizure relapse was observed in 22% of cases.

Propofol and thiopental

Propofol is an IV general anesthetic agent with the property of a positive modulator of GABA receptors. It generally has a short half-life and is easily titratable; it is rapid acting, enabling rapid recovery after drug discontinuation, although with prolonged administration, the terminal half-life may amount to several days.
In adults, propofol has evidence-based utility in management of RSE or super-refractory status epilepticus (SRSE) due to its aforementioned rapid onset of action and prompt awakening of patients upon withdrawal. Propofol induces burst-suppression within 35 min of initiation; however, frequent titration is required to maintain adequate suppression. Adult studies have shown that propofol infusion terminates RSE/SRSE in 67% of cases [36, 37].
Unfortunately, the high risk of “propofol infusion syndrome” (PRIS) often limits its use in children [38]. It has been reported that PRIS could lead to early Brugada–Brugada-like ECG changes, cardiac failure, rhabdomyolysis, metabolic acidosis, renal failure, and sometimes even death [39, 40, 127, 128]. Propofol also has a propensity to induce hyperkinesias, which can mimic breakthrough seizures [41]. However, these are not true seizures, but extrapyramidal hyperkinesias given their pathomechanism: Propofol inhibits cortical than basal ganglia excitatory activity much faster, which leads to disinhibition of basal ganglia motor programs and the hyperkinesias [129, 130].
Reported risk factors associated with a higher risk of PRIS include younger age, higher doses, prolonged use, concurrent use of with catecholamines and corticosteroids, and a low body mass index or presence of malnutrition [42, 43]. There have been descriptions of PRIS in patients receiving propofol doses greater than 5 mg/kg/h for more than 48 h. However, more recent data suggest that even shorter durations or lower doses of propofol (less than 4 mg/kg/h) can also result in development of PRIS, exposing pediatric patients to high risk of death and morbidity [4446]. When propofol was administered in conjunction with a ketogenic diet, more severe adverse events including case fatalities were reported [47]. It also worth noting that the propofol syndrome may not be caused by propofol in entirety but may have resulted from drug-induced cerebral suppression.
A retrospective study assessed the effectiveness of propofol in comparison with thiopental in 33 children aged between 4 months and 15 years with RSE and concluded that propofol was more effective than thiopental, with propofol being successful in terminating seizures in 64% and thiopental in only 55% of cases [48]. However, propofol had to be discontinued in 18% of patients due to adverse events, which included rhabdomyolysis and hypertriglyceridemia.
All these factors may limit the utility of propofol as a third-tier agent in the management of pediatric SE, and stronger evidence is required to assess the associated safety concerns.

Refractory status epilepticus

Refractory status epileptics is defined as SE that persists despite administration of at least two appropriately selected and dosed parenteral mediations including benzodiazepines. No specific seizure duration is required for this diagnosis. Approximately 15–35% of patients with RSE fail to achieve the desired therapeutic endpoint and progress to SRSE.

Super-refractory status epilepticus

Super-refractory status epilepticus is defined as SE that continue for 24 h or more, despite anesthetic treatment, or recurs on attempted weaning off the anesthetic regimen requiring reintroduction of anesthetic agents [49, 50].
By the time SRSE ensues, it is presumed to be very likely that the neuronal damage has been already established and the goal of management thus deviates from simply halting and preventing seizures to anticipatory monitoring and pre-emptive managing down-stream consequences, which include multiorgan dysfunction [17].
Based on a variety of pathophysiological mechanisms, resistance to conventional medication is established by the time SRSE is established [51]. Almost all conventional anesthetic agents used in treatment of SRSE act on inhibitory GABAA receptors [51]. Experimental models suggest that with continuing seizures, GABAA receptors composed of alpha, beta, and gamma subunits are internalized while those receptors containing delta subunits and localized extrasynaptically remain pharmacologically responsive to very high doses of midazolam and neurosteroids [131]. By contrast, excitatory N-methyl-d-aspartate (NMDA) receptors are mobilized to the cellular membrane, which results in a clearly hyperexcitable state at the synapse [52]. This excess glutamate release with ongoing seizure activity lasting for longer than 1 h is postulated to unfavorably alter the balance between neuronal excitation and inhibition and to sustain seizure activity.
Neurological sequelae are expected in more than 50% of the pediatric population with refractory convulsive SE despite best available care; refractory convulsive SE has a mortality rate of 2.7–5.2% [15, 53].
There is a significant lack of consensus or a clear evidence-based guidelines on how to approach pediatric RSE and/or SRSE. A variety of different approaches are utilized worldwide.
A retrospective cohort study demonstrated that in a group of 31 pediatric patients with SRSE, a single continuous drug infusion was effective in 48.4% of cases for resolution of SE. Two infusions with different drugs were utilized in 32.3% and three or more different drugs were used in 19.4% of cases. Most of the patients with SR SE (96.8%) received midazolam as the first choice [54]. The same study observed that SRSE patients had delayed initiation of first non-benzodiazepine antiseizure medication in comparison with the non- SRSE patients (149 min vs. 62 min; p = 0.030). Additional therapies that were administered in 17 out of 31 patients (54.8%) in this study cohort included corticosteroids, ketogenic diet, and intravenous immunoglobulin (IVIG) as the most favored options.
Some of the other nonconventional or debated management modalities that are available for treatment of SRSE are discussed next.

Intravenous valproic acid

Valproic acid is a conventional antiseizure medication that is used as maintenance therapy, but several case reports are available to demonstrate that valproic acid given as a continuous infusion can be effective in terminating RSE [55]. In a case series of 41 children with RSE, a continuous infusion of valproic acid successfully terminated seizures in 78% when used in addition to a conventional second-line medication such as phenobarbital [56].
However, the use of valproic acid in children can be problematic with metabolic conditions being a more common underlying etiology of SE in children than in adults, predisposing them to hepatotoxicity and pancreatitis as a consequence. Certain mitochondrial metabolic conditions such as mutations of the POLG gene or in the urea cycle enzyme system, such as ornithine transcarbamylase (OTC) deficiency, are accepted contraindications for its use, which may remain undiagnosed at the time of presentation. There is also a known age restriction where children below the age of 2 years are generally excluded from receiving valproic acid. Universal unavailability of IV formulations can also act as another limiting factor.

Ketamine

There is increasing evidence to suggest that ketamine may be effective in treating refractory convulsive SE compared to conventional anesthetics in adults [57]. Ketamine is a potent N-methyl-D-aspartate (NMDA) antagonist. The NMDA receptors are still functional at later stages of SE even after the GABA receptors have been internalized rendering conventional anesthetics that utilize this pathway inefficacious [58]. It also has sympathomimetic properties with utility in neuroprotection [59]. Ketamine has neither cardiac nor respiratory depressant properties. Therefore, endotracheal intubation may not be required with ketamine in comparison with other conventional anesthetics, especially considering that mechanical ventilation itself is considered a factor that increases morbidity and mortality risk in critically ill pediatric patients [60, 61]. It is tempting to speculate that earlier, i.e., before it enters the refractory stage, use of ketamine in SE may be more effective than the administration in the (super-)refractory phase only.
Ilvento et al., in their study of 13 children, aged between 2 months and 11 years 5 months, with a total of 19 treatment episodes using ketamine at a median dose of 30 µg/kg/min, observed that refractory convulsive SE was resolved in 14 out of 19 episodes. Four patients who received ketamine in lieu of conventional anesthetics achieved resolution of status and thus did not require endotracheal intubation [62]. A burst suppression EEG pattern was obtained in 10 of 19 episodes. Aside from a slight increased saliva production and a transient mild increase of liver enzymes in some of the children receiving concomitant phenobarbital, no serious adverse events were observed. Add-on midazolam at 1 mcg/kg/min was administered as per protocol to prevent hallucinations, which are an expected side effect.
The KETASER01 trial (Ketamine in Refractory Convulsive Status Epilepticus in Pediatric Convulsive RSE) was planned to evaluate the efficacy of ketamine in this population of interest. This multicenter, randomized controlled trial had to be halted because of much too slow recruitment [63, 132].

Inhaled anesthetics

The mechanisms of action of inhaled anesthetics are not well established, but both isoflurane and desflurane are known to produce dose-dependent changes in the EEG. The postulated mechanism of action is likely via potentiation of inhibitory postsynaptic GABAA receptor-mediated currents or through its effects on the thalamocortical pathways [64]. Isoflurane and desflurane both are easily titratable and they both have favorable pharmacokinetic and pharmacodynamic properties that are effective in achieving burst-suppression patterns on EEG [65].
However, only limited data are available for the use of isoflurane and desflurane and their efficacy in the management of pediatric RSE with most of the validated data coming from the adult population. In addition, many hospitals stopped or heavily restricted the use of fluranes because of their catastrophic effect on the environment.
In a case series of seven patients with RSE aged between 17–71 years, isoflurane with and without desflurane was effective in seizure cessation and achieving burst suppression [66]. Complications were frequent, the commonest complication being hypotension with all of the patients in the series requiring volume resuscitation and vasopressors. Among other complications were atelectasis (7/7), infections (5/7), paralytic ileus (3/7), and deep venous thrombosis (2/7).
In another case series where five pediatric patients were included, Kofke et al. reported seizure response and achievement of burst suppression in all five patients with isoflurane [67]. However, potential neurotoxicity with inhaled anesthetics is worth considering in light of recent case reports of magnetic resonance imaging (MRI) changes observed in variable brain regions with prolonged isoflurane exposure [68, 69].
These MRI changes were particularly observed in the hippocampal region, hypothalamus and the cerebellum associated with prolonged use in RSE compared with those who received only an intravenous anesthetics.
Under these circumstances risks versus benefits need to be considered in using inhaled anesthetics in the pediatric population.

Immunomodulatory therapy

Immunomodulatory therapy in the management of epilepsy and SE is a relatively novel approach gaining popularity over the years, especially in the management of immune-mediated epilepsies and autoimmune encephalitis. It has been identified that immune-mediated processes may modulate epileptogenicity. Evidence has emerged on the use of immune modularity therapy in successful treatment of intractable childhood epilepsies and epilepsies with a known or a presumed underlying inflammatory or autoimmune pathology [7075].
Immunomodulatory therapy may also be efficacious in patients with no established underlying immune-mediated etiology primarily because not all anti-neuronal antibodies can be identified by current commercially available panels. Therefore, a trial of immunomodulatory therapy can be justified in pediatric RSE/SRSE patients with no obvious contraindication, particularly for those presenting with temporal lobe seizures without a known underlying immune-mediated pathology.
Steroids in particular may have an additional benefit in managing cerebral edema and raised intracranial pressure that may occur concomitantly with prolonged seizures [76].
Based on a literature review by Ferlisi et al., only 5% of patients with RSE and SRSE have adequate seizure control with steroids, IVIG, and plasma exchange [77].

Neurosteroids

Neuroactive steroids have been studied in experimental epilepsies in animals and were shown to alter their cortical excitability and therefore reduce seizure propensity.
Brexanolone, an aqueous formulation of allopregnanolone, was experimented in adults with SRSE as an adjunctive medication in a phase I/II open-label trial. Out of 25 patients with a median age range of 47.6 ± 19.5 years, 17 were successfully weaned off third-line anesthetics with the use of the study medication. No brexanolone-specific serious safety concerns were identified. However, the phase 3 trial reported that the primary endpoint was not significantly different between the study drug (43.9%) and the placebo (42.4%, p = 0.88) at the end of the double-blind period [78].
Methylprednisolone pulse therapy was observed to be beneficial for presumed immunologically driven epilepsies such as FIRES and NORSE [79].

Intravenous immunoglobulins

It has been shown that IVIG may be of benefit in some of the antibody-mediated epilepsies and CNS neuroinflammatory processes as well as other types of epilepsies that are not yet proven to be immune mediated such as  Continuous Spike and Wave during Slow wave sleep (CSWS) and refractory focal epilepsies.
Out of 33 adults in a systematic review who had received IVIG for RSE, 14 achieved seizure freedom with no reported adverse effects [80]. Furthermore, IVIG also demonstrated some efficacy in managing FIRES- (16.6%) and NORSE-related (29.4%) SE [79].

Plasmapheresis

Use of plasmapheresis for RSE has been reported in 27 adults with autoimmune conditions, with 13 patients achieving seizure control [81]. Plasmapheresis was effective in two of 18 patients with FIRES and six of 15 patients with NORSE [79]. However, the reports have been less encouraging in children, with only seven of 37 patients achieving seizure control and two other patients experiencing a decrease in seizure burden [82].
In the realm of pediatric SE, more evidence is required with regard to potential long- and intermediate-term side effects as well as evidence on whether any particular combination of different immunomodulatory therapies, rather than relying on one modality, might be of more benefit [83].

Dietary therapy

Ketogenic diet (KD) is a therapeutic diet that is high in fat and low in carbohydrate with proven efficacy in certain drug-resistant epilepsies such as infantile spasms, childhood epileptic encephalopathies, inborn errors of metabolism, as well as drug-resistant focal epilepsies. Over the past several years there is emerging evidence for the use of KD in the context of RSE and SRSE.
The exact mechanism of action of KD, especially in abolishing refractory seizures, is not precisely understood, but it has been postulated that KD potentially disrupts the pathophysiology of RSE at multiple levels including, but not limited to, neurotransmitter receptor trafficking, neurotransmitter localization and release, excitotoxicity, alteration in the gut microbiome, and blood–brain barrier (BBB) permeability with downstream effects on action of various inflammatory mediators and on mitochondrial dynamics [84, 85, 133].
Fortunately, most of the studies investigating the efficacy of KD on RSE are pediatric studies, possibly due to its ease of administration in this population. In most cases, KD is delivered via the enteral route in a ratio of 4:1 and occasionally in the form of ketogenic total parenteral nutrition (TPN).
A case series reported that 10 of 14 pediatric patients had electrographic seizure resolution and ≥ 50% suppression within 7 days of initiating KD [86]. The median delay in starting KD after the onset of SE in this series was 14 days.
Another pediatric case series reported resolution of SRSE in nine out of 10 patients after a median of 7 days’ KD duration [87]. Anesthesia could be weaned off in eight of these 10 patients within 15 days of KD initiation.
In another series of 10 pediatric RSE patients, two patients had seizure resolution and five patients had > 50% decrease in seizure frequency over a mean duration of 5 days after initiating KD [88].
A more recently published systematic review assessed 147 pediatric patients who were started on a KD with 96% of them achieving ketosis and SRSE resolution in 60% over an average of 6.3 days [89]. A total of 34% were reported to have mild side effects in this analyzed population and 4% had side effects categorized as being severe or serious.
However, KD takes relatively longer time to take effect in comparison with other nonsurgical approaches of RSE management (weeks vs. days). There are also concerns about variability in the definition of ketosis and representative outcomes in this population.
While the incidence of adverse effects associated with the use of KD for treatment of RSE/SRSE has reportedly been low, commonly observed adverse effects include metabolic derangements such as keto acidosis and hypoglycemia and common gastrointestinal symptoms such as emesis and constipation [86, 89, 90].

Therapeutic hypothermia

Therapeutic hypothermia has demonstrated antiseizure and neuroprotective properties under experimental conditions, which are presumed to be exerted via several different pathways including decreasing cerebral oxygen utilization, metabolic rate, excitotoxicity, calcium release, free radical production, release of reactive oxidative molecular species, and altering BBB permeability to inflammatory intermediates [9193].
Although hypothermia was used in isolation to control SE in the first reported series of patients, today therapeutic hypothermia is almost never used alone in the management of RSE and is usually used in conjunction with other available modalities.
In an adult case series of four adult patients in a neurology/neurosurgery critical care setting, endovascular cooling (31–35 °C) successfully controlled RSE, allowing for gradual withdrawal of IV anticonvulsants and vasopressors [94]. Two out of four patients had sustained seizure freedom, and all patients showed a marked reduction in seizures.
The only class‑1 evidence for the use of hypothermia in management of RSE comes from the HYBERNATUS study, which is a multicenter study recruiting 270 patients with convulsive RSE who were on assisted ventilation. The trial found that the efficacy of therapeutic hypothermia was not better than placebo for RSE/SRSE, and there was no difference in achieving the primary endpoint of a Glasgow outcome score of 5 (minimal or no neurologic deficit) at 90 days between the intervention and control groups (49 vs. 43%; [95]).
With most of the available evidence coming from descriptive studies, a very early pediatric case series describes three patients with generalized SE who were treated successfully with induction of therapeutic hypothermia of 30–31 °C in conjunction with 5–55 mg/kg/h of thiopental infusion [96].
A case report of an infant with RSE due to hemimegalencephaly described decreased seizure frequency with induction of mild hypothermia at 36 °C [97]. Two additional case reports described prompt and sustained control of RSE in two pediatric patients diagnosed with FIRES with induction of moderate hypothermia at 33 °C [98]. In another case series of five pediatric patients with RSE, including four with relapse on attempted discontinuation of pentobarbital, Guilliams et al. reported that therapeutic hypothermia of 32–35 °C could bring about sustained decrease in seizure burden even when midazolam, pentobarbital, and ketamine had failed [99].
The adverse effects that were highlighted in these case series associated with even mild hypothermia included coagulopathy, venous thromboembolism, renal tubular dysfunction with attendant acid–base and electrolyte abnormalities, bradyarrhythmias, and intestinal paresis.
Further questions that remain unanswered with regard to the practicality of this approach are the optimal core body temperature to be targeted, optimal time lag to achieve this target temperature, optimal duration of hypothermia to be sustained, and the target percentage suppression of EEG as an electrographic goal. Other management questions also remain unanswered, such as the routine use of prophylactic heparin for prevention of coagulopathy, the mode of induction of the hypothermia such as surface cooling versus endovascular cooling, and the benefits versus the risks of each of these methods. Although very few of these questions could be answered with evidence extrapolated from the neonatal age group who underwent therapeutic hypothermia for perinatal ischemic injury, these may not be applicable to older age groups.

Pyridoxine

Pyridoxine-dependent epilepsy results from pathogenic mutations in the ALDH7A gene causing a functional antiquation deficiency. Pyridoxine has been effectively used to treat SE associated with pyridoxine-dependent epilepsy [49, 100]. Pyridoxine has also been reported to be helpful for SE occurring in the context of isoniazid toxicity or with pyridoxine deficiency associated with prolonged isoniazid therapy [101, 102].
An empiric trial with pyridoxine may be considered in the management of RSE in infants or younger children. Side effects of acute IV pyridoxine infusion include potential respiratory depression and hypotonia. However, most RSE/SRSE patients are likely to be ventilated at the stage where pyridoxine is given as a trial.

Magnesium

The mechanism of action of magnesium in the management of RSESE has not been completely elucidated. Magnesium—at the resting potential in mammalian (also human) neurons—blocks the ionophore of the glutamatergic NMDA receptor. During physiological depolarization, the magnesium ion exits and renders the ionophore permeable, i.e., excitable. Hypomagnesemia, as a consequence, facilitates hyperexcitability because the neurons became excitable already at the resting state potential voltage [134]. Nevertheless, magnesium is rarely used in light of the very limited evidence outside of specific contexts such as treatment of eclampsia or in patients with provoked seizures due to hypomagnesemia from gastrointestinal or renal losses [103].
A single case report of two teenage girls with juvenile-onset Alper’s syndrome due to POLG1 variants describes remission of SE in response to IV magnesium [104].
In a systematic review including 28 patients (including nine children), seizure control was achieved in 12 patients but half of them had seizure recurrence after withdrawal of magnesium [105].
Significant side effects that are reported with hypermagnesemia (with serum levels > 4.5 mg/dL) are hypotonia, weakness, or even paralysis resulting from the inhibition of acetylcholine release at the neuromuscular junctions. Vasodilation is also known to occur at higher concentrations (with magnesium concentrations exceeding > 15 mg/dL) and can result in complete heart block or cardiac arrest.

Surgical management

Surgical resection of an area of the cortex with or without an identified epileptogenic lesion, with the presence of clear evidence to indicate that the seizures are originating from this particular region or lesion, can be curative in RSE. This may not be a straightforward decision if the imaging is negative or is discordant with electrophysiological data and may mandate extensive work-up including intracranial EEG monitoring and/or electrical stimulation mapping, quite similar to a presurgical evaluation to assess surgical candidacy. However, carrying out a presurgical evaluation on an emergent basis in a medically unstable patient is not only difficult but also is confounded by ongoing ictal activity and by the effects of concomitantly administered anesthetic infusions.
Surgical planning and decision-making should also incorporate the location, size, and nature of such lesions as well as the functional significance of the surrounding eloquent cortex.
In a series of adults between the ages of 20 and 68 years, five out of nine patients were found to be seizure free at 1 month to 7 years after electrocorticographically guided resections [106]. Five out of nine patients had focal or diffuse nonspecific hyperintense lesions and three patients had solitary lesions, which included an intra-parenchymal hemorrhage, a resection cavity, and evidence of mesial temporal sclerosis with unilateral hippocampal atrophy.
As for evidence on pediatric patients, a case report of a 10-year-old boy with generalized convulsive RSE after a mild febrile illness and restricted diffusion in the right hippocampus described termination of RSE with right anterior temporal lobectomy [107].
Furthermore, left occipital lobectomy in a 7-year-old boy with anti-NMDA receptor encephalitis 3 months after onset of RSE led to seizure resolution and so did Electro Corticography(ECoG)-guided resection of the right dorsolateral frontal cortex in a 20-year-old man with CNS vasculitis [108, 109].
On rare occasions, multiple subpial transections have also been tried, usually as an adjunct to cortical resection [110]. An isolated multiple subpial transection of the sensorimotor cortex was successful in abolishing seizures in a 6-year-old child with MRI-negative SRSE of 60 days’ duration, with a clearly localizable seizure-onset zone [110].
Palliative surgical options include neurostimulatory options such as vagus nerve stimulation (VNS) and complete corpus callosotomy and functional hemispherectomy with several case reports of successful control of RSE [111, 112].

Neurostimulation

Delivery of an external electrical stimulus that is of adequate voltage and is properly timed in refractory epilepsy can prolong the post-excitation refractory period of the hyper-synchronous discharges and has the potential to interrupt an ongoing seizure.

Vagus nerve stimulation

There are case reports of young adults who underwent VNS surgery for prolonged RSE refractory to infusions of anesthetics resulting in seizure termination after the procedure enabling the tapering of other infusions [113, 114].
A 13-year-old boy with a history of previous 90% callosotomy underwent left VNS implantation 15 days after onset of RSE with a rapid response of seizures with the VNS settings of 30 Hz of frequency, pulse width 500 ms, on time 7 s, off time 120 s and current 0.25 mA [115].
Other pediatric case reports suggesting efficacy of neurostimulation in SE include a case report of a 17-year-old girl with generalized convulsive RSE demonstrating electro-clinical improvement of RSE after deep brain stimulation targeting the anterior thalamic nucleus [116, 117].

Electroconvulsive therapy

Electroconvulsive therapy (ECT) is an unpopular modality of neurostimulation that is hypothesized to be effective in terminating seizures by increasing presynaptic release of GABA resulting in a prolonged postictal refractory period [118].
In a 13-year-old boy with polymicrogyria and epileptic encephalopathy, consecutive ECT sessions over 3 days were successful in controlling RSE [119].
Another case report describes termination of SRSE with ECT in a 4-year-old boy with FIRES after multiple other therapeutic strategies failed including benzodiazepines, phenytoin, barbiturates, corticosteroids, plasmapheresis, immunoglobulins, propofol, lidocaine, ketamine, inhaled desflurane, KD, lacosamide, and even therapeutic hypothermia [120].
Overall, neurostimulation (transcranial magnetic stimulation, Deep Brain Stimulation (DBS), and ECT) in treatment of RSE/SRSE remains poorly understood and utilized at present. Candidate selection, identifying stimulation targets, stimulation frequency, dose and regimen as well as the safety profile need to be evaluated prior to adaptation [83].

Noninvasive neurostimulation

Noninvasive methods of neurostimulation such as transcranial magnetic stimulation are emerging modalities for treating RSE/SRSE, which may be of use in the future, but they require better quality evidence prior to adaptation.

Management and prognosis of neonatal status epilepticus

Neonates are considered a special subgroup within the pediatric population, taking into consideration a variety of physiological and maturational factors. Likewise, neonatal seizures are considered an entity on their own based on the electro-clinical characteristics that differ widely from the rest of the pediatric population. Therefore, it is only fair that neonatal seizures and their management are considered separately, adapting a distinct approach.
Neonates are at a greater risk of developing seizures in comparison with the older children or adults, with acute provoked seizure being the commonest in this age group. The International League Against Epilepsy (ILAE) defines a neonatal seizure as an electrographic event with a definitive beginning and an end and clear electrographic evidence of evolution, but with no defined minimum duration, which is a significant deviation from seizures defined in other age groups [122]. Also, no universally accepted definition exists for neonatal SE, although neonatal SE is quite common and is associated with a higher burden of neurodevelopmental disability [123]. A cohort study by Pisani and colleagues observed that only one out of eight newborns with neonatal SE would have a normal developmental outcome and six out of eight of them went on to develop cerebral palsy later in life. In this study, a mortality rate of 19% was observed with the presence of either SE or recurrent neonatal seizures, emphasizing the degree of associated disability [124].
The NEOLEV2 trial is a multicenter, randomized, blinded, phase IIb trial investigating the efficacy and safety of levetiracetam (40 mg/kg bolus followed by maintenance) compared with phenobarbital (20 mg/kg bolus followed by maintenance) as a first-line treatment of seizures in 83 neonates. In total, 80% of neonates who received phenobarbital were electrographically seizure free at 24 h while only 28% in the levetiracetam arm were able to achieve a similar outcome [125]. Beyond this, there is no evidence-driven data to guide management of SE in this specific population of patients.

Future directions

It is abundantly clear that the burden of disability and acute and long-term morbidity risks that are associated with pediatric SE (SE) is significant. Large knowledge gaps exist in the management of pediatric SE with several questions remaining unanswered. In contrast to 5–10 years ago, we now have a substantial amount of evidence to support the use of several treatment options as first- and second-line agents in managing pediatric SE. However, beyond this point the evidence is sparse and the management protocols and guidelines are largely institutional based and expert opinion driven.
The challenge of developing evidence-based guidelines beyond this point is in the lack of properly designed trials to assess each of the available modalities individually for efficacy and safety. By the time refractory SE ensues, ongoing management in a critical care setting includes multiple other instituted medication regimens, which will invariably confound the efficacy and safety profile of the trial medication. It is often hard to overcome this barrier in designing good-quality trials.
Prehospital treatment initiation of pediatric SE aiming to prevent progression into established SE is another area that lacks clear guidelines or protocols.
It is also worthwhile to explore other potential therapeutic targets in the treatment of SE, which expands beyond the realms of extensively explored current targets. Some of the potential candidates include but are not limited to targeting specific inflammatory mediators exploring the benefits of utilizing therapeutic agents such as anakinra or tocilizumab. Exploring and managing altered gene expression in SE such as alterations of the mammalian target-of-rapamycin (mTOR) pathway with the use of agents such as rapamycin, and exploring dysregulations in cholesterol synthesis, cholinergic pathways, and mitochondrial oxidation are other potential options.
Incorporating machine learning to develop predictive models and reliable biomarkers to predict and identify risk factors for progression into refractory SE and super-refractory SE will also hasten the management of SESE and improve outcomes.
Developing universally accepted, evidence-based clear guidelines leading to rational use of polytherapy targeting one or more etiology-specific targets with minimal adverse events would be the ultimate goal to pursue.

Declarations

Conflict of interest

E. Swarnalingam, K. Woodward, M. Esser and J. Jacobs declare that they have no competing interests.
For this article no studies with human participants or animals were performed by any of the authors. All studies mentioned were in accordance with the ethical standards indicated in each case.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

Clinical Epileptology

Print-Titel

• Übersichten, Originalarbeiten, Kasuistiken

• Aktuelles aus der epileptologischen Diagnostik und Therapie  


e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
2.
Zurück zum Zitat Chin RF, Neville BG, Peckham C et al (2006) Incidence, cause, and short-term outcome of convulsive status epilepticus in childhood: prospective population-based study. Lancet 368:222–229PubMed Chin RF, Neville BG, Peckham C et al (2006) Incidence, cause, and short-term outcome of convulsive status epilepticus in childhood: prospective population-based study. Lancet 368:222–229PubMed
3.
Zurück zum Zitat Novorol CL, Chin RF, Scott RC (2007) Outcome of convulsive status epilepticus: a review. Arch Dis Child 92:948–951PubMedPubMedCentral Novorol CL, Chin RF, Scott RC (2007) Outcome of convulsive status epilepticus: a review. Arch Dis Child 92:948–951PubMedPubMedCentral
4.
Zurück zum Zitat Gurcharran K, Grinspan ZM (2019) The burden of pediatric status epilepticus: epidemiology, morbidity, mortality, and costs. Seizure 68:3–8PubMed Gurcharran K, Grinspan ZM (2019) The burden of pediatric status epilepticus: epidemiology, morbidity, mortality, and costs. Seizure 68:3–8PubMed
5.
Zurück zum Zitat Maytal J, Shinnar S, Moshe SL, Alvarez LA (1989) Low morbidity and mortality of status epilepticus in children. Pediatrics 83:323–331PubMed Maytal J, Shinnar S, Moshe SL, Alvarez LA (1989) Low morbidity and mortality of status epilepticus in children. Pediatrics 83:323–331PubMed
6.
Zurück zum Zitat Wu YW, Shek DW, Garcia PA, Zhao S, Johnston SC (2002) Incidence and mortality of generalized convulsive status epilepticus in California. Neurology 58:1070–1076PubMed Wu YW, Shek DW, Garcia PA, Zhao S, Johnston SC (2002) Incidence and mortality of generalized convulsive status epilepticus in California. Neurology 58:1070–1076PubMed
7.
Zurück zum Zitat Hussain N, Appleton R, Thorburn K et al (2007) Etiology, course and outcome of children admitted to pediatric intensive care with convulsive status epilepticus: a retrospective 5‑year review. Seizure 16:305–312PubMed Hussain N, Appleton R, Thorburn K et al (2007) Etiology, course and outcome of children admitted to pediatric intensive care with convulsive status epilepticus: a retrospective 5‑year review. Seizure 16:305–312PubMed
8.
Zurück zum Zitat Glauser T, Shinnar S, Gloss D et al (2016) Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report of the guideline committee of the American epilepsy society. Epilepsy Curr 16:48–61PubMedPubMedCentral Glauser T, Shinnar S, Gloss D et al (2016) Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report of the guideline committee of the American epilepsy society. Epilepsy Curr 16:48–61PubMedPubMedCentral
9.
Zurück zum Zitat DeLorenzo RJ, Hauser WA, Towne AR, Boggs JG, Pellock JM, Penberthy L, Garnett L, Fortner CA, Ko D (1996) A prospective, population-based epidemiologic study of Status epilepticus in Richmond, Virginia. Neurology 46:1029–1035PubMed DeLorenzo RJ, Hauser WA, Towne AR, Boggs JG, Pellock JM, Penberthy L, Garnett L, Fortner CA, Ko D (1996) A prospective, population-based epidemiologic study of Status epilepticus in Richmond, Virginia. Neurology 46:1029–1035PubMed
10.
Zurück zum Zitat van Baalen A, Häusler M, Boor R, Rohr A, Sperner J, Kurlemann G, Panzer A, Stephani U, Kluger G (2010) Febrile infection-related epilepsy syndrome (FIRES): a nonencephalitic encephalopathy in childhood. Epilepsia 51:1323–1328PubMed van Baalen A, Häusler M, Boor R, Rohr A, Sperner J, Kurlemann G, Panzer A, Stephani U, Kluger G (2010) Febrile infection-related epilepsy syndrome (FIRES): a nonencephalitic encephalopathy in childhood. Epilepsia 51:1323–1328PubMed
11.
Zurück zum Zitat Costello DJ, Kilbride RD, Cole AJ (2009) Cryptogenic New Onset Refractory Status Epilepticus (NORSE) in adults-Infectious or not? J Neurol Sci 277:26–31PubMed Costello DJ, Kilbride RD, Cole AJ (2009) Cryptogenic New Onset Refractory Status Epilepticus (NORSE) in adults-Infectious or not? J Neurol Sci 277:26–31PubMed
12.
Zurück zum Zitat Pujar SS, Martinos MM, Cortina-Borja M et al (2018) Long-term prognosis after childhood convulsive status epilepticus: a prospective cohort study. Lancet Child Adolesc Health 2:103–111PubMed Pujar SS, Martinos MM, Cortina-Borja M et al (2018) Long-term prognosis after childhood convulsive status epilepticus: a prospective cohort study. Lancet Child Adolesc Health 2:103–111PubMed
13.
Zurück zum Zitat Pujar SS, Neville BG, Scott RC, Chin RF (2011) North London Epilepsy Research Network, Death within 8 years after childhood convulsive status epilepticus: a population-based study. Brain 134:2819–2827PubMedPubMedCentral Pujar SS, Neville BG, Scott RC, Chin RF (2011) North London Epilepsy Research Network, Death within 8 years after childhood convulsive status epilepticus: a population-based study. Brain 134:2819–2827PubMedPubMedCentral
14.
Zurück zum Zitat Alldredge BK, Wall DB, Ferriero DM (1995) Effect of prehospital treatment on the outcome of status epilepticus in children. Pediatr Neurol 12:213–216PubMed Alldredge BK, Wall DB, Ferriero DM (1995) Effect of prehospital treatment on the outcome of status epilepticus in children. Pediatr Neurol 12:213–216PubMed
15.
Zurück zum Zitat Chin RF, Neville BG, Peckham C, Wade A, Bedford H, Scott RC (2008) Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 7:696–703PubMedPubMedCentral Chin RF, Neville BG, Peckham C, Wade A, Bedford H, Scott RC (2008) Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 7:696–703PubMedPubMedCentral
16.
Zurück zum Zitat Raspall-Chaure M, Chin RF, Neville BG, Scott RC (2006) Outcome of pediatric convulsive status epilepticus: a systematic review. Lancet Neurol 5:769–779PubMed Raspall-Chaure M, Chin RF, Neville BG, Scott RC (2006) Outcome of pediatric convulsive status epilepticus: a systematic review. Lancet Neurol 5:769–779PubMed
18.
Zurück zum Zitat Lewena S, Pennington V, Acworth J et al (2009) Emergency management of pediatric convulsive status epilepticus: a multicenter study of 542 patients. Pediatr Emerg Care 25:83–87PubMed Lewena S, Pennington V, Acworth J et al (2009) Emergency management of pediatric convulsive status epilepticus: a multicenter study of 542 patients. Pediatr Emerg Care 25:83–87PubMed
19.
Zurück zum Zitat Chamberlain JM, Okada P, Holsti M, Mahajan P, Brown KM, Vance C et al (2014) Pediatric emergency care applied research network (PECARN). Lorazepam vs diazepam for pediatric status epilepticus: a randomized clinical trial. JAMA 311:1652–1660PubMed Chamberlain JM, Okada P, Holsti M, Mahajan P, Brown KM, Vance C et al (2014) Pediatric emergency care applied research network (PECARN). Lorazepam vs diazepam for pediatric status epilepticus: a randomized clinical trial. JAMA 311:1652–1660PubMed
20.
Zurück zum Zitat Silbergleit R, Durkalski V, Lowenstein D, Conwit R, Pancioli A, Palesch Y et al (2012) Intramuscular versus intravenous therapy for prehospital status epilepticus. N Engl J Med 366:591–600PubMedPubMedCentral Silbergleit R, Durkalski V, Lowenstein D, Conwit R, Pancioli A, Palesch Y et al (2012) Intramuscular versus intravenous therapy for prehospital status epilepticus. N Engl J Med 366:591–600PubMedPubMedCentral
22.
Zurück zum Zitat Abend NS, Huh JW, Helfaer MA, Dlugos DJ (2008) Anticonvulsant medications in the pediatric emergency room and intensive care unit. Pediatr Emerg Care 24:705–718PubMed Abend NS, Huh JW, Helfaer MA, Dlugos DJ (2008) Anticonvulsant medications in the pediatric emergency room and intensive care unit. Pediatr Emerg Care 24:705–718PubMed
23.
Zurück zum Zitat Lyttle MD, Rainford NEA, Gamble C, Messahel S, Humphreys A, Hickey H et al (2019) Paediatric Emergency Research in the United Kingdom Ireland (PERUKI) collaborative. Levetiracetam versus phenytoin for second-line treatment of paediatric convulsive status epilepticus (EcLiPSE): a multicentre, open-label, randomised trial. Lancet 393:2125–2134PubMedPubMedCentral Lyttle MD, Rainford NEA, Gamble C, Messahel S, Humphreys A, Hickey H et al (2019) Paediatric Emergency Research in the United Kingdom Ireland (PERUKI) collaborative. Levetiracetam versus phenytoin for second-line treatment of paediatric convulsive status epilepticus (EcLiPSE): a multicentre, open-label, randomised trial. Lancet 393:2125–2134PubMedPubMedCentral
24.
Zurück zum Zitat Dalziel SR, Borland ML, Furyk J, Bonisch M, Neutze J, Donath S et al (2019) PREDICT research network. Levetiracetam versus phenytoin for second-line treatment of convulsive status epilepticus in children (ConSEPT): an open-label, multicentre, randomised controlled trial. Lancet 393:2135–2145PubMed Dalziel SR, Borland ML, Furyk J, Bonisch M, Neutze J, Donath S et al (2019) PREDICT research network. Levetiracetam versus phenytoin for second-line treatment of convulsive status epilepticus in children (ConSEPT): an open-label, multicentre, randomised controlled trial. Lancet 393:2135–2145PubMed
25.
Zurück zum Zitat Kapur J, Elm J, Chamberlain JM, Barsan W, Cloyd J, Lowenstein D et al (2019) NETT and PECARN investigators. Randomized trial of three anticonvulsant medications for status epilepticus. N Engl J Med 381:2103–2113PubMedPubMedCentral Kapur J, Elm J, Chamberlain JM, Barsan W, Cloyd J, Lowenstein D et al (2019) NETT and PECARN investigators. Randomized trial of three anticonvulsant medications for status epilepticus. N Engl J Med 381:2103–2113PubMedPubMedCentral
26.
Zurück zum Zitat Griesdale DE, Bosma TL, Kurth T et al (2008) Complications of endotracheal intubation in the critically ill. Int Care Med 34:1835–1842 Griesdale DE, Bosma TL, Kurth T et al (2008) Complications of endotracheal intubation in the critically ill. Int Care Med 34:1835–1842
27.
Zurück zum Zitat Fernández IS, Abend NS, Agadi S et al (2014) Gaps and opportunities in refractory status epilepticus research in children: a multi-center approach by the Pediatric Status Epilepticus Research Group (pSERG). Seizure 23:87–97 Fernández IS, Abend NS, Agadi S et al (2014) Gaps and opportunities in refractory status epilepticus research in children: a multi-center approach by the Pediatric Status Epilepticus Research Group (pSERG). Seizure 23:87–97
28.
Zurück zum Zitat Abend NS, Dlugos DJ (2008) Treatment of refractory status epilepticus: literature review and a proposed protocol. Pediatr Neurol 38:377–390PubMed Abend NS, Dlugos DJ (2008) Treatment of refractory status epilepticus: literature review and a proposed protocol. Pediatr Neurol 38:377–390PubMed
29.
Zurück zum Zitat Bleck TP (2005) Refractory status epilepticus. Curr Opin Crit Care 11:117–120PubMed Bleck TP (2005) Refractory status epilepticus. Curr Opin Crit Care 11:117–120PubMed
30.
Zurück zum Zitat Lowenstein DH (2006) The management of refractory status epilepticus: an update. Epilepsia 47(Suppl 1):S35–S40 Lowenstein DH (2006) The management of refractory status epilepticus: an update. Epilepsia 47(Suppl 1):S35–S40
31.
Zurück zum Zitat Meierkord H, Boon P, Engelsen B et al (2010) European Federation of Neurological Societies: EFNS guideline on the management of status epilepticus in adults. Eur J Neurol 17:348–355PubMed Meierkord H, Boon P, Engelsen B et al (2010) European Federation of Neurological Societies: EFNS guideline on the management of status epilepticus in adults. Eur J Neurol 17:348–355PubMed
32.
Zurück zum Zitat Pediatric Status Epilepticus Research Group, Tasker RC, Goodkin HP, Sánchez-Fernández I, Chapman KE, Abend NS, Arya R et al (2016) Refractory status epilepticus in children: intention to treat with continuous infusions of midazolam and pentobarbital. Pediatr Crit Care Med 17:968–975 Pediatric Status Epilepticus Research Group, Tasker RC, Goodkin HP, Sánchez-Fernández I, Chapman KE, Abend NS, Arya R et al (2016) Refractory status epilepticus in children: intention to treat with continuous infusions of midazolam and pentobarbital. Pediatr Crit Care Med 17:968–975
33.
Zurück zum Zitat Wilkes R, Tasker RC. (2014) Intensive care treatment of uncontrolled status epilepticus in children: systematic literature search of midazolam and anesthetic therapies. Pediatr Crit Care Med 15:632–639PubMed Wilkes R, Tasker RC. (2014) Intensive care treatment of uncontrolled status epilepticus in children: systematic literature search of midazolam and anesthetic therapies. Pediatr Crit Care Med 15:632–639PubMed
34.
Zurück zum Zitat Rogawski MA, Löscher W (2004) The neurobiology of antiepileptic drugs. Nat Rev Neurosci 5:553–564PubMed Rogawski MA, Löscher W (2004) The neurobiology of antiepileptic drugs. Nat Rev Neurosci 5:553–564PubMed
35.
Zurück zum Zitat Kim SJ, Lee DY, Kim JS (2001) Neurologic outcomes of pediatric epileptic patients with pentobarbital coma. Pediatr Neurol 25:217–220PubMed Kim SJ, Lee DY, Kim JS (2001) Neurologic outcomes of pediatric epileptic patients with pentobarbital coma. Pediatr Neurol 25:217–220PubMed
36.
Zurück zum Zitat Rossetti AO, Reichhart MD, Schaller MD, Despland PA, Bogousslavsky J (2004) Propofol treatment of refractory status epilepticus: a study of 31 episodes. Epilepsia 45:757–763PubMed Rossetti AO, Reichhart MD, Schaller MD, Despland PA, Bogousslavsky J (2004) Propofol treatment of refractory status epilepticus: a study of 31 episodes. Epilepsia 45:757–763PubMed
37.
Zurück zum Zitat Parviainen I, Uusaro A, Kalviainen R, Mervaala E, Ruokonen E (2006) Propofol in the treatment of refractory status epilepticus. Intensive Care Med 32:1075–1079PubMed Parviainen I, Uusaro A, Kalviainen R, Mervaala E, Ruokonen E (2006) Propofol in the treatment of refractory status epilepticus. Intensive Care Med 32:1075–1079PubMed
38.
Zurück zum Zitat Kam PC, Cardone D (2007) Propofol infusion syndrome. Anaesthesia 62:690–701PubMed Kam PC, Cardone D (2007) Propofol infusion syndrome. Anaesthesia 62:690–701PubMed
39.
Zurück zum Zitat Augoustides JG, Culp KE, Ochroch AE, Milas BL (2005) Total suppression of cerebral activity by thiopental mimicking propofol infusion syndrome: a fatal common pathway? Anesth Analg 100:1865PubMed Augoustides JG, Culp KE, Ochroch AE, Milas BL (2005) Total suppression of cerebral activity by thiopental mimicking propofol infusion syndrome: a fatal common pathway? Anesth Analg 100:1865PubMed
40.
Zurück zum Zitat Felmet K, Nguyen T, Clark RS et al (2003) The FDA warning against prolonged sedation with propofol in children remains warranted. Pediatrics 112:1002–1003PubMed Felmet K, Nguyen T, Clark RS et al (2003) The FDA warning against prolonged sedation with propofol in children remains warranted. Pediatrics 112:1002–1003PubMed
41.
Zurück zum Zitat McHugh P (1991) Acute choreoathetoid reaction to propofol. Anaesthesia 46:425PubMed McHugh P (1991) Acute choreoathetoid reaction to propofol. Anaesthesia 46:425PubMed
42.
Zurück zum Zitat Chidambaran V, Costandi A, D’Mello A (2015) Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs 29:543–563PubMedPubMedCentral Chidambaran V, Costandi A, D’Mello A (2015) Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs 29:543–563PubMedPubMedCentral
43.
Zurück zum Zitat Krajčová A, Waldauf P, Anděl M et al (2015) Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care 19:398PubMedPubMedCentral Krajčová A, Waldauf P, Anděl M et al (2015) Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care 19:398PubMedPubMedCentral
44.
Zurück zum Zitat Hemphill S, McMenamin L, Bellamy MC et al (2019) Propofol infusion syndrome: a structured literature review and analysis of published case reports. Br J Anaesth 122:448–459PubMedPubMedCentral Hemphill S, McMenamin L, Bellamy MC et al (2019) Propofol infusion syndrome: a structured literature review and analysis of published case reports. Br J Anaesth 122:448–459PubMedPubMedCentral
45.
Zurück zum Zitat Chidambaran V, Costandi A, D’Mello A (2015) Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs 29:543–563PubMedPubMedCentral Chidambaran V, Costandi A, D’Mello A (2015) Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs 29:543–563PubMedPubMedCentral
46.
Zurück zum Zitat Krajčová A, Waldauf P, Anděl M et al (2015) Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care 19:398PubMedPubMedCentral Krajčová A, Waldauf P, Anděl M et al (2015) Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care 19:398PubMedPubMedCentral
47.
Zurück zum Zitat Baumeister FA, Oberhoffer R, Liebhaber GM, Kunkel J, Eberhardt J, Holthausen H, Peters J (2004) Fatal propofol infusion syndrome in association with ketogenic diet. Neuropediatrics 35:250–252PubMed Baumeister FA, Oberhoffer R, Liebhaber GM, Kunkel J, Eberhardt J, Holthausen H, Peters J (2004) Fatal propofol infusion syndrome in association with ketogenic diet. Neuropediatrics 35:250–252PubMed
48.
Zurück zum Zitat van Gestel JP, Blusse van Oud-Alblas HJ, Malingre M, Ververs FF, Braun KP, van Nieuwenhuizen O (2005) Propofol and thiopental for refractory status epilepticus in children. Neurology 65:591–592PubMed van Gestel JP, Blusse van Oud-Alblas HJ, Malingre M, Ververs FF, Braun KP, van Nieuwenhuizen O (2005) Propofol and thiopental for refractory status epilepticus in children. Neurology 65:591–592PubMed
49.
Zurück zum Zitat Shorvon S, Ferlisi M (2011) The treatment of super-refractory status epilepticus: a critical review of available therapies and a clinical treatment protocol. Brain 134:2802–2818PubMed Shorvon S, Ferlisi M (2011) The treatment of super-refractory status epilepticus: a critical review of available therapies and a clinical treatment protocol. Brain 134:2802–2818PubMed
50.
Zurück zum Zitat Mayer SA, Claassen J, Lokin J, Mendelsohn F, Dennis LJ, Fitzsimmons BF (2002) Refractory status epilepticus: frequency, risk factors, and impact on outcome. Arch Neurol 59:205–210PubMed Mayer SA, Claassen J, Lokin J, Mendelsohn F, Dennis LJ, Fitzsimmons BF (2002) Refractory status epilepticus: frequency, risk factors, and impact on outcome. Arch Neurol 59:205–210PubMed
51.
Zurück zum Zitat Loscher W (2007) Mechanisms of drug resistance in status epilepticus. Epilepsia 48(Suppl. 8):S74–S77 Loscher W (2007) Mechanisms of drug resistance in status epilepticus. Epilepsia 48(Suppl. 8):S74–S77
52.
Zurück zum Zitat Sofou K, Kristjansdòttir R, Papachatzakis NE et al (2009) Management of prolonged seizures and status epilepticus in childhood: a systematic review. J Child Neurol 24:918–926PubMed Sofou K, Kristjansdòttir R, Papachatzakis NE et al (2009) Management of prolonged seizures and status epilepticus in childhood: a systematic review. J Child Neurol 24:918–926PubMed
53.
Zurück zum Zitat Raspall-Chaure M, Chin RF, Neville BG et al (2006) Outcome of pediatric convulsive status epilepticus: a systematic review. Lancet Neurol 5:769–779PubMed Raspall-Chaure M, Chin RF, Neville BG et al (2006) Outcome of pediatric convulsive status epilepticus: a systematic review. Lancet Neurol 5:769–779PubMed
54.
Zurück zum Zitat Vasquez A, Farias-Moeller R, Sánchez-Fernández I, Abend N, Amengual-Gual M, Anderson A et al (2021) Super-refractory status epilepticus in children: a retrospective cohort study. Pediatr Crit Care Med 22:e613–e625PubMed Vasquez A, Farias-Moeller R, Sánchez-Fernández I, Abend N, Amengual-Gual M, Anderson A et al (2021) Super-refractory status epilepticus in children: a retrospective cohort study. Pediatr Crit Care Med 22:e613–e625PubMed
55.
Zurück zum Zitat Van Matre ET, Cook AM (2016) Steady-state pharmacokinetic simulation of intermittent vs. continuous infusion valproic acid therapy in non-critically ill and critically ill patients. Neurol Res 38:786–791PubMed Van Matre ET, Cook AM (2016) Steady-state pharmacokinetic simulation of intermittent vs. continuous infusion valproic acid therapy in non-critically ill and critically ill patients. Neurol Res 38:786–791PubMed
56.
Zurück zum Zitat Uberall MA, Trollmann R, Wunsiedler U et al (2000) Intravenous valproate in pediatric epilepsy patients with refractory status epilepticus. Neurology 54:2188–2189PubMed Uberall MA, Trollmann R, Wunsiedler U et al (2000) Intravenous valproate in pediatric epilepsy patients with refractory status epilepticus. Neurology 54:2188–2189PubMed
57.
Zurück zum Zitat Zeiler FA, Teitelbaum J, Gillman LM, West M (2014) NMDA antagonists for refractory seizures. Neurocrit Care 20:502–513PubMed Zeiler FA, Teitelbaum J, Gillman LM, West M (2014) NMDA antagonists for refractory seizures. Neurocrit Care 20:502–513PubMed
59.
Zurück zum Zitat Fujikawa DG (2019) Starting ketamine for neuroprotection earlier than its current use as an anesthetic/antiepileptic drug late in refractory status epilepticus. Epilepsia 60:373–380PubMed Fujikawa DG (2019) Starting ketamine for neuroprotection earlier than its current use as an anesthetic/antiepileptic drug late in refractory status epilepticus. Epilepsia 60:373–380PubMed
60.
Zurück zum Zitat Carroll CL, Spinella PC, Corsi JP, Stoltz P, Zucker AR (2010) Emergent endotracheal intubations in children: be careful if it’s late when you intubate. Pediatr Crit Care Med 11:343–348PubMed Carroll CL, Spinella PC, Corsi JP, Stoltz P, Zucker AR (2010) Emergent endotracheal intubations in children: be careful if it’s late when you intubate. Pediatr Crit Care Med 11:343–348PubMed
61.
Zurück zum Zitat Griesdale DE, Bosma TL, Kurth T, Isac G, Chittock DR (2008) Complications of endotracheal intubation in the critically ill. Int Care Med 34:1835–1842 Griesdale DE, Bosma TL, Kurth T, Isac G, Chittock DR (2008) Complications of endotracheal intubation in the critically ill. Int Care Med 34:1835–1842
63.
Zurück zum Zitat Rosati A, Ilvento L, L’Erario M et al (2016) Efficacy of ketamine in refractory convulsive status epilepticus in children: a protocol for a sequential design, multi centre, randomized, controlled, open-label, non-profit trial (KETASER01). BMJ Open 6:e1156 Rosati A, Ilvento L, L’Erario M et al (2016) Efficacy of ketamine in refractory convulsive status epilepticus in children: a protocol for a sequential design, multi centre, randomized, controlled, open-label, non-profit trial (KETASER01). BMJ Open 6:e1156
64.
Zurück zum Zitat Langmoen IA, Hegstad E, Berg-Johnsen J (1992) An experimental study of the effect of isoflurane on epileptiform bursts. Epilepsy Res 11:153–157PubMed Langmoen IA, Hegstad E, Berg-Johnsen J (1992) An experimental study of the effect of isoflurane on epileptiform bursts. Epilepsy Res 11:153–157PubMed
65.
Zurück zum Zitat Eger EI 2nd (1994) New inhaled anesthetics. Anesthesiology 80:906–922PubMed Eger EI 2nd (1994) New inhaled anesthetics. Anesthesiology 80:906–922PubMed
66.
Zurück zum Zitat Mirsattari SM, Sharpe MD, Young GB (2004) Treatment of refractory status epilepticus with inhalational anesthetic agents isoflurane and desflurane. Arch Neurol 61:1254–1259PubMed Mirsattari SM, Sharpe MD, Young GB (2004) Treatment of refractory status epilepticus with inhalational anesthetic agents isoflurane and desflurane. Arch Neurol 61:1254–1259PubMed
67.
Zurück zum Zitat Kofke WA, Young RS, Davis P et al (1989) Isoflurane for refractory status epilepticus: a clinical series. Anesthesiology 71:653–659PubMed Kofke WA, Young RS, Davis P et al (1989) Isoflurane for refractory status epilepticus: a clinical series. Anesthesiology 71:653–659PubMed
68.
Zurück zum Zitat Fugate JE, Burns JD, Wijdicks EF, Warner DO, Jankowski CJ, Rabinstein AA (2010) Prolonged high-dose isoflurane for refractory status epilepticus: is it safe? Anesth Analg 111:1520–1524PubMed Fugate JE, Burns JD, Wijdicks EF, Warner DO, Jankowski CJ, Rabinstein AA (2010) Prolonged high-dose isoflurane for refractory status epilepticus: is it safe? Anesth Analg 111:1520–1524PubMed
69.
Zurück zum Zitat Ikeda KM, Connors R, Lee DH, Khandji AG, Claassen J, Young GB (2017) Isoflurane use in the treatment of super-refractory status epilepticus is associated with hippocampal changes on MRI. Neurocrit Care 26:420–427PubMed Ikeda KM, Connors R, Lee DH, Khandji AG, Claassen J, Young GB (2017) Isoflurane use in the treatment of super-refractory status epilepticus is associated with hippocampal changes on MRI. Neurocrit Care 26:420–427PubMed
70.
Zurück zum Zitat Mikati MA, Kurdi R, El-Khoury Z et al (2010) Intravenous immunoglobulin therapy in intractable childhood epilepsy: open-label study and review of the literature. Epilepsy Behav 17:90–94PubMed Mikati MA, Kurdi R, El-Khoury Z et al (2010) Intravenous immunoglobulin therapy in intractable childhood epilepsy: open-label study and review of the literature. Epilepsy Behav 17:90–94PubMed
71.
Zurück zum Zitat Sinclair DB (2003) Prednisone therapy in pediatric epilepsy. Pediatr Neurol 28:194–198PubMed Sinclair DB (2003) Prednisone therapy in pediatric epilepsy. Pediatr Neurol 28:194–198PubMed
72.
Zurück zum Zitat Marescaux C, Hirsch E, Finck S et al (1990) Landau-Kleffner syndrome: a pharmacologic study of five cases. Epilepsia 31:768–777PubMed Marescaux C, Hirsch E, Finck S et al (1990) Landau-Kleffner syndrome: a pharmacologic study of five cases. Epilepsia 31:768–777PubMed
73.
Zurück zum Zitat Villani F, Avanzini G (2002) The use of immunoglobulins in the treatment of human epilepsy. Neurol Sci 23(Suppl 1):S33–S37PubMed Villani F, Avanzini G (2002) The use of immunoglobulins in the treatment of human epilepsy. Neurol Sci 23(Suppl 1):S33–S37PubMed
74.
Zurück zum Zitat Vezzani A, French J, Bartfai T et al (2011) The role of inflammation in epilepsy. Nat Rev Neurol 7:31–40PubMed Vezzani A, French J, Bartfai T et al (2011) The role of inflammation in epilepsy. Nat Rev Neurol 7:31–40PubMed
75.
Zurück zum Zitat Lopinto-Khoury C, Sperling MR (2013) Autoimmune status epilepticus. Curr Treat Options Neurol 15:545–556PubMed Lopinto-Khoury C, Sperling MR (2013) Autoimmune status epilepticus. Curr Treat Options Neurol 15:545–556PubMed
76.
Zurück zum Zitat Graus F, Titulaer MJ, Balu R, Benseler S, Bien CG, Cellucci T et al (2016) A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 15:391–404PubMedPubMedCentral Graus F, Titulaer MJ, Balu R, Benseler S, Bien CG, Cellucci T et al (2016) A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 15:391–404PubMedPubMedCentral
77.
Zurück zum Zitat Ferlisi M, Shorvon S (2012) The outcome of therapies in refractory and super-refractory convulsive status epilepticus and recommendations for therapy. Brain 135:2314–2328PubMed Ferlisi M, Shorvon S (2012) The outcome of therapies in refractory and super-refractory convulsive status epilepticus and recommendations for therapy. Brain 135:2314–2328PubMed
78.
Zurück zum Zitat Rosenthal ES, Claassen J, Wainwright MS, Husain AM, Vaitkevicius H, Raines S et al (2017) Brexanolone as adjunctive therapy in super-refractory status epilepticus. Ann Neurol 82:342–352PubMedPubMedCentral Rosenthal ES, Claassen J, Wainwright MS, Husain AM, Vaitkevicius H, Raines S et al (2017) Brexanolone as adjunctive therapy in super-refractory status epilepticus. Ann Neurol 82:342–352PubMedPubMedCentral
79.
Zurück zum Zitat Gaspard N, Hirsch LJ, Sculier C, Loddenkemper T, van Baalen A, Lancrenon J et al (2018) New-onset refractory status epilepticus (NORSE) and febrile infection-related epilepsy syndrome (FIRES): state of the art and perspectives. Epilepsia 59:745–752PubMed Gaspard N, Hirsch LJ, Sculier C, Loddenkemper T, van Baalen A, Lancrenon J et al (2018) New-onset refractory status epilepticus (NORSE) and febrile infection-related epilepsy syndrome (FIRES): state of the art and perspectives. Epilepsia 59:745–752PubMed
80.
Zurück zum Zitat Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2017) Intravenous immunoglobulins for refractory status epilepticus, part I: A scoping systematic review of the adult literature. Seizure 45:172–180PubMed Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2017) Intravenous immunoglobulins for refractory status epilepticus, part I: A scoping systematic review of the adult literature. Seizure 45:172–180PubMed
81.
Zurück zum Zitat Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2016) Plasmapheresis for refractory status epilepticus, part I: A scoping systematic review of the adult literature. Seizure 43:14–22PubMed Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2016) Plasmapheresis for refractory status epilepticus, part I: A scoping systematic review of the adult literature. Seizure 43:14–22PubMed
82.
Zurück zum Zitat Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2016) Plasmapheresis for refractory status epilepticus part II: a scoping systematic review of the pediatric literature. Seizure 43:61–68PubMed Zeiler FA, Matuszczak M, Teitelbaum J, Kazina CJ, Gillman LM (2016) Plasmapheresis for refractory status epilepticus part II: a scoping systematic review of the pediatric literature. Seizure 43:61–68PubMed
83.
Zurück zum Zitat Arya R, Rotenberg A (2019) Dietary, immunological, surgical, and other emerging treatments for pediatric refractory status epilepticus. Seizure 68:89–96PubMed Arya R, Rotenberg A (2019) Dietary, immunological, surgical, and other emerging treatments for pediatric refractory status epilepticus. Seizure 68:89–96PubMed
84.
Zurück zum Zitat Likhodii SS, Serbanescu I, Cortez MA, Murphy P, Snead OC 3rd, Burnham WM (2003) Anticonvulsant properties of acetone, a brain ketone elevated by the ketogenic diet. Ann Neurol 54:219–226PubMed Likhodii SS, Serbanescu I, Cortez MA, Murphy P, Snead OC 3rd, Burnham WM (2003) Anticonvulsant properties of acetone, a brain ketone elevated by the ketogenic diet. Ann Neurol 54:219–226PubMed
85.
Zurück zum Zitat Rho JM, Anderson GD, Donevan SD, White HS (2002) Acetoacetate, acetone, and dibenzylamine (a contaminant in l‑(+)-beta-hydroxybutyrate) exhibit direct anticonvulsant actions in vivo. Epilepsia 43:358–361PubMed Rho JM, Anderson GD, Donevan SD, White HS (2002) Acetoacetate, acetone, and dibenzylamine (a contaminant in l‑(+)-beta-hydroxybutyrate) exhibit direct anticonvulsant actions in vivo. Epilepsia 43:358–361PubMed
86.
Zurück zum Zitat Arya R, Peariso K, Gainza-Lein M, Harvey J, Bergin A, Brenton JN et al (2018) Efficacy and safety of ketogenic diet for treatment of pediatric convulsive refractory status epilepticus. Epilepsy Res 144:1–6PubMed Arya R, Peariso K, Gainza-Lein M, Harvey J, Bergin A, Brenton JN et al (2018) Efficacy and safety of ketogenic diet for treatment of pediatric convulsive refractory status epilepticus. Epilepsy Res 144:1–6PubMed
87.
Zurück zum Zitat Appavu B, Vanatta L, Condie J, Kerrigan JF, Jarrar R (2016) Ketogenic diet treatment for pediatric super-refractory status epilepticus. Seizure 41:62–65PubMed Appavu B, Vanatta L, Condie J, Kerrigan JF, Jarrar R (2016) Ketogenic diet treatment for pediatric super-refractory status epilepticus. Seizure 41:62–65PubMed
88.
Zurück zum Zitat Caraballo RH, Flesler S, Armeno M, Fortini S, Agustinho A, Mestre G et al (2014) Ketogenic diet in pediatric patients with refractory focal status epilepticus. Epilepsy Res 108:1912–1916PubMed Caraballo RH, Flesler S, Armeno M, Fortini S, Agustinho A, Mestre G et al (2014) Ketogenic diet in pediatric patients with refractory focal status epilepticus. Epilepsy Res 108:1912–1916PubMed
89.
Zurück zum Zitat Schoeler NE, Simpson Z, Zhou R, Pujar S, Eltze C, Cross JH (2021) Dietary management of children with super-refractory status Epilepticus: a systematic review and experience in a single UK tertiary centre. Front Neurol 12:643105PubMedPubMedCentral Schoeler NE, Simpson Z, Zhou R, Pujar S, Eltze C, Cross JH (2021) Dietary management of children with super-refractory status Epilepticus: a systematic review and experience in a single UK tertiary centre. Front Neurol 12:643105PubMedPubMedCentral
90.
Zurück zum Zitat Cobo NH, Sankar R, Murata KK, Sewak SL, Kezele MA, Matsumoto JH (2015) The ketogenic diet as broad-spectrum treatment for super-refractory pediatric status epilepticus: challenges in implementation in the pediatric and neonatal intensive care units. J Child Neurol 30:259–266PubMed Cobo NH, Sankar R, Murata KK, Sewak SL, Kezele MA, Matsumoto JH (2015) The ketogenic diet as broad-spectrum treatment for super-refractory pediatric status epilepticus: challenges in implementation in the pediatric and neonatal intensive care units. J Child Neurol 30:259–266PubMed
91.
Zurück zum Zitat Niquet J, Gezalian M, Baldwin R, Wasterlain CG (2015) Neuroprotective effects of deep hypothermia in refractory status epilepticus. Ann Clin Transl Neurol 2:1105–1115PubMedPubMedCentral Niquet J, Gezalian M, Baldwin R, Wasterlain CG (2015) Neuroprotective effects of deep hypothermia in refractory status epilepticus. Ann Clin Transl Neurol 2:1105–1115PubMedPubMedCentral
92.
Zurück zum Zitat Lundgren J, Smith ML, Blennow G, Siesjö BK (1994) Hyperthermia aggravates and hypothermia ameliorates epileptic brain damage. Exp Brain Res 99:43–55PubMed Lundgren J, Smith ML, Blennow G, Siesjö BK (1994) Hyperthermia aggravates and hypothermia ameliorates epileptic brain damage. Exp Brain Res 99:43–55PubMed
93.
Zurück zum Zitat Hrncic D, Vucevic D, Rasic A, Radosavljevic T, Mladenovic D, Susic V et al (2007) Moderate body hypothermia alleviates behavioral and EEG manifestations of audiogenic seizures in metaphit-treated rats. Can J Physiol Pharmacol 85:1032–1037PubMed Hrncic D, Vucevic D, Rasic A, Radosavljevic T, Mladenovic D, Susic V et al (2007) Moderate body hypothermia alleviates behavioral and EEG manifestations of audiogenic seizures in metaphit-treated rats. Can J Physiol Pharmacol 85:1032–1037PubMed
94.
Zurück zum Zitat Corry JJ, Dhar R, Murphy T, Diringer MN (2008) Hypothermia for refractory status epilepticus. Neurocrit Care 9:189–197PubMed Corry JJ, Dhar R, Murphy T, Diringer MN (2008) Hypothermia for refractory status epilepticus. Neurocrit Care 9:189–197PubMed
95.
Zurück zum Zitat HYBERNATUS Study Group, Legriel S, Lemiale V, Schenck M, Chelly J, Laurent V, Daviaud F et al (2016) Hypothermia for neuroprotection in convulsive status epilepticus. N Engl J Med 375:2457–2467 HYBERNATUS Study Group, Legriel S, Lemiale V, Schenck M, Chelly J, Laurent V, Daviaud F et al (2016) Hypothermia for neuroprotection in convulsive status epilepticus. N Engl J Med 375:2457–2467
96.
Zurück zum Zitat Orlowski JP, Erenberg G, Lueders H, Cruse RP (1984) Hypothermia and barbiturate coma for refractory status epilepticus. Crit Care Med 12:367–372PubMed Orlowski JP, Erenberg G, Lueders H, Cruse RP (1984) Hypothermia and barbiturate coma for refractory status epilepticus. Crit Care Med 12:367–372PubMed
97.
Zurück zum Zitat Elting JW, Naalt Jv, Fock JM (2010) Mild hypothermia for refractory focal status epilepticus in an infant with hemimegalencephaly. Eur J Paediatr Neurol 14:452–455PubMed Elting JW, Naalt Jv, Fock JM (2010) Mild hypothermia for refractory focal status epilepticus in an infant with hemimegalencephaly. Eur J Paediatr Neurol 14:452–455PubMed
98.
Zurück zum Zitat Lin JJ, Lin KL, Hsia SH, Wang HS, CHEESE Study Group (2012) Therapeutic hypothermia for febrile infection-related epilepsy syndrome in two patients. Pediatr Neurol 47:448–450PubMed Lin JJ, Lin KL, Hsia SH, Wang HS, CHEESE Study Group (2012) Therapeutic hypothermia for febrile infection-related epilepsy syndrome in two patients. Pediatr Neurol 47:448–450PubMed
99.
Zurück zum Zitat Guilliams K, Rosen M, Buttram S et al (2013) Hypothermia for pediatric refractory status epilepticus. Epilepsia 54:1586–1594PubMedPubMedCentral Guilliams K, Rosen M, Buttram S et al (2013) Hypothermia for pediatric refractory status epilepticus. Epilepsia 54:1586–1594PubMedPubMedCentral
100.
Zurück zum Zitat Mills PB, Struys E, Jakobs C, Plecko B, Baxter P, Baumgartner M et al (2006) Mutations in antiquitin in individuals with pyridoxine-dependent seizures. Nat Med 12:307–309PubMed Mills PB, Struys E, Jakobs C, Plecko B, Baxter P, Baumgartner M et al (2006) Mutations in antiquitin in individuals with pyridoxine-dependent seizures. Nat Med 12:307–309PubMed
101.
Zurück zum Zitat Caksen H, Odabas D, Erol M, Anlar O, Tuncer O, Atas B (2003) Do not overlook acute isoniazid poisoning in children with status epilepticus. J Child Neurol 18:142–143PubMed Caksen H, Odabas D, Erol M, Anlar O, Tuncer O, Atas B (2003) Do not overlook acute isoniazid poisoning in children with status epilepticus. J Child Neurol 18:142–143PubMed
102.
Zurück zum Zitat Minns AB, Ghafouri N, Clark RF (2010) Isoniazid-induced status epilepticus in a pediatric patient after inadequate pyridoxine therapy. Pediatr Emerg Care 26:380–381PubMed Minns AB, Ghafouri N, Clark RF (2010) Isoniazid-induced status epilepticus in a pediatric patient after inadequate pyridoxine therapy. Pediatr Emerg Care 26:380–381PubMed
103.
Zurück zum Zitat Which anticonvulsant for women with eclampsia? Evidence from the collaborative eclampsia trial. Lancet 1995; 345:1455–1463. Which anticonvulsant for women with eclampsia? Evidence from the collaborative eclampsia trial. Lancet 1995; 345:1455–1463.
104.
Zurück zum Zitat Visser NA, Braun KP, Leijten FS, van Nieuwenhuizen O, Wokke JH, van den Bergh WM (2011) Magnesium treatment for patients with refractory status epilepticus due to POLG1-mutations. J Neurol 258:218–222PubMed Visser NA, Braun KP, Leijten FS, van Nieuwenhuizen O, Wokke JH, van den Bergh WM (2011) Magnesium treatment for patients with refractory status epilepticus due to POLG1-mutations. J Neurol 258:218–222PubMed
105.
Zurück zum Zitat Zeiler FA, Matuszczak M, Teitelbaum J, Gillman LM, Kazina CJ (2015) Magnesium sulfate for non-eclamptic status epilepticus. Seizure 32:100–108PubMed Zeiler FA, Matuszczak M, Teitelbaum J, Gillman LM, Kazina CJ (2015) Magnesium sulfate for non-eclamptic status epilepticus. Seizure 32:100–108PubMed
106.
Zurück zum Zitat Basha MM, Suchdev K, Dhakar M, Kupsky WJ, Mittal S, Shah AK (2017) Acute resective surgery for the treatment of refractory status epilepticus. Neurocrit Care 27:370–380PubMed Basha MM, Suchdev K, Dhakar M, Kupsky WJ, Mittal S, Shah AK (2017) Acute resective surgery for the treatment of refractory status epilepticus. Neurocrit Care 27:370–380PubMed
107.
Zurück zum Zitat Mohamed IS, Otsubo H, Imai K, Shroff M, Sharma R, Chuang SH et al (2007) Surgical treatment for acute symptomatic refractory status epilepticus: a case report. J Child Neurol 22:435–439PubMed Mohamed IS, Otsubo H, Imai K, Shroff M, Sharma R, Chuang SH et al (2007) Surgical treatment for acute symptomatic refractory status epilepticus: a case report. J Child Neurol 22:435–439PubMed
108.
Zurück zum Zitat Barros P, Brito H, Ferreira PC, Ramalheira J, Lopes J, Rangel R et al (2014) Resective surgery in the treatment of super-refractory partial status epilepticus secondary to NMDAR antibody encephalitis. Eur J Paediatr Neurol 18:449–452PubMed Barros P, Brito H, Ferreira PC, Ramalheira J, Lopes J, Rangel R et al (2014) Resective surgery in the treatment of super-refractory partial status epilepticus secondary to NMDAR antibody encephalitis. Eur J Paediatr Neurol 18:449–452PubMed
109.
Zurück zum Zitat Atkinson M, Atkinson B, Norris G, Shah A (2012) Refractory status epilepticus secondary to CNS vasculitis, a role for epilepsy surgery. J Neurol Sci 315:156–159PubMed Atkinson M, Atkinson B, Norris G, Shah A (2012) Refractory status epilepticus secondary to CNS vasculitis, a role for epilepsy surgery. J Neurol Sci 315:156–159PubMed
110.
Zurück zum Zitat D’Giano CH, Del CGM, Pomata H, Rabinowicz AL (2001) Treatment of refractory partial status epilepticus with multiple subpial transection: case report. Seizure 10:382–385PubMed D’Giano CH, Del CGM, Pomata H, Rabinowicz AL (2001) Treatment of refractory partial status epilepticus with multiple subpial transection: case report. Seizure 10:382–385PubMed
111.
Zurück zum Zitat Lupashko S, Malik S, Donahue D, Hernandez A, Perry MS (2011) Palliative functional hemispherectomy for treatment of refractory status epilepticus associated with Alpers’ disease. Childs Nerv Syst 27:1321–1323PubMed Lupashko S, Malik S, Donahue D, Hernandez A, Perry MS (2011) Palliative functional hemispherectomy for treatment of refractory status epilepticus associated with Alpers’ disease. Childs Nerv Syst 27:1321–1323PubMed
112.
Zurück zum Zitat Greiner HM, Tillema JM, Hallinan BE, Holland K, Lee KH, Crone KR (2012) Corpus callosotomy for treatment of pediatric refractory status epilepticus. Seizure 21:307–309PubMedPubMedCentral Greiner HM, Tillema JM, Hallinan BE, Holland K, Lee KH, Crone KR (2012) Corpus callosotomy for treatment of pediatric refractory status epilepticus. Seizure 21:307–309PubMedPubMedCentral
113.
Zurück zum Zitat O’Neill BR, Valeriano J, Synowiec A, Thielmann D, Lane C, Wilberger J (2011) Refractory status epilepticus treated with vagal nerve stimulation: case report. Neurosurgery 69:E1172–E1175PubMed O’Neill BR, Valeriano J, Synowiec A, Thielmann D, Lane C, Wilberger J (2011) Refractory status epilepticus treated with vagal nerve stimulation: case report. Neurosurgery 69:E1172–E1175PubMed
114.
Zurück zum Zitat Patwardhan RV, Dellabadia J Jr., Rashidi M, Grier L, Nanda A (2005) Control of refractory status epilepticus precipitated by anticonvulsant withdrawal using left vagal nerve stimulation: a case report. Surg Neurol 64:170–173PubMed Patwardhan RV, Dellabadia J Jr., Rashidi M, Grier L, Nanda A (2005) Control of refractory status epilepticus precipitated by anticonvulsant withdrawal using left vagal nerve stimulation: a case report. Surg Neurol 64:170–173PubMed
115.
Zurück zum Zitat Levisohn BR, Miller J, Winston KR, Levisohn P, Miller BR, Freeman J (2001) Vagal nerve stimulation for status epilepticus. Pediatr Neurosurg 34:190–192PubMed Levisohn BR, Miller J, Winston KR, Levisohn P, Miller BR, Freeman J (2001) Vagal nerve stimulation for status epilepticus. Pediatr Neurosurg 34:190–192PubMed
116.
Zurück zum Zitat Lee CY, Lim SN, Wu T, Lee ST (2017) Successful treatment of refractory status epilepticus using anterior thalamic nuclei deep brain stimulation. World Neurosurg 99:14–18PubMed Lee CY, Lim SN, Wu T, Lee ST (2017) Successful treatment of refractory status epilepticus using anterior thalamic nuclei deep brain stimulation. World Neurosurg 99:14–18PubMed
117.
Zurück zum Zitat Lehtimäki K, Långsjö JW, Ollikainen J, Heinonen H, Möttönen T, Tähtinen T et al (2017) Successful management of super-refractory status epilepticus with thalamic deep brain stimulation. Ann Neurol 81:142–146PubMed Lehtimäki K, Långsjö JW, Ollikainen J, Heinonen H, Möttönen T, Tähtinen T et al (2017) Successful management of super-refractory status epilepticus with thalamic deep brain stimulation. Ann Neurol 81:142–146PubMed
118.
Zurück zum Zitat Sanacora G, Mason GF, Rothman DL, Hyder F, Ciarcia JJ, Ostroff RB et al (2003) Increased cortical GABA concentrations in depressed patients receiving ECT. Am J Psychiatry 160(3):577–579PubMed Sanacora G, Mason GF, Rothman DL, Hyder F, Ciarcia JJ, Ostroff RB et al (2003) Increased cortical GABA concentrations in depressed patients receiving ECT. Am J Psychiatry 160(3):577–579PubMed
119.
Zurück zum Zitat Griesemer DA, Kellner CH, Beale MD, Smith GM (1997) Electroconvulsive therapy for treatment of intractable seizures. Initial findings in two children. Neurology 49:1389–1392PubMed Griesemer DA, Kellner CH, Beale MD, Smith GM (1997) Electroconvulsive therapy for treatment of intractable seizures. Initial findings in two children. Neurology 49:1389–1392PubMed
120.
Zurück zum Zitat Mirás Veiga A, Moreno DC, Menéndez AI, Siscart IM, Fernández MD, Sánchez EG et al (2017) Effectiveness of electroconvulsive therapy for refractory status epilepticus in febrile infection-related epilepsy syndrome. Neuropediatrics 48:45–48PubMed Mirás Veiga A, Moreno DC, Menéndez AI, Siscart IM, Fernández MD, Sánchez EG et al (2017) Effectiveness of electroconvulsive therapy for refractory status epilepticus in febrile infection-related epilepsy syndrome. Neuropediatrics 48:45–48PubMed
121.
Zurück zum Zitat Rosati A, L’Erario M, Ilvento L et al (2012) Efficacy and safety of ketamine in refractory status epilepticus in children. Neurology 79:2355–2358PubMed Rosati A, L’Erario M, Ilvento L et al (2012) Efficacy and safety of ketamine in refractory status epilepticus in children. Neurology 79:2355–2358PubMed
122.
Zurück zum Zitat Pressler RM, Cilio MR, Mizrahi EM, Moshé SL, Nunes ML, Plouin P et al (2021) The ILAE classification of seizures and the epilepsies: modification for seizures in the neonate. Position paper by the ILAE Task Force on Neonatal Seizures. Epilepsia 62:615–628PubMed Pressler RM, Cilio MR, Mizrahi EM, Moshé SL, Nunes ML, Plouin P et al (2021) The ILAE classification of seizures and the epilepsies: modification for seizures in the neonate. Position paper by the ILAE Task Force on Neonatal Seizures. Epilepsia 62:615–628PubMed
123.
124.
Zurück zum Zitat Pisani F, Cerminara C, Fusco C, Sisti L (2007) Neonatal status epilepticus vs recurrent neonatal seizures. Neurology 69:2177–2185PubMed Pisani F, Cerminara C, Fusco C, Sisti L (2007) Neonatal status epilepticus vs recurrent neonatal seizures. Neurology 69:2177–2185PubMed
125.
Zurück zum Zitat NEOLEV2 INVESTIGATORS, Sharpe C, Reiner GE, Davis SL, Nespeca M, Gold JJ, Rasmussen M et al (2020) Levetiracetam versus phenobarbital for neonatal seizures: a randomized controlled trial. Pediatrics 145:e20193182 NEOLEV2 INVESTIGATORS, Sharpe C, Reiner GE, Davis SL, Nespeca M, Gold JJ, Rasmussen M et al (2020) Levetiracetam versus phenobarbital for neonatal seizures: a randomized controlled trial. Pediatrics 145:e20193182
126.
Zurück zum Zitat Sutter R, Dittrich T, Semmlack S, Rüegg S, Marsch S, Kaplan PW (2018) Acute systemic complications of convulsive status epilepticus—a systematic review. Crit Care Med 46(1):138–145PubMed Sutter R, Dittrich T, Semmlack S, Rüegg S, Marsch S, Kaplan PW (2018) Acute systemic complications of convulsive status epilepticus—a systematic review. Crit Care Med 46(1):138–145PubMed
127.
Zurück zum Zitat Vernooy K, Delhaas T, Cremer OL, Di Diego JM, Oliva A, Timmermans C, Volders PG, Prinzen FW, Crijns HJ, Antzelevitch C, Kalkman CJ, Rodriguez LM, Brugada R (2006) Electrocardiographic changes predicting sudden death in propofol-related infusion syndrome. Heart Rhythm 3(2):131–137PubMedPubMedCentral Vernooy K, Delhaas T, Cremer OL, Di Diego JM, Oliva A, Timmermans C, Volders PG, Prinzen FW, Crijns HJ, Antzelevitch C, Kalkman CJ, Rodriguez LM, Brugada R (2006) Electrocardiographic changes predicting sudden death in propofol-related infusion syndrome. Heart Rhythm 3(2):131–137PubMedPubMedCentral
128.
Zurück zum Zitat Pérez Riera AR, Uchida AH, Schapachnik E, Dubner S, Ferreira Filho C, Ferreira C (2010) Propofol infusion syndrome and Brugada syndrome electrocardiographic phenocopy. Cardiol J 17(2):130–135 Pérez Riera AR, Uchida AH, Schapachnik E, Dubner S, Ferreira Filho C, Ferreira C (2010) Propofol infusion syndrome and Brugada syndrome electrocardiographic phenocopy. Cardiol J 17(2):130–135
129.
Zurück zum Zitat Walder B, Tramèr MR, Seeck M (2002) Seizure-like phenomena and propofol: a systematic review. Neurology 58(9):1327–1332PubMed Walder B, Tramèr MR, Seeck M (2002) Seizure-like phenomena and propofol: a systematic review. Neurology 58(9):1327–1332PubMed
130.
Zurück zum Zitat Brown EN, Lydic R, Schiff ND (2010) General anesthesia, sleep, and coma. N Engl J Med 363(27):2638–2650PubMedPubMedCentral Brown EN, Lydic R, Schiff ND (2010) General anesthesia, sleep, and coma. N Engl J Med 363(27):2638–2650PubMedPubMedCentral
131.
Zurück zum Zitat Burman RJ, Rosch RE, Wilmshurst JM, Sen A, Ramantani G, Akerman CJ, Raimondo JV (2022) Why won’t it stop? The dynamics of benzodiazepine resistance in status epilepticus. Nat Rev Neurol 18(7):428–441PubMed Burman RJ, Rosch RE, Wilmshurst JM, Sen A, Ramantani G, Akerman CJ, Raimondo JV (2022) Why won’t it stop? The dynamics of benzodiazepine resistance in status epilepticus. Nat Rev Neurol 18(7):428–441PubMed
132.
Zurück zum Zitat Rosati A, L’Erario M, Bianchi R, Olivotto S, Battaglia DI, Darra F, Biban P, Biggeri A, Catelan D, Danieli G, Mondardini MC, Cordelli DM, Amigoni A, Cesaroni E, Conio A, Costa P, Lombardini M, Meleleo R, Pugi A, Tornaboni EE, Santarone ME, Vittorini R, Sartori S, Marini C, Vigevano F, Mastrangelo M, Pulitanò SM, Izzo F, Fusco L (2022) KETASER01 protocol: What went right and what went wrong. Epilepsia Open 7(3):532–540PubMedPubMedCentral Rosati A, L’Erario M, Bianchi R, Olivotto S, Battaglia DI, Darra F, Biban P, Biggeri A, Catelan D, Danieli G, Mondardini MC, Cordelli DM, Amigoni A, Cesaroni E, Conio A, Costa P, Lombardini M, Meleleo R, Pugi A, Tornaboni EE, Santarone ME, Vittorini R, Sartori S, Marini C, Vigevano F, Mastrangelo M, Pulitanò SM, Izzo F, Fusco L (2022) KETASER01 protocol: What went right and what went wrong. Epilepsia Open 7(3):532–540PubMedPubMedCentral
134.
Zurück zum Zitat Antonov SM, Johnson JW (1999) Permeant ion regulation of N‑methyl-D-aspartate receptor channel block by Mg(2+). Proc Natl Acad Sci U S A 96(25):14571–14576PubMedPubMedCentral Antonov SM, Johnson JW (1999) Permeant ion regulation of N‑methyl-D-aspartate receptor channel block by Mg(2+). Proc Natl Acad Sci U S A 96(25):14571–14576PubMedPubMedCentral
Metadaten
Titel
Management and prognosis of pediatric status epilepticus
verfasst von
Eroshini Swarnalingam
Kristine Woodward
Micheal Esser
Julia Jacobs
Publikationsdatum
10.11.2022
Verlag
Springer Medizin
Erschienen in
Clinical Epileptology / Ausgabe 4/2022
Print ISSN: 2948-104X
Elektronische ISSN: 2948-1058
DOI
https://doi.org/10.1007/s10309-022-00538-0

Weitere Artikel der Ausgabe 4/2022

Zeitschrift für Epileptologie 4/2022 Zur Ausgabe

Informations de la Ligue Suisse contre l‘Epilepsie

Informations de la Ligue Suisse contre l‘Epilepsie

Mitteilungen der Österreichischen Gesellschaft für Epileptologie

Mitteilungen der Österreichischen Gesellschaft für Epileptologie

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.