5

Modern Treatment of Status Epilepticus in Adults

Kjell Heuser1 Ketil Berg Olsen1,3 Line Bédos Ulvin1 Leif Gjerstad1,2 Erik Taubøll1,2

1Department of Neurology, Oslo University Hospital – Rikshospitalet, Oslo, Norway; 2Faculty of Medicine, University of Oslo, Norway; 3Department of Neurology, Østfold Hospital Trust, Norway

Abstract: Status epilepticus is a serious epileptic condition associated with significant morbidity and mortality. It can be divided into four stages: developing (seizures leading up to status epilepticus), established (>5 minutes convulsive status epilepticus, nonconvulsive/focal status epilepticus >10 min), refractory (failure of two adequately dosed antiseizure medications in different drug classes), and super-refractory (persisting despite >24 hours of anesthesia). All seizure types can develop to status epilepticus. Generalized convulsive status epilepticus has the largest potential for brain damage. The drug treatment algorithms for status epilepticus generally go through three stages, starting with benzodiazepines: lorazepam, midazolam, or diazepam as first-line drugs, moving to levetiracetam, valproate or fos-phenytoin as second-line drugs, ending, if necessary, with anesthetics like propofol or midazolam. Lacosamide, topiramate, phenobarbital, brivaracetam, and perampanel are potential alternatives, and pentobarbital is commonly used when long-term anesthesia is needed. Super-refractory status epilepticus is the form that usually causes the most concern.Treatment guidelines for newer treatment alternatives are often based only on case reports and small patient series. Such alternatives include ketamine, immunotherapy, ketogenic diet, inhalation anesthetics and hypothermia, and several surgical neurostimulation procedures. This chapter provides an overview of current treatment alternatives, with particular focus on refractory status epilepticus and super-refractory status epilepticus.

Keywords: antiseizure medications; immune therapy; ketamine; non-pharmacological treatment; status epilepticus

Author for Correspondence: Erik Taubøll, Department of Neurology, Oslo University Hospital – Rikshospitalet, PO Box 4950 Nydalen, 0424 Oslo, Norway. Email: erik.tauboll@medisin.uio.no

Cite this chapter as: Heuser K, Olsen KB, Ulvin LB, Gjerstad L, Taubøll E. Modern Treatment of Status Epilepticus in Adults. In: Czuczwar SJ, editor. Epilepsy. Brisbane (AU): Exon Publications. Online first 2022 Feb 25.

Doi: https://doi.org/10.36255/exon-publications-epilepsy-status-epilepticus

In: Czuczwar SJ, editor. Epilepsy. Exon Publications, Brisbane, Australia. ISBN: 978-0-6453320-4-9. Doi: https://doi.org/10.36255/exon-publications-epilepsy

Copyright: The Authors.

License: This open access article is licenced under Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/

INTRODUCTION

Status epilepticus (SE) defined as continuous epileptic activity, is a major medical and neurological emergency requiring immediate treatment to avoid severe morbidity and mortality. All types of epileptic seizures, including focal and absence seizures, can turn into SE, but the most common and serious form of SE is generalized convulsive SE. Persistent epileptic activity, whether generalized or focal, causes excessive stress to both the organism in general and the brain in particular.

According to the previous International League Against Epilepsy (ILAE) definition, SE was defined as 30 minutes of ongoing epileptic activity or a series of seizures without recovery for a period of > 30 minutes. The rationale behind this definition was based on animal studies showing irreversible neuronal injury after long-lasting seizure activity, and it was agreed somewhat arbitrarily on a time limit of 30 minutes (13). In 2015, the ILAE proposed a new definition of SE (4) that reduced the time limits to 5 minutes of ongoing seizure activity to diagnose convulsive SE (CSE), and 10 minutes for focal and absence SE (4). Several types of SE with prominent motor phenomena at any time (including CSE) were distinguished from those without (i.e., nonconvulsive SE (NCSE)). The new ILAE definition introduced the concept of two important time points, t1 and t2. The first, t1, is the time point at which the seizure should be regarded as an “abnormally prolonged seizure”. The second time point, t2, is the time of ongoing seizure activity beyond which there is a risk of long-term consequences, such as irreversible brain damage and neurological sequelae (4). Evidence for these time points is, however, incomplete. Therefore,these time points should be considered as the best estimates currently available (4) (Table 1).

TABLE 1 Operational dimensions of t1 and t2 for status epilepticus

Type of SE Operational dimension 1 Operational dimension 2
Time (t1) when a seizure is likely to be prolonged leading to continuous seizure activity Time (t2) when a seizure may cause long term consequences (including neuronal injury, neuronal death, alteration of neuronal networks and functional deficits)
Tonic-clonic SE 5 min 30 min
Focal SE with impaired consciousness 10 mina >60 min
Absence SE 10-15 mina Unknown

a Evidence for the time frame is currently limited and future data may lead to modifications. Operational dimensions with t1 indicating the time that emergency treatment of SE should be started, and t2 indicating the time at which long-term consequences may be expected. Modified from Trinka et al 2015 (4).

The incidence of SE varies considerably between studies, from below 10 to above 40 per 100,000 adults per year (58). The new definition of SE will invariably lead to a change in incidence. In a population-based study, the incidence of SE was found to be 10% higher when the new definition was applied (5). Mortality in SE varies between 9–37%, highly dependent on etiology and time in SE (913). In refractory SE, mortality rate is approximately three times higher than for non-refractory SE (10). Morbidity is high in SE with about 40% of patients having a reduced modified Rankin Scale at hospital discharge (13). There are also indications that focal SE has a negative impact on cognitive function (1315).

SE can be divided into four stages as follows: (i) developing––seizures leading up to SE; (ii) established––> 5 minutes (CSE, Table 1); (iii) refractory––failure of two adequately dosed antiseizure medications (ASMs) in different drug classes (a benzodiazepine + another ASM); (iv) super-refractory––SE persisting despite > 24 hours of anesthesia (4, 16). SE requires immediate treatment to prevent successive brain injury. A series of treatment guidelines and protocols have been published, most of which recommend initial use of benzodiazepines, followed by intravenous ASMs starting with valproate, levetiracetam or, less frequently, fos-phenytoin (fos-PHT), and subsequently induction of anesthesia. Molecular studies indicate that rapid cellular and subcellular changes occur with ongoing SE, such as internalization of GABA-receptors into the cytoplasm of neurons (17) and up-regulation of NMDA-receptors in the synapses after ~30 min (18). Starting treatment early and giving adequate dosages of appropriate medication is therefore a prerequisite for a good outcome. Good diagnostic evaluation of the many possible causes of SE, together with prompt and suitable treatment could prevent development to refractory SE (RSE). This chapter provides a brief overview of non-surgical SE treatment, with major focus on the most severe and resistant form: super-refractory status epilepticus (SRSE). Lack of compliance and forgotten medication are very common cause of SE. Consider starting with a bolus dose of the drug that the patient is already taking.

EARLY-STAGE STATUS EPILEPTICUS

The treatment of SE has been discussed for many years, but no consensus has been reached when it comes to the preferred use of ASM, and especially regarding the use of anesthetics. Many guidelines and protocols have been published (4, 1927), recommending initial use of benzodiazepines, followed by intravenous ASMs and, subsequently, induction of anesthesia.

All international guidelines recommend the use of benzodiazepines in the early stages of SE (22, 2628). Which benzodiazepine to use and what is the best route of administration have been discussed for years, but more important is to use a sufficiently high dose of the chosen drug as soon as possible. Lorazepam at a dose of 2-4 mg intravenously (IV) is often advocated as first-line treatment. However, because lorazepam is not listed for use in all countries, diazepam is often used as a first-line drug, at a dosage of 10-20 mg IV with an infusion rate of 5 mg/min. A few comparative studies and a Cochrane report showed a possible slightly better effect and less need for additional ASMs for lorazepam compared with diazepam (29, 30). However, a more recent systematic review including a meta-analysis of five randomized controlled trials (RCTs) and a total of 656 patients could not find any differences between lorazepam and diazepam regarding effect and side-effects (31). Alternatively, diazepam can be administered rectally and recently also intranasally, an administration route well suited also for children (32). The third drug of choice in early-stage SE is midazolam, at a dosage of 10-20 mg and with buccal, intranasal, or intramuscular administration (33, 34). For prehospital treatment, it is of special interest to note that an intramuscular administration of midazolam was more effective and median time to active treatment was shorter than for IV lorazepam (34). This probably reflects the practical problems with IV administration to a patient with an ongoing seizure outside hospital, an aspect that should be considered when establishing local guidelines for prehospital treatment. Clonazepam 1-3 mg may also be used as an alternative benzodiazepine (35). Generally, the same doses can be administered again after 5 min for all benzodiazepines if the response is inadequate, with individual considerations in the elderly and patients with body weight below 50 kg (10).

ESTABLISHED STATUS EPILEPTICUS

ASMs other than benzodiazepines are the preferred drugs for this stage of SE. The drugs most recommended are valproate, levetiracetam or fos-PHT (3638).

Valproate

Valproate is most often recommended at dosages of IV 30-40 mg/kg body weight over a period of 10-20 min (36, 39). In the ESETT study, a bolus dose of 40 mg/kg over 10 min was used (37). Initial serum levels drop early as valproate distributes into body fat, so maintenance dosing should be started 30 minutes to 2 hours after IV loading. We have not experienced any difficulties in immediately following up the bolus dose with continuous infusion of 100-200 mg/hour over the next 24 h. Alternatively, maintenance dose of about 5 mg/kg every 6 hours can be used. Following the valproate serum concentration repetitively at this stage is highly recommended.

Levetiracetam

Levetiracetam has been used with increasing frequency in both RSE and SRSE since the first case reports appeared in 2005 (40). Several studies on the effect of levetiracetam have been performed demonstrating an effect ranging from around 50-70% (4144). An extensive review by Trinka and Dobesberger collecting 156 cases treated with levetiracetam for SE (42) found an overall success rate of 65.4%, confirmed by data from Yasiry and Shorvon describing an efficacy of 68.5% (44). The recently published ESETT study found an efficacy of ~ 50% for the primary outcome of termination of clinical seizures plus improvement in the level of consciousness by 60 minutes after IV administration of levetiracetam (37). In an earlier study, Eue et al. (45) described a slightly better efficacy of levetiracetam for non-convulsive than generalized convulsive SE, but this has not been confirmed. When it comes to the loading doses of levetiracetam, most recent studies have used 20-30 mg/kg (42, 44, 46). However, the ESETT study employed a loading dose of 60 mg/kg levetiracetam and compared this with respective loading doses of 20 mg/kg for fos-PTH and 40 mg/kg for valproate, all leading to similar results (efficacy in about 50% of cases after 60 min) (37).

Fos-PHT

Fos-PHT is a prodrug of phenytoin with pharmacokinetic bioequivalence (47, 48), and dosage is therefore specified in Phenytoin Equivalents (PE). It is generally recommended to administer fos-PHT IV with a loading dose of 20 mg PE/kg body weight. This is given as an infusion at a rate of 100 mg PE/min (50-150 mg PE/min). A maintenance dose of 5 mg PE/kg body weight is often used, given twice daily, with the first dose typically 6-8 hours after the loading dose. Side-effects of fos-PHT are like those of phenytoin but are usually more moderate (48).

Phenobarbital

Phenobarbital (10 mg/kg iv bolus dose at a max rate of 100 mg/min) is still used as second line treatment for SE in some centres and countries, but because of its depressive effect on respiration it has now been largely replaced by other drugs (49).

Which drug to choose for established status epilepticus, and at what dose?

Taken together, no differences between valproate, levetiracetam or fos-PHT were detected in efficacy or primary safety outcome by drug within each age group (50). However, drug safety should be evaluated carefully for each drug as individual comorbidities may favor or disfavor specific drugs. As an example, valproate should be used with caution in patients with mitochondrial dysfunction or severe liver diseases, although there are no reports of liver failure because of treatment with valproate in the acute phase of SE (51). Fos-PHT might not be the best choice in certain patients with cardiac conduction problems due to its pro-arrhythmogenic side effects (52). Finally, levetiracetam is known to cause behavioural side effects and one should be aware of possible negative consequences in patients with pre-existing behavioural deviations. In most cases, the SE patient is continued on the same drug as used to stop SE. Careful evaluation of which drug to choose after termination of SE should therefore be mandatory already in the early stage of SE treatment. As a precaution, valproate should not be used in pregnant women with SE, although there are no data showing that short-term use of valproate has any negative effect on the fetus, given that valproate is later shifted to another ASM after cessation of SE.

The question of which maximum loading dose to use for second line treatment of SE has not been settled yet. In this respect, it is interesting that the ESETT study (37) utilized a maximum loading dose for patients above 75 kg (4500 mg for levetiracetam, 1500 mg for fos-PHT and 3000 mg for valproate). Consequently, people with body weight >75 kg received a lower mg/kg dosage than those with body weight <75 kg, but the effect on SE was equal in those above and below body weight of 75 kg (53). This would imply that traditional loading doses may have been higher than necessary. This is also in line with data from Rossetti et al. who found that elevating the loading dose of levetiracetam above 3000 mg was unlikely to provide any additional benefit (54). In an earlier study on valproate treatment in children with SE, the effect was 46% when a bolus dose at 20-30 mg/kg was given, and 73% at bolus dose of 30-40 mg/kg. Paradoxically the effect was again reduced to 40% at bolus doses above 40 mg/kg (55). It therefore seems plausible to use maximum bolus doses of 20 mg/kg for fos-PHT, 40 mg/kg for valproate and 40 to 60 mg/kg for levetiracetam in most adults with body weights up to 75 kg and only increase maximum dose in those with higher body weights after individual considerations.

REFRACTORY STATUS EPILEPTICUS

After failure of two adequately dosed ASMs in different drug classes including a benzodiazepine and one of the above-mentioned drugs (valproate, levetiracetam or fos-PHT), anesthetics should be considered. However, use of anesthesia and thereby therapeutic coma (TC) may also have several negative consequences. A study comparing the use of TC for SE treatment in the US (Harvard) versus Europe (Geneva), showed that TC was much more frequently used in the US. While this did not affect mortality, more extensive TC use increased the length of hospital stay and related costs significantly (56). On the contrary, it was recently found that early induction of TC after unsuccessful first-line treatment was associated with shorter SE duration as well as intensive care unit/hospital stay, than when a second line ASM was applied first (57). Novy and co-workers reported that escalating treatment with a non-sedating ASM terminated refractory SE in more than half their patients (58). In another study where lorazepam was followed sequentially by phenytoin, levetiracetam and valproate, SE was controlled in 92% of cases thereby avoiding TC (59). On the other hand, the risks from using anesthetics must be balanced against the risk of long-lasting seizure activity. The question of when to use TC is therefore not finally settled, but must be discussed on an individual basis, especially for focal and non-convulsive forms of SE (60).

If TC is decided, patients with RSE, independent of generalized or focal/nonconvulsive, should be treated with general anesthesia. Recommended anesthetics are propofol, midazolam, or barbiturates, especially pentobarbital. Loading doses and infusion rates vary noticeably in the literature, ranging from 0.5-20 mg/kg/h for barbiturates, 0.1-24 mg/kg/h for propofol, and 0.02-1.8 mg/kg/h for midazolam (36, 38, 47).

Anesthetic drug preferences have been extensively discussed (10, 16, 25, 36, 38, 47), but comparative trials are still lacking. In two systemic reviews, none of the treatments currently available was found to be superior to another (25, 61). Patients with SE based on severe pathological conditions, such as hypoxia, are often treated with barbiturates, as are patients that do not respond to either midazolam or propofol. Complications, such as respiratory depression and hypotension, have been reported for all three drugs. Therapy failure due to side-effects occurred in 6% of patients who received propofol, versus 3% in barbiturate- and <1% in midazolam-treated patients. Death during therapy was most prevalent in patients receiving barbiturates (19%), versus 8% of propofol- and 2% of midazolam-treated patients. This probably reflects the use of barbiturates in the most severe cases (47).

Generally, propofol or midazolam tend to be the first-line anesthetics, whereas barbiturates are often reserved for SE refractory to these drugs (10). Propofol is advantageous with respect to its short half-life, making frequent neurological exams possible. It should, however, be noted that long-term propofol infusion may lead to propofol infusion syndrome. This is a life-threatening condition, including rhabdomyolysis, cardiac and renal failure, and metabolic acidosis. Propofol treatment should therefore be restricted to 48 h (62). To avoid withdrawal seizures, we recommend that propofol is gradually reduced over 24 h before treatment is stopped. Duration of anesthesia or degree of electrographic suppression is controversial; common practice is to achieve electrographic burst suppression for 24 to 48 h, but the rationale for this is limited. Whether or not burst suppression is necessary can be discussed, but all epileptic activity must be continuously suppressed for a certain period. However, as most hospitals do not have unlimited access (24 h per day) to continuous EEG, burst suppression can be regarded as a practical approach for ascertaining that anesthesia has been sufficient.

It is important to remember that therapy failure is probably the result of an insufficient target dose of the anesthetic drug, including dose limitation due to side-effects, rather than choice of drug. We therefore recommend to load the patient fully with the chosen ASM to therapeutic, or even supra-therapeutic, levels when the patient is still in anesthesia to avoid insufficient serum concentrations and seizure breakthrough when anesthesia is tapered off.

SUPER-REFRACTORY STATUS EPILEPTICUS

SRSE is one of the most difficult-to-treat conditions in neurology and life-threatening. Due to a large variation in etiology and clinical presentation (including heterogeneity in age, comorbidity etc.), as well as its rarity, robust clinical studies of SRSE are extremely difficult to perform. One major challenge is the increasing amount of treatment options, both with diverse ASMs but also with a wide range of alternative therapies reported to be helpful in case reports or small patient series. Such reports should be considered cautiously due to the possibility of publication bias. Herein, the treatment of SRSE is discussed under three groups: (i) the use of ASMs other than PHT/fos-PHT, valproate, levetiracetam or phenobarbital, which are used in the earlier phases of SE and commented on above; (iii) other drugs or supplementations given IV, perorally, or through inhalation; and (iii) other non-pharmacologic treatments. In addition to valproate, PHT/fos-PHT, levetiracetam, and phenobarbital, especially lacosamide and topiramate, and even more recently, perampanel and brivaracetam, have gained attention. While lacosamide and brivaracetam can be administered IV, topiramate and perampanel can only be used perorally, hampering its use in SRSE. For all ASMs mentioned here, evidence for their use in SRSE and their effects on SE in general, is very sparse and should be considered with great caution. Studies also vary markedly regarding etiologies of seizures and SRSE, outcome criteria, and definitions of efficacy. Several studies are not even specific regarding the type of SE.

Lacosamide

To date, lacosamide has been used in a limited number of SE. In their review from 2013, Höfler and Trinka (63) refer to 136 episodes of RSE (50% nonconvulsive status epilepticus, 31% focal status epilepticus, and 19% convulsive status epilepticus) treated with lacosamide. The most frequently used bolus dose used was 200-400 mg IV over 3-5 min. The overall success rate was 56% (76/136). This is very much in line with a recently published systematic review comparing lacosamide (n=115) with phenytoin (n=166) lacking significant differences between the two drugs for either seizure control (57% for lacosamide versus 46% for phenytoin) and for side effects (64). Adverse events of lacosamide are generally mild with dizziness, ataxia, diplopia, headache, and nausea, but one should be aware of possible cardiac effects of the drug. Especially, atrial arrhythmias, AV block, and ventricular tachyarrhythmia may occur with rapid intravenous loading (65, 66). It should therefore be used with caution in patients with underlying pro-arrhythmic conditions. The exact loading dose to achieve a sufficient serum concentration has been discussed. As mentioned, 200 to 400 mg has been used in most studies, but to be more specific, doses above 9 mg/kg seem to be necessary to swiftly achieve therapeutic serum levels (67). In another study, doses of above 8 mg/kg led to effective drug concentrations of 15-20 μg/ml (68). With a loading dose of 10 mg/kg one would be on the safe side, and this has therefore been suggested (38).

Topiramate

As for lacosamide, data on use of topiramate for RSE, and especially SRSE, are scarce. In one study comprising 35 adults with either generalized or focal SE, topiramate 100 mg x 4-6/day was considered effective in 44% (14/35) after 3 days (69). In another study (70), topiramate was used as an add-on treatment after therapy failure of 1-6 (median 4) previously administered ASMs. It was introduced after a median of 2 (range 2-23) days, for a period of 1-24 (median 3) days. Overall, SRSE was terminated in 71% of patients within 72 h after first topiramate administration, and in 9% of patients within 24 h. In a large study comprising 106 patients with RSE or SRSE, the rate of seizure cessation after topiramate was 27% (71). Topiramate was started at a median of 8.5 days after SE with 100 mg/day and titrated up to 400 mg/day. A possible effect of topiramate was also seen in a small series published in 2017 by Brigo and co-workers (72). As with lacosamide, it seems that there is some evidence that topiramate may be effective and could be considered in very difficult cases.

Newer antiseizure medications

A wide range of other ASMs have been tried to treat SE, and there are ample case reports describing a possible benefit. These include brivaracetam, perampanel, pregabalin, oxcarbazepine, stiripentol, and others.

Brivaracetam is a high-affinity synaptic vesicle glycoprotein 2A ligand that is structurally related to levetiracetam. Due to its more lipophilic characteristics, it may have a quicker penetration across the blood-brain barrier and potentially also a stronger anticonvulsant effect. It would therefore theoretically be a good alternative to levetiracetam in SE. Brivaracetam has been tried in several case reports and small series with promising results, much in line with other ASMs. The largest study so far is an Italian study of 56 cases. Brivaracetam was administered IV as the first drug after benzodiazepine failure in 21% and as second and third (or later used) drug in 38% and 38%, respectively (73). Median loading dose was 100 mg ranging from 50 to 300 mg over 10 to15 min (only 3 min in three cases without complications observed). Brivaracetam was judged effective in 57% which is in line with valproate, levetiracetam and fos-PHT.

Perampanel is a selective non-competitive AMPA-receptor antagonist, the only one among the available ASMs. The drug seems to be one more alternative in the armamentarium of ASMs to treat SE with its unique mode of action (74). In the largest study so far, 81 patients with different kinds of SE were treated with perampanel after having failed a median of three drugs. Loading doses ranged from 2 to 36 mg followed by median daily doses of 12 mg (75). Thirty-three percentage of cases were judged as responders. Response to perampanel was defined as being clinically and electrographically seizure-free within 72 h of initiation of perampanel if no further drugs were added. No cardiorespiratory adverse events or laboratory abnormalities were noted with perampanel treatment. Personal experience in three cases with 36 mg on day one, 24 mg on day two and 12 mg on day three followed by dose adjustments according to serum concentrations was effective in one SRSE patient (mitochondrial disease) stopping clinical and electroencephalographical seizure activity for >72 h with no observed side effects in any of the patients. Seizures later resumed and the patient died a few weeks later.

However, data are generally limited for both perampanel, brivaracetam, and other “try-and error” ASMs. Such drugs are often only tried in patients that has not responded to more conventional treatment. A further discussion on their role in SRSE treatment is not within the scope of this review. Nevertheless, this demonstrates the urgent need for systematic registration of use of different ASMs on a broader scale to provide information on efficacy and side-effects.

NON-ANTISEIZURE MEDICATIONS USED IN THE TREATMENT OF STATUS EPILEPTICUS

The most promising new treatment strategies for SE, especially SRSE are, in our opinion, ketamine and immunotherapy. Both have been tried in a sufficient number of patients to evaluate their potential role in SRSE treatment.

Ketamine

In the later stages of SE, and with prolonged seizure activity, GABA-A receptors are internalized and sensitivity to GABAergic drugs is reduced. Excitatory synapses show changes in the opposite direction from those of GABA synapses with NMDA receptor subunits being recruited to the synaptic membrane, where they form additional receptors (76). NMDA receptors may therefore be promising targets for anti-seizure treatment in SRSE. Ketamine is a non-competitive NMDA receptor antagonist and highly efficacious in stopping SE in animals and even with a possible neuroprotective effect (77, 78). Based on these properties, ketamine has been used for SE treatment in humans, but most published data are from single cases or small case series, implying clear limitations as described in detail below; however, the general impression is positive.

Experiences with IV ketamine in the treatment of SRSE have been published in a multicenter study from several hospitals in North America and Europe collecting 60 SRSE episodes in 46 adults and 12 children (79). More than half the patients had new-onset SRSE of unknown origin (NORSE). The main findings were: “permanent control” of SRSE in 57% (34 of 60), defined as no recurrence of SE during the same ICU stay; “likely response” to ketamine in 12%, defined as permanent control of SE within 24 h of initiation of ketamine when ketamine was the last drug added; and “possible response” in 20%, defined as permanent control of SRSE within 24 h of initiation when ketamine was not the last drug added. Taken together, ketamine was considered to have contributed to permanent control in 32% (19 of 60, including seven in which ketamine was the last drug added considered as a “likely response”). Median loading doses used by the participating centres was 1.5 mg/kg, with a maximum of 5 mg/kg, followed by continuous infusion (median 2.75 mg/kg/h; max 10 mg/kg/h).

Another study investigating the effect of ketamine in SRSE in adults consisted of 68 cases collected between 2009 - 2018 from a tertiary epilepsy care centre (80). Seizures were completely controlled in 63% of ketamine recipients; an additional 18% had a greater than 50% reduction in seizures. Average dose of ketamine infusion used in this study was 2.2 mg/kg/h with a median treatment duration of 2 (1-4) days. The same study attested no adverse effects of high-dose ketamine on intracranial pressure; rather the doses used in this study were associated with a decreased need for vasopressor application.

The two abovementioned relatively large studies are supported by previously published minor reports and case series in both adults and children in which doses as high as 7.5 mg/kg/h were used for up to 14 days and were described as effective and safe (8185). According to one report, even doses up to 10 mg/kg/h for up to 27 days have not been associated with increased complications or mortality (86). Ketamine treatment seems to be effective in cases of both generalized convulsive and focal non-convulsive SE. Data are however insufficient to provide definite therapeutic recommendations regarding the effect of ketamine on different types of SE.

Likewise, optimal loading dose and doses for continuation are not finally settled, but in an ongoing randomised controlled trial in adults (NCT03115489), an IV loading dose of 2.5 mg/kg of ketamine is given followed by a continuous infusion at 3 mg/kg/h with the possibility to increase up to 10 mg/kg/h until burst suppression in EEG. This seems to be a good starting point for further clinical practice.

Side-effects of ketamine are generally few and no increase in mortality rate has been described (7980), although this can obviously be difficult to evaluate in SRSE. One advantage with ketamine is its cardiovascular effect as a slight vasopressor; one study (83) clearly showed that ketamine improved haemodynamic stability and reduced the need for vasopressor support.

A potential neurotoxic effect of ketamine, especially in the developing brain, has been discussed, but ketamine has also been associated with neuroprotective actions (78, 84). The unconfirmed risk of neurotoxicity must thus be balanced against the severity of SRSE and the possibility of serious brain damage due to the SRSE.

Immunotherapy

The first report of epilepsy due to brain inflammation dates back to 1958 when the Canadian neurosurgeon Theodore Brown Rasmussen from Montreal Neurological Institute described a few children with intractable focal seizures, progressive hemiparesis, and one-sided inflammatory changes in the brain (87). Since then, evidence has accrued that brain inflammation is involved in both ictogenesis, the consequence of single seizures, and in epileptogenesis (8890). In recent years, a wide range of autoantibodies have been identified in relation to severe forms of epilepsy, like FIRES, NORSE and Rasmussens encephalitis, and autoantibodies against the NMDA receptor, leucine-rich glioma-inactivated I (LGI1) complex (a protein associated with the voltage-gated potassium channel VGKC), and other autoantibodies against VGKC, VGCC, GAD, GABA etc. have been found| (90). When assessing a patient with SRSE, autoantibodies should now be measured routinely, and immune therapy considered early. However, lack of RCTs or larger (empirical) studies challenge the choice of immunotherapy. Steroids, intravenous immunoglobulins, plasma exchange/ plasmapheresis, rituximab, efalizumab, natalizumab, tofacitinib, azathioprine, cyclophosphamide, anakinra and other therapies have been tried (25, 9093). These drugs are often used in different combinations and in combination with other therapies. In FIRES for example, the combination of anakinra and ketogenic diet has recently been suggested (94).

In our clinic, high-dose steroid therapy, intravenous immunoglobulins, and rituximab are most often considered first. Also, anakinra, an interleukin-1 receptor antagonist is another promising drug with possible antiseizure (9597) as well as antiepileptogenic effect (98). RCTs designed to study immunotherapy in presumed autoimmune epilepsy are urgently needed to provide more valuable information and, at best, guidelines.

Neurosteroids

A promising new antiseizure drug, brexanolone, has recently been tested in SRSE. The active substance of brexanolone is allopregnanolone, a naturally occurring neurosteroid and progesterone metabolite. Progesterone and its metabolites are well known to have strong neuroactive properties decreasing brain excitability with allopregnanolone being the most potent (99). Allopregnanolone is a positive allosteric modulator of the GABAA receptor (100102) and acts on a broad range of GABAA receptor isoforms including an extrasynaptic GABAA receptor (103).

Several minor studies showed very promising results in SE, and in a study of 25 patients with superrefractory cases, brexanolone was effective in 73%. No serious side effects were observed (104).

These encouraging results gave rise to the multicenter study SAGE-547 (https://clinicaltrials.gov/ct2/show/NCT02477618 [accessed on 21 December 2021]). SAGE-547 is a randomized, double-blinded, placebo-controlled trial designed to evaluate the efficacy and safety of SAGE-547 (brexanolone) administered as continuous IV infusion in patients with SRSE. In this study, brexanolone infusion was given for 6 days. Patients were classified as responders when all third-line agents could be weaned off (and remained off for≥24 hours) before the infusion (drug or placebo) was stopped. 122 study sites participated, and 132 patients were included, of which 66 received placebo and 67 brexanolone. Seizure freedom was found in 43.9% in the group receiving brexanolone in addition to standard treatment (29 of 53 that completed) versus 42.4% in the group receiving standard treatment with placebo as add-on (28 of 50 that completed).

This result is apparently disappointing and challenges present concepts of drug development and testing also for other treatment options for RSE and SRSE. Many currently available treatment alternatives, both pharmacological and non-pharmacological ones, are based on promising results described in case reports, small case series, or open studies that cannot be confirmed in a properly designed randomized, double blinded, placebo-controlled trial as demonstrated in this case. The SAGE-547 results underline the urgent need for larger studies, due to the rarity of RSE and SRSE probably only achievable as multicenter RCT studies, and through the establishment of national and international registries.

OTHER TREATMENT OPTIONS

A broad spectrum of alternative therapies has been suggested and tried to treat SE. For all these therapies generally only case reports and small case series exist while systematic comparative trials are lacking. Publication bias should therefore be regarded as high. Hypothermia, magnesium infusion, pyridoxine infusion (highly effective in cases of inborn error of pyridoxine metabolism, also found as responsible for SE in rare cases in adults), lidocaine infusion, ketogenic diet, nerve stimulation (vagal, trigeminal, deep brain), resective neurosurgery, electroconvulsive therapy (ECT), cerebrospinal fluid drainage, and many others have been tried and may be beneficial in individual cases. The numerous therapeutic possibilities are summarized in multiple review articles (10, 25, 105110).

Magnesium infusion

Magnesium has a potential role in SE as it blocks the NMDA receptor and has an antiepileptic effect in experimental models (111). Magnesium is the drug of choice when treating seizures in eclampsia and has been found effective in a very few case studies in patients with mitochondrial disease, porphyria, and even in a case with myoclonic SE (106). There is no consensus regarding dose, but a serum level up to 1.7 mmol/l has been suggested (16, 60, 112).

Inhalation anesthetics

Inhalation anesthetics have been tried in several small studies, with isoflurane usually being the preferred drug. Although isoflurane suppresses seizure activity while the drug is applied, recurrence rate is high and complications probably frequent (16). In a small study including 7 patients, all 7 developed hypotension, 5/7 infections, 3/7 paralytic ileus, and 2/7 deep vein thrombosis (113), but mortality and long-term morbidity were considered as related to the underlying disease and duration of RSE, rather than to isoflurane. A review comprising 13 studies with a total of 82 SE episodes treated with isoflurane draws a different picture attesting fewer and milder complications, with hypotension as the most frequent side effect (108). Given the high recurrence rate after cessation of inhalation anesthetics, but also possible complications, and practical and logistical challenges with long-term use, this therapy approach should be limited to highly selected cases. Despite this, several national guidelines including the newly published guidelines from the German neurological society (27) mention inhalation anesthetics as a possibility in highly refractory cases.

Ketogenic diet

Data on treatment of RSE/SRSE with ketogenic diet are sparse, but several small uncontrolled case series have indicated a favourable outcome in both children (114, 115) and adults (116, 117). The effect usually seems to occur after 2-5 days. In a multicenter study including 15 adult patients, ketogenic diet was started in patients with SRSE with ketosis state reached on average after two days, and successfully stopped SRSE in 11 patients (73%) (117). This is in line with another study comprising 16 children (median age 8 yrs.) in which the diet was effective in 9 cases (56%) (118). With the relatively rapid onset of action, ketogenic diet can be tried, but, if not effective, should be withdrawn 3-5 days after ketosis is reached. Ketogenic diet may affect serum concentrations of ASMs (119) and may also influence certain forms of intensive care treatment. Notably, ketogenic diet is contraindicated in patients with hepatic failure, acute pancreatitis, or metabolic acidosis (109). It is also recommended not to combine ketogenic diet with propofol, since it increases the risk for fatal propofol related infusion syndrome (120). However, more studies and clinical experience on use and safety of ketogenic diet in SE is needed to make more concrete therapy recommendations. So far, no randomized trials or controlled trials are available.

Hypothermia

Hypothermia exerts anti-epileptic and neuroprotective effects in experimental SE and has also been used in the treatment of SRSE in humans. Data are sparse and very few cases have been properly described (107). The usual recommendation is hypothermia to around 31-35 °C, but even lower body temperatures have been tried. The frequency of complications is high and general anesthesia is always required when hypothermia is established (121). One multicenter RCT exists on the efficacy of hypothermia as add-on therapy in critically ill and ventilated patients with convulsive SE (target temperature 32–34 °C for 24 h) (122). In this study, hypothermia seemed to reduce the number of patients progressing to EEG-verified SE but did not improve clinical outcome at three months post SE. Common adverse effects included deep vein thrombosis, coagulopathy, increased risk for infection, and hemodynamic alterations including bradycardia and hypotension (121, 122).

Electroconvulsive therapy

There are no general recommendations on the use of Electroconvulsive therapy (ECT) in SRSE. However, several case reports, small series, and reviews on ECT treatment in SE are available (123127). In a review by Lambrecq and colleagues a favourable outcome was reported including cessation of SRSE in 80% and full recovery in 27% of patients (123). More recently, a case series of 6 patients receiving ECT lead to termination of SRSE in all cases (126). However, up to 12 ECT sessions were required posing a challenge to evaluation of the effect of ECT per se. ECT is stated as one out of several treatment options for SRSE in the newly published guidelines by the German Neurological Society (27). There are, however, several practical and ethical considerations when treating patients with ECT for SRSE. Notably, a reduction of anesthesia is required to assure recurrence of seizures, and only analgesia is given. Usually, ECT is applied in multiple series, necessitating frequent changes in the anesthesia level. Therefore, a rigorous ethical discussion should be conducted before ECT is performed in patients with SRSE, and this procedure should, in our opinion, be regarded as last resort.

Electrical or magnetic stimulation

Case reports and small case series on the use of vagus nerve stimulation (VNS), trigeminal stimulation (TNS), deep brain stimulation, and transcranial magnetic stimulation have been published, but the data are scarce, and no firm conclusions can be drawn (127). In our own limited experience, treatment with VNS and 2 cases of TNS proved unsuccessful, contrasting the case reports in the literature describing a positive effect. Systematic larger studies evaluating the effect of these treatment options are needed.

CONCLUSION

Most cases of established SE are treatable. However, much more challenging is the treatment of RSE/SRSE in which about 25% of cases are not controlled despite extensive treatment (25). Certain requirements should be fulfilled to avoid that established SE develops to RSE/SRSE. First, it is vital to start treatment rapidly with benzodiazepines immediately followed by levetiracetam, valproate or fos-PHT. Second, these drugs must be given in sufficient doses. Our strategy plan for SE treatment is given in Table 2.

TABLE 2 Flow-chart for treatment of status epilepticus, Stage 1-5

Stage 1
ABC – airway, breathing, circulation

Unknown patient:

  1. Consider hypoglycemia – give glucose 50 ml, 50%
  2. Consider alcohol related seizures – give thiamine 200 mg i.v. (can be dissolved in 100 ml NaCl). Give thiamine before glucose
  3. Avoid hyperthermia
Discuss: Etiology and triggering factors. Intoxication/drugs?
Stage 2
Benzodiazepines
Diazepam 10 mg i.v. (can be give rectally) OR
Midazolam 10 mg p.o. (can be given i.m. or intranasally) OR
Lorazepam 2-4 mg i.v.
Individual assessments must be done. Initial diazepam dose often increased up to 20 mg i.v. in adults. All drugs can be given in repeated doses if needed. Consider reducing dose (up to 50%) in the elderly and in patients <50kg
Stage 3
Valproic acid, 40 mg/kg, i.v., infusion rate 10 min (Max dose often set to 3000 mg)
Levetiracetam, 40 mg/kg i.v., infusion rate 10 min (Max dose often set to 4500 mg)
Phenytoin/fos-phenytoin, 20 mg/kg (20 mg/kg PE) i.v. infusion rate 50 to max 150 mg/min
Stage 4
Anesthesia
Propofol, 2-5 mg/kg i.v., infusion 2-5 (10) mg/h. Try to restrict to 48 h treatment, OR
Midazolam 0.2 mg/kg i.v., infusion 0.2-0.6 mg/kg/h
If still not controlled, consider pentobarbital
Stage 5 – Super-refractory SE*
Ethics. Discuss continuously how far treatment shall go, must be related to underlying pathology and an evaluation of the prognosis. When should active treatment be ended in the individual case?
Should be tried/discussed
Ketamine (loading dose 2.5 mg/kg (1-3), continuous infusion at 3 mg/kg/h until burst suppression in EEG or up to a maximum dose of 10 mg/kg/h.
Ketogenic diet. Ketosis usually reached within 2-3 days. Evaluate effect after 3-5 days in ketosis. If not effective after 5 days, stop treatment
Immunotherapy; steroids, immunoglobulins, rituximab, anakinra
Inhalation anesthetics (isoflurane) (low evidence, can be discussed)
Alternative ASMs
Lacosamide, 400 mg i.v. followed by 200 mg x 2 until se-concentration measurements
Perampanel, up to 36 mg through ngs, reduce gradually to 12 mg/day after 2 days. Adjustments according to se-concentrations
Brivaracetam, 100-300 mg iv loading dose?
Other treatment alternatives to be discussed in individual case (very low evidence)
 Magnesium
 Hypothermia
 Pyridoxine (May be relevenat also for adults with uncontrolled seizures/SE)
 Lidocain
 Electroconvulsive treatment (ECT)?
End treatment? Ethical discussions

*No consensus on doses and infusion rates. Suggestions based on literature search and personal experience. Should be discussed individually in each case.

Furthermore, it is essential to quickly try to identify the underlying cause of SE, and to detect possible factors that may have triggered the current SE episode. Treatment should then be directed towards these factors, the pathological cause (if detectable) and, naturally, neuronal hyperactivity. Triggering factors and causes of SE will often be identified as metabolic abnormalities, fever, change of medication, non-compliance, intoxications, and different types of brain insults.

It is important to stop both clinical and subclinical neuronal hyperactivity. Most current rational treatment programs are geared towards this by influencing glutamatergic and GABA-ergic mechanisms. However, SE is based on a complex and yet mostly unclear range of mechanisms reaching far beyond glutamatergic and GABAergic imbalance, including genetic, epigenetic, molecular, and other cellular, intercellular and network alterations. As an example, it has been shown in an experimental epilepsy model using lithium-pilocarpine in rats, SE ceased when scopolamine was administered together with diazepam and phenobarbital (128).

With the complex mechanisms involved, several forms of rational polytherapy should probably be tried out. It is only the combination of diazepam and PHT that has been shown to be effective in humans (type 1 evidence) (129). We recommend that different combinations of therapies should be tested systematically in both animal models and clinical studies, and that clinical data should be collected in an international database. This would reduce the large number of “try and error treatment” attempts and would also counteract publication bias.

The primary cause of SE should be in greater focus. As an example, we are currently witnessing an increasing number of putative immunological causes for SE, opening for more specific diagnostic tools and more personalized treatment options in the future.

A highly important matter is the discussion of the ethical aspects of SRSE treatment. Treatment facilities dealing with SRSE should discuss ethical issues profoundly and define procedures to aid more streamlined treatment approaches including how many trials to use and when to stop the treatment. This is not an easy task as the origin of SRSE seems to be highly individual and by this makes general guidelines difficult. Some of the ethical questions are: what is right for the patient? What is ethical for family and relatives? What is ethical for other patients also requiring the often limited ICU resources in most hospitals? These considerations and questions have been excellently discussed by Crippen et al (130). One major challenge is to distinguish at an early stage between patients with a good prognosis and potential for a worthy life, versus those patients expecting severe brain damage or death. Presently no clear risk factors or biomarkers are available that are near to allow distinction between these groups. The suggested prognostic scoring systems like, STESS and EMSE do not hold sufficient sensitivity and specificity. This poses a particular challenge to decision-making on an individual basis.

Regarding SRSE, there are probably cases where the cause of the condition is so unique that it is very difficult, or even impossible, to either diagnose, or to treat. Other cases of SRSE, however, probably represent RSE that has been developing and that could, perhaps, have been prevented by better treatment at an earlier stage. This underlines the importance of prompt and appropriate treatment from the moment a patient with SE enters the clinic.

Conflict of Interest: The authors declare no potential conflict of interest with respect to research, authorship and/or publication of this chapter.

Copyright and Permission Statement: The authors confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s), and all original sources have been appropriately acknowledged or referenced.

REFERENCES

  1. Meldrum BS, Horton RW. Physiology of status epilepticus in primates. Arch Neurol 1973;28(1):1–9. https://doi.org/10.1001/archneur.1973.00490190019001
  2. Epilepsy Foundation of America. Treatment of convulsive status epilepticus. Recommendations of the Epilepsy Foundation of America’s Working Group on Status Epilepticus. JAMA 1993;270(7):854–9. https://doi.org/10.1001/jama.270.7.854
  3. International League Against Epilepsy. Guidelines for epidemiologic studies on epilepsy. Commission on Epidemiology and Prognosis. Epilepsia 1993;34(4):592–6. https://doi.org/10.1111/j.1528-1157.1993.tb00433.x
  4. Trinka E, Cock H, Hesdorffer D, Rossetti AO, Scheffer IE, Shinnar S, et al. A definition and classification of status epilepticus - Report of the ILAE Task Force on Classification of Status Epilepticus. Epilepsia 2015;56(10):1515–23. https://doi.org/10.1111/epi.13121
  5. Leitinger M, Trinka E, Giovannini G, Zimmermann G, Florea C, Rohracher A, et al. Epidemiology of status epilepticus in adults: A population-based study on incidence, causes, and outcomes. Epilepsia 2019;60(1):53–62. https://doi.org/10.1111/epi.14607
  6. Knake S, Rosenow F, Vescovi M, Oertel WH, Mueller HH, Wirbatz A, et al. Incidence of status epilepticus in adults in Germany: a prospective, population-based study. Epilepsia 2001;42(6):714–8. https://doi.org/10.1046/j.1528–1157.2001.01101.x
  7. Coeytaux A, Jallon P, Galobardes B, Morabia A. Incidence of status epilepticus in French-speaking Switzerland: (EPISTAR). Neurology 2000;55(5):693–7. https://doi.org/10.1212/WNL.55.5.693
  8. Vignatelli L, Tonon C, D’Alessandro R. Bologna Group for the Study of Status Epilepticus. Incidence and short-term prognosis of status epilepticus in adults in Bologna, Italy. Epilepsia 2003;44(7):964–8. https://doi.org/10.1046/j.1528-1157.2003.63702.x
  9. Logroscino G, Hesdorffer DC, Cascino G, Annegers JF, Hauser WA. Short-term mortality after a first episode of status epilepticus. Epilepsia 1997;38(2):1344–49. https://doi.org/10.1111/j.1528-1157.1997.tb00073.x
  10. Betjemann JP, Lowenstein DH. Status epilepticus in adults. Lancet Neurol 2015;14(6):615–24. https://doi.org/10.1016/S1474-4422(15)00042-3
  11. Giovannini G, Monti G, Polisi MM, Mirandola L, Marudi A, Pinelli G, et al. A one-year prospective study of refractory status epilepticus in Modena, Italy. Epilepsy Behav 2015;49:141–5. https://doi.org/10.1016/j.yebeh.2015.05.022
  12. Delaj L, Novy J, Ryvlin P, Marchi NA, Rossetti AO. Refractory and super-refractory status epilepticus in adults: a 9-year cohort study. Acta Neurol Scand 2017;135(1):92–9. https://doi.org/10.1111/ane.12605
  13. Ulvin LB, Heuser K, Olsen KB, Taubøll E. Factors associated with refractoriness and outcome in an adult status epilepticus cohort. Seizure 2018;61:111–8. https://doi.org/10.1016/j.seizure.2018.07.020
  14. Power KN, Gramstad A, Gilhus NE, Engelsen BA. Adult nonconvulsive status epilepticus in a clinical setting: Semiology, aetiology, treatment and outcome. Seizure 2015;24:102–6. https://doi.org/10.1016/j.seizure.2014.09.007
  15. Gramstad A, Power KN, Engelsen BA. Neuropsychological Performance 1 Year After Status Epilepticus in Adults. Arch Clin Neuropsychol 2021;36(3):329–38. https://doi.org/10.1093/arclin/acz069
  16. Ferlisi M, Shorvon S. The treatment of super-refractory status epilepticus: a critical review of available therapies and a clinical treatment protocol. Brain 2011;134(Pt 10):2802–18. https://doi.org/10.1093/brain/awr215
  17. Naylor DE, Liu H, Wasterlain CG. Trafficking of GABA(A) receptors, loss of inhibition, and a mechanism for pharmacoresistance in status epilepticus. J Neurosci 2005;25(34):7724–33. https://doi.org/10.1523/JNEUROSCI.4944-04.2005
  18. Naylor DE, Liu H, Niquet J, Wasterlain CG. Rapid surface accumulation of NMDA receptors increases glutamatergic excitation during status epilepticus. Neurobiol Dis 2013;54:225–38. https://doi.org/10.1016/j.nbd.2012.12.015
  19. Delgado-Escueta AV, Wasterlain C, Treiman D, Porter R. Status epilepticus: summary. Adv Neurol 1983 34:537-41.20. Kälviäinen R, Eriksson K, Parviainen I. Refractory generalized convulsive status epilepticus: a guide to treatment. CNS Drugs 2005;19(9):759–68. https://doi.org/10.2165/00023210-200519090-00003
  20. Meierkord H, Boon P, Engelsen B, Göcke K, Shorvon S, Tinuper P, et al. EFNS guideline on the management of status epilepticus. Eur J Neurol 2006;13(5):445–50. https://doi.org/10.1111/j.1468-1331.2006.01397.x
  21. Meierkord H, Boon P, Engelsen B, Göcke K, Shorvon S, Tinuper P, et al. EFNS guideline on the management of status epilepticus in adults. Eur J Neurol 2010;17(3): 348–55. https://doi.org/10.1111/j.1468-1331.2009.02917.x
  22. Rossetti AO. Treatment options in the management of status epilepticus. Curr Treat Options Neurol 2010;12(2):100–12. https://doi.org/10.1007/s11940-010-0060-2
  23. Brophy GM, Bell R, Claassen J, Alldredge B, Bleck TP, Glauser T, et al. Guidelines for the Evaluation and Management of Status Epilepticus. Neurocrit Care 2012;17(1):3–23. https://doi.org/10.1007/s12028-012-9695-z
  24. Shorvon S, Ferlisi M. The outcome of therapies in refractory and super-refractory convulsive status epilepticus and recommendations for therapy. Brain 2012;135(Pt8): 2314–28. https://doi.org/10.1093/brain/aws091
  25. Glauser T, Shinnar S, Gloss D, Alldredge B, Arya R, Bainbridge J, et al. Evidence-Based Guideline: Treatment of Convulsive Status Epilepticus in Children and Adults: Report of the Guideline Committee of the American Epilepsy Society. Epilepsy Curr 2016;16(1):48–61. https://doi.org/10.5698/1535-7597-16.1.48
  26. Rosenow F, Weber J. S2k guidelines: status epilepticus in adulthood: Guidelines of the German Society for Neurology. Nervenarzt 2021;92(10):1002–30. https://doi.org/10.1007/s00115-020-01036-2
  27. Rogalski R, Rogalski A. Benzodiazepine selection in the management of status epilepticus. A review. Adv Emerg Nurs J 2015;37(2): 83–94. https://doi.org/10.1097/TME.0000000000000064
  28. Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich S, et al. A comparison of lorazepam, diazepam, and placebo for the treatment of out-of-hospital status epilepticus. N Engl J Med 2001;345(9):631–7. https://doi.org/10.1056/NEJMoa002141
  29. Prasad M, Krishnan PR, Sequeira R, Al-Roomi K. Anticonvulsant therapy for status epilepticus. Cochrane Database Syst Rev 2014;2014(9):CD003723. https://doi.org/10.1002/14651858.CD003723.pub3
  30. Brigo F, Bragazzi NL, Bacigaluppi S, Nardone R, Trinka E. Is intravenous lorazepam really more effective and safe than intravenous diazepam as first-line treatment for convulsive status epilepticus? A systematic review with meta-analysis of randomized controlled trials. Epilepsy Behav. 2016;64(Pt A):29–36. https://doi.org/10.1016/j.yebeh.2016.09.020
  31. Higdon LM, Sperling MR. A review of a diazepam nasal spray for the treatment of acute seizure clusters and prolonged seizures. Expert Rev Neurother 2021:21(11):1207–12. https://doi.org/10.1080/14737175.2021.1965880
  32. Wheless JW. A critical evaluation of midazolam nasal spray for the treatment of patients with seizure clusters. Expert Rev Neurother 2021:21(11):1195–1205. https://doi.org/10.1080/14737175.2021.1890033
  33. Silbergleit R, Durkalski V, Lowenstein D, Conwit R, Pancioli A, Palesch Y, et al. Intramuscular versus intravenous therapy for prehospital status epilepticus. NETT Investigators. N Engl J Med 2012;366(7):591–600. https://doi.org/10.1056/NEJMoa1107494
  34. Alvarez V, Lee JW, Drislane FW, Westover MB, Novy J, Dworetzky BA, et al. Practice variability and efficacy of clonazepam, lorazepam, and midazolam in status epilepticus: A multicenter comparison. Epilepsia 2015 56(8):1275–85. https://doi.org/10.1111/epi.13056
  35. Trinka E, Kälviäinen R. 25 years of advances in the definition, classification and treatment of status epilepticus. Seizure 2017;44:65–73. https://doi.org/10.1016/j.seizure.2016.11.001
  36. Kapur J, Elm J, Chamberlain JM, Barsan W, Cloyd J, Lowenstein D, et al. Randomized Trial of Three Anticonvulsant Medications for Status Epilepticus. NETT and PECARN Investigators. N Engl J Med 2019;381(22):2103–13. https://doi.org/10.1056/NEJMoa1905795
  37. Vossler DG, Bainbridge JL, Boggs JG, Novotny EJ, Loddenkemper T, Faught E, et al. Treatment of Refractory Convulsive Status Epilepticus: A Comprehensive Review by the American Epilepsy Society Treatments Committee. Epilepsy Curr 2020;20(5):245–64. https://doi.org/10.1177/1535759720928269
  38. Trinka E, Höfler J, Zerbs, Brigo F. Efficacy and safety of intravenous valproate for status epilepticus: a systematic review. CNS Drugs 2014;28(7): 623–39. https://doi.org/10.1007/s40263-014-0167-1
  39. Rossetti AO, Bromfield EB. Levetiracetam in the treatment of status epilepticus in adults: a study of 13 episodes. Eur Neurol. 2005;54(1):34–8. https://doi.org/10.1159/000087385
  40. Möddel G, Bunten S, Dobis C, Kovac S, Dogan M, Fischera M, et al. Intravenous levetiracetam: a new treatment alternative for refractory status epilepticus. J Neurol Neurosurg Psychiatry 2009;80(6):689–92. https://doi.org/10.1136/jnnp.2008.145458
  41. Trinka E, Dobesberger J. New treatment options in status epilepticus: a critical review on intravenous levetiracetam. Ther Adv Neurol Disord 2009;2:79–91. https://doi.org/10.1177/1756285608100460
  42. Rantsch K, Walter U, Wittstock M, Benecke R, Rösche J. Treatment and course of different subtypes of status epilepticus. Epilepsy Res 2013;107(1-2):156–62. https://doi.org/10.1016/j.eplepsyres.2013.08.001
  43. Yasiry Z, Shorvon SD. The relative effectiveness of five antiepileptic drugs in treatment of benzodiazepine-resistant convulsive status epilepticus: a meta-analysis of published studies. Seizure 2014;23(3):167–74. https://doi.org/10.1016/j.seizure.2013.12.007
  44. Eue S, Grumbt M, Müller M, Schulze A. Two years of experience in the treatment of status epilepticus with intravenous levetiracetam. Epilepsy Behav 2009;15(4):467–9. https://doi.org/10.1016/j.yebeh.2009.05.020
  45. Chakravarthi S, Goyal MK, Modi M, Bhalla A, Singh P. Levetiracetam versus phenytoin in management of status epilepticus. J Clin Neurosci 2015;22(6):959–963. https://doi.org/10.1016/j.jocn.2014.12.013
  46. Alvarez V, Januel JM, Burnand B, Rossetti AO. Second-line status epilepticus treatment: comparison of phenytoin, valproate, and levetiracetam. Epilepsia 2011;52(7):1292–6. https://doi.org/10.1111/j.1528-1167.2011.03056.x
  47. Shorvon S. Clinical trials in acute repetitive seizures and status epilepticus. Epileptic Disorders 2012;14(2):138–47. https://doi.org/10.1684/epd.2012.0514
  48. Hocker S, Clark S, Britton J. Parenteral phenobarbital in status epilepticus revisited: Mayo Clinic experience. Epilepsia 2018;59 (Suppl 2):193–7. https://doi.org/10.1111/epi.14488
  49. Chamberlain JM, Kapur J, Shinnar S, Elm J, Holsti M, Babcock L, et al. Efficacy of levetiracetam, fosphenytoin, and valproate for established status epilepticus by age group (ESETT): a double-blind, responsive-adaptive, randomised controlled trial. Lancet 2020;395(10231):1217–1224. https://doi.org/10.1016/S0140-6736(20)30611-5
  50. Rahman S. Mitochondrial disease and epilepsy. Dev Med Child Neurol 2012;54(5):397–406. https://doi.org/10.1111/j.1469-8749.2011.04214.x
  51. Zaccara G, Giorgi FS, Amantini A, Giannasi G, Campostrini R, Giovannelli F, et al. Why we prefer levetiracetam over phenytoin for treatment of status epilepticus. Acta Neurol Scand 2018;137(6):618–622. https://doi.org/10.1111/ane.12928
  52. Sathe AG, Elm JJ, Cloyd JC, Chamberlain JM, Silbergleit R, Kapur J, et al. The association of patient weight and dose of fosphenytoin, levetiracetam, and valproic acid with treatment success in status epilepticus. Epilepsia 2020;61(6):e66–e70. https://doi.org/10.1111/epi.16534
  53. Rossetti AO, Bromfield EB. Determinants of success in the use of oral levetiracetam in status epilepticus. Epilepsy Behav 2006;8(3):651–654. https://doi.org/10.1016/j.yebeh.2006.01.006
  54. Uberall MA, Trollmann R, Wunsiedler U, Wenzel D. Intravenous valproate in pediatric epilepsy patients with refractory status epilepticus. Neurology 2000;54(11):2188–9. https://doi.org/10.1212/WNL.54.11.2188-a
  55. Alvarez V, Lee JW, Westover MB, Drislane FW, Novy J, Faouzi M, et al. Therapeutic coma for status epilepticus: Differing practices in a prospective multicenter study. Neurology 2016;87(16):1650–9.https://doi.org/10.1212/WNL.0000000000003224
  56. De Stefano P, Baumann SM, Semmlack S, Rüegg S, Marsch S, Seeck M, et al. Safety and Efficacy of Coma Induction Following First-Line Treatment in Status Epilepticus: A 2-Center Study. Neurology 2021;97(6):e564–e576. https://doi.org/10.1212/WNL.0000000000012292
  57. Novy J, Logroscino G, Rossetti AO. Refractory status epilepticus: a prospective observational study. Epilepsia 2010;51(2):251–6. https://doi.org/10.1111/j.1528-1167.2009.02323.x
  58. Mundlamuri RC, Sinha S, Subbakrishna DK, Prathyusha PV, Nagappa M, Bindu PS et al. Management of generalised convulsive status epilepticus (SE): a prospective randomised controlled study of combined treatment with intravenous lorazepam with either phenytoin, sodium valproate or levetiracetam-pilot study. Epilepsy Res 2015;114(1):52–8. https://doi.org/10.1016/j.eplepsyres.2015.04.013
  59. Sutter R, Marsch S, Fuhr P, Kaplan PW, Ruegg S. Anesthetic drugs in status epilepticus: risk or rescue? A 6-year cohort study. Neurology 2014;82(8):656–64. https://doi.org/10.1212/WNL.0000000000000009
  60. Claassen J, Hirsch LJ, Emerson RG, Mayer SA. Treatment of refractory status epilepticus with pentobarbital, propofol, or midazolam: a systematic review. Epilepsia 2002;43(2):146–53. https://doi.org/10.1046/j.1528-1157.2002.28501.x
  61. Parviainen I, Kälviäinen R, Ruokonen E. Propofol and barbiturates for the anesthesia of refractory convulsive status epilepticus: pros and cons. Neurol Res 2007;29(7):667–71. https://doi.org/10.1179/016164107X240044
  62. Höfler J, Trinka E. Lacosamide as a new treatment option in status epilepticus. Epilepsia 2013;54(3):393–404. https://doi.org/10.1111/epi.12058
  63. Panda PK, Panda P, Dawman L, Sharawat IK. Efficacy of lacosamide and phenytoin in status epilepticus: A systematic review. Acta Neurol Scand 2021;144(4):366–74. https://doi.org/10.1111/ane.13469
  64. FDA. Vimpat prescribing information. . U.S. Food and Drug Administration. 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/022253s042lbl.pdf
  65. Kim HK, Lee H, Bae E-K, Kim DW. Cardiac effects of rapid intravenous loading of lacosamide in patients with epilepsy. Epilepsy Res 2021;176:106710. https://doi.org/10.1016/j.eplepsyres.2021.106710
  66. Perrenoud M, Andre P, Alvarez V, Stähli C, DecosterdLA, Sorressi AO et al. Intravenous lacosamide in status epilepticus: correlation between loading dose, serum levels, and clinical response. Epilepsy Res. 2017;135(1):38–42. https://doi.org/10.1016/j.eplepsyres.2017.05.007
  67. Chimakurthy AK, Ramsay RE, Sabharwal V, Menon U. Safety, tolerability, and pharmacokinetics of weight-based IV loading dose of lacosamide in the ICU. Epilepsy Behav 2021;114(Pt A):107449. https://doi.org/10.1016/j.yebeh.2020.107449
  68. Synowiec AS, Yandora KA, Yenugadhati V, Valeriano JP, Schramke CJ, Kelly KM. The efficacy of topiramate in adult refractory status epilepticus: Experience of a tertiary care center. Epilepsy Res 2012;98(2-3):232–7. https://doi.org/10.1016/j.eplepsyres.2011.09.017
  69. Hottinger A, Sutter R, Marsch S, Rüegg S. Topiramate as an adjunctive treatment in patients with refractory status epilepticus: an observational cohort study. CNS Drugs 2012;26(9):761–72. https://doi.org/10.2165/11633090-000000000-00000
  70. Fechner A, Hubert K, Jahnke K, Knake S, Konczalla J, Menzler K, et al. Treatment of refractory and superrefractory status epilepticus with topiramate: A cohort study of 106 patients and a review of the literature. Epilepsia 2019;60(12):2448–58. https://doi.org/10.1111/epi.16382
  71. Brigo F, Bragazzi NL, Igwe SC, Nardone R, Trinka E. Topiramate in the Treatment of Generalized Convulsive Status Epilepticus in Adults: A Systematic Review with Individual Patient Data Analysis. Drugs2017;77(1):67–74. https://doi.org/10.1007/s40265-016-0672-2
  72. Orlandi N, Bartolini E, Audenino D, Coletti Moja M, Urso L, d’Orsi G, et al. Intravenous brivaracetam in status epilepticus: A multicentric retrospective study in Italy. Seizure 2021;86:70–6. https://doi.org/10.1016/j.seizure.2021.01.014
  73. Willems LM, Bauer S, Rosenow F, Strzelczyk A. Recent advances in the pharmacotherapy of epilepsy: brivaracetam and perampanel as broad-spectrum antiseizure drugs for the treatment of epilepsies and status epilepticus. Expert Opin Pharmacother 2019;20(14):1755–65. https://doi.org/10.1080/14656566.2019.1637420
  74. Lim SN, Wu T, Tseng WJ, Chiang HI, Cheng MY, Lin WR, et al. Efficacy and safety of perampanel in refractory and super-refractory status epilepticus: cohort study of 81 patients and literature review. J Neurol 2021;268(10):3744–57. https://doi.org/10.1007/s00415-021-10506-9
  75. Wasterlain CG, Chen JWY. Mechanistic and pharmacologic aspects of status epilepticus and its treatment with new antiepileptic drugs. Epilepsia 2008;49(Suppl 9): 63–73. https://doi.org/10.1111/j.1528-1167.2008.01928.x
  76. Amengual-Gual M, Sanchez Fernandez I, Wainwright MS. Novel drugs and early polypharmacotherapy in status epilepticus. Seizure 2019;68(3):79–88. https://doi.org/10.1016/j.seizure.2018.08.004
  77. Fujikawa DG. Starting ketamine for neuroprotection earlier than its current use as an anesthetic/antiepileptic drug late in refractory status epilepticus. Epilepsia 2019;60(5):373–80. https://doi.org/10.1111/epi.14676
  78. Gaspard N, Foreman B, Judd LM, Brenton JN, Nathan BR, McCoy BM, et al. Intravenous ketamine for the treatment of refractory status epilepticus: a retrospective multicenter study. Epilepsia 2013;54(8):1498–503. https://doi.org/10.1111/epi.12247
  79. Alkhachroum A, Der-Nigoghossian CA, Mathews E, Massad N, Letchinger R, Doyle K, et al. Ketamine to treat super-refractory status epilepticus. Neurology 2020;95(16):e2286–e94. https://doi.org/10.1212/WNL.0000000000010611
  80. Kramer AH. Early ketamine to treat refractory status epilepticus. Neurocrit Care 2012;16(2):299–305. https://doi.org/10.1007/s12028-011-9668-7
  81. Rosati A, L’Erario M, Ilvento L, Cecchi C, Pisano T, Mirabile L, et al. Efficacy and safety of ketamine in refractory status epilepticus in children. Neurology 2012;79(24):2355–8. https://doi.org/10.1212/WNL.0b013e318278b685
  82. Synowiec AS, Singh DS, Yenugadhati V, Valeriano JP, Schramke CJ, Kelly KM. Ketamine use in the treatment of refractory status epilepticus. Epilepsy Res 2013;105(1-2):183–8. https://doi.org/10.1016/j.eplepsyres.2013.01.007
  83. Yan J, Jiang H. Dual effects of ketamine: neurotoxicity versus neuroprotection in anesthesia for the developing brain. J Neurosurg Anesthesiol 2014;26(2):155–60. https://doi.org/10.1097/ANA.0000000000000027
  84. Zeiler FA. Early use of the NMDA receptor antagonist ketamine in refractory and superrefractory status epilepticus. Crit Care Res Pract 2015e;2015e: 831260. https://doi.org/10.1155/2015/831260
  85. Borris DJ, Bertram EH, Kapur J. Ketamine controls prolonged status epilepticus. Epilepsy Res 2000;42(2-3):117–22. https://doi.org/10.1016/S0920-1211(00)00175-3
  86. Rasmussen T, Olszewski J, Lloydsmith D. Focal seizures due to chronic localized encephalitis. Neurology 1958;8(6):435–45. https://doi.org/10.1212/WNL.8.6.435
  87. Vezzani A. Epilepsy and inflammation in the brain: overview and pathophysiology. Epilepsy Currs 2014;14(1 Suppl):3–7. https://doi.org/10.5698/1535-7511-14.s2.3
  88. Vezzani A, Balosso S, Ravizza T. Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol 2019;15(8):459–72. https://doi.org/10.1038/s41582-019-0217-x
  89. Tan TH, Perucca P, O’Brien TJ, Kwan P, Monif M. Inflammation, ictogenesis, and epileptogenesis: An exploration through human disease. Epilepsia 2021;62(2):303–24. https://doi.org/10.1111/epi.16788
  90. Bien C, Bauer J. Autoimmune epilepsies. Neurotherapeutics 2014;11(2): 311–8. https://doi.org/10.1007/s13311-014-0264-3
  91. Khawaja AM, DeWolfe JL, Miller DW, Szaflarski JP. New-onset refractory status epilepticus (NORSE) - The potential role for immunotherapy. Epilepsy Behav 2015;47:17–23. https://doi.org/10.1016/j.yebeh.2015.04.054
  92. Jang Y, Lee WJ, Lee HS, Chu K, Lee SK, Lee ST. Tofacitinib treatment for refractory autoimmune encephalitis. Epilepsia 2021;62(4):e53–e59. https://doi.org/10.1111/epi.16848
  93. Koh S, Wirrell E, Vezzani A, Nabbout R, Muscal E, Kaliakatsos M, et al. Proposal to optimize evaluation and treatment of Febrile infection-related epilepsy syndrome (FIRES): A Report from FIRES workshop. Epilepsia Open 2021;6(1):62–72. https://doi.org/10.1002/epi4.12447
  94. Yamanaka G, Ishida Y, Kanou K, Suzuki S, Watanabe Y, Takamatsu T, et al. Towards a Treatment for Neuroinflammation in Epilepsy: Interleukin-1 Receptor Antagonist, Anakinra, as a Potential Treatment in Intractable Epilepsy. Int J Mol Sci 2021;22(12):6282. https://doi.org/10.3390/ijms22126282
  95. Choi CH, Ma SH, Ma KK, Leung H, Mok VC. Super-refractory status epilepticus in autoimmune encephalitis treated with interleukin-1 receptor antagonist, anakinra. Epileptic Disord 2021;23(3):500–5. https://doi.org/10.1684/epd.2021.1283
  96. Mochol M, Taubøll E, Sveberg L, Tennøe B, Berg Olsen K, Heuser K, et al. Seizure control after late introduction of anakinra in a patient with adult onset Rasmussen’s encephalitis. Epilepsy Behav Rep 2021;16:100462. https://doi.org/10.1016/j.ebr.2021.100462
  97. Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, et al. Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis 2017;99:12–23. https://doi.org/10.1016/j.nbd.2016.12.007
  98. Taubøll E, Isojärvi JIT, Herzog AG. The interactions between reproductive hormones and epilepsy. Handb Clin Neurol 2021;182:155–174. https://doi.org/10.1016/B978-0-12-819973-2.00011-3
  99. Majewska MD, Harrison NL, Schwartz RD, Barker JL, Paul SM. Steroid hormone metabolites are barbiturate-like modulators of the GABA receptor. Science 1986;232(4753): 1004–1007. https://doi.org/10.1126/science.2422758
  100. Taubøll E, Lindström S. The effect of progesterone and its metabolite 5 alpha-pregnan-3 alpha-ol-20-one on focal epileptic seizures in the cat’s visual cortex in vivo. Epilepsy Res 1993;14(1):17–30.https://doi.org/10.1016/0920-1211(93)90071-E
  101. Taubøll E, Gjerstad L. Comparison of 5 alpha-pregnan-3 alpha-ol-20-one and phenobarbital on cortical synaptic activation and inhibition studied in vitro. Epilepsia 1993;34(2):228–35. https://doi.org/10.1111/j.1528-1157.1993.tb02403.x
  102. Joshi S, Kapur J. Neurosteroid regulation of GABA(A) receptors: A role in catamenial epilepsy. Brain Res 2019;1703:31–40. https://doi.org/10.1016/j.brainres.2018.02.031
  103. Rosenthal ES, Claassen J, Wainwright MS, Husain AM, Vaitkevicius H, Raines S, et al. Brexanolone as adjunctive therapy in super-refractory status epilepticus. Ann Neurol 2017;82(3):342–352. https://doi.org/10.1002/ana.25008
  104. Zeiler FA, Zeiler KJ, Kazina CJ, Teitelbaum J, Gillman LM, West M. Lidocaine for status epilepticus in adults. Seizure. 2015a;31:41–8. https://doi.org/10.1016/j.seizure.2015.07.003
  105. Zeiler FA, Matuszczak M, Teitelbaum J, Gillman LM, Kazina CJ. Magnesium sulfate for non-eclamptic status epilepticus. Seizure. 2015b, Nov;32:100–8. https://doi.org/10.1016/j.seizure.2015.09.017
  106. Zeiler FA, Zeiler KJ, Teitelbaum J, Gillman LM, West M. Therapeutic Hypothermia for Refractory Status Epilepticus. Can J Neurol Sci 2015c;42(4):221–9. https://doi.org/10.1017/cjn.2015.31
  107. Zeiler FA, Zeiler KJ, Teitelbaum J, Gillman LM, West M. Modern inhalational anesthetics for refractory status epilepticus. Can J Neurol Sci 2015d; 42(2):106–15. https://doi.org/10.1017/cjn.2014.121
  108. Alolayan YS, McKinley K, Bhatia R, Alkhachroum A. Review and Updates on the Treatment of Refractory and Super Refractory Status Epilepticus. J Clin Med 2021;10(14):3028. https://doi.org/10.3390/jcm10143028
  109. Kirmani BF, Au K, Ayari L, John M, Shetty P, Delorenzo RJ. Super-Refractory Status Epilepticus: Prognosis and Recent Advances in Management. Aging Dis 2021;12(4):1097–1119. https://doi.org/10.14336/AD.2021.0302
  110. Nowak L, Bregestovski P, Ascher P, Herbet A, Prochiantz A. Magnesium gates glutamate-activated channels in mouse central neurones. Nature 1984;307(5950):462–5. https://doi.org/10.1038/307462a0
  111. Visser NA, Braun KP, Leijten FS, van Nieuwenhuizen O, Wokke JH, van den Bergh WM. Magnesium treatment for patients with refractory status epilepticus due to POLG1-mutations. J Neurol 2011;258(2):218–22. https://doi.org/10.1007/s00415-010-5721-2
  112. Mirsattari SM, Sharpe MD, Young GB. Treatment of refractory status epilepticus with inhalational anesthetic agents isoflurane and desflurane. Arch Neurol 2004;61(8):1254–9. https://doi.org/10.1001/archneur.61.8.1254
  113. Caraballo RH, Flesler S, Armeno M, Fortini S, Agustinho A, Mestre G, et al. Ketogenic diet in pediatric patients with refractory focal status epilepticus. Epilepsy Res 2014;108(10):1912–6. https://doi.org/10.1016/j.eplepsyres.2014.09.033
  114. Kossoff EH, Zupec-Kania BA, Auvin S, Ballaban-Gil KR, Bergqvist AC, Blackford R, et al. Optimal clinical management of children receiving dietary therapies for epilepsy: Updated recommendations of the International Ketogenic Diet Study Group. Epilepsia Open 2018;3(2):175–92. https://doi.org/10.1002/epi4.12225
  115. Thakur KT, Probasco JC, Hocker SE, Roehl K, Henry B, Kossoff EH et al. Ketogenic diet for adults in super-refractory status epilepticus. Neurology 2014;82(8): 665–70. https://doi.org/10.1212/WNL.0000000000000151
  116. Cervenka MC, Hocker S, Koenig M, Bar B, Henry-Barron B, Kossoff EH, et al. Phase I/II multicenter ketogenic diet study for adult superrefractory status epilepticus. Neurology. 2017;88(10):938–43.https://doi.org/10.1212/WNL.0000000000003690
  117. Park EG, Lee J, Lee J. The ketogenic diet for super-refractory status epilepticus patients in intensive care units. Brain Dev 2019;41(5):420–7. https://doi.org/10.1016/j.braindev.2018.12.007
  118. Kverneland M, Taubøll E, Molteberg E, Veierød MB, Selmer KK, Nakken KO, et al. Pharmacokinetic interaction between modified Atkins diet and antiepileptic drugs in adults with drug-resistant epilepsy. Epilepsia 2019;60(11):2235–2244. https://doi.org/10.1111/epi.16364
  119. Baumeister FA, Oberhoffer R, Liebhaber GM, Kunkel J, Eberhardt J, Holthausen H, et al. Fatal propofol infusion syndrome in association with ketogenic diet. Neuropediatrics 2004;35(4):250–2. doi: 10.1055/s-2004-820992. https://doi.org/10.1055/s-2004-820992
  120. Corry JJ, Dhar R, Murphy T, Diringer MN. Hypothermia for refractory status epilepticus. Neurocrit Care 2008;9(2):189–97. https://doi.org/10.1007/s12028-008-9092-9
  121. Legriel S, Lemiale V, Schenck M, Chelly J, Laurent V, Daviaud F, et al. Hypothermia for Neuroprotection in Convulsive Status Epilepticus. N Engl J Med 2016;375(25):2457–67. doi: 10.1056/NEJMoa1608193.https://doi.org/10.1056/NEJMoa1608193
  122. Lambrecq V, Villéga F, Marchal C, Michel V, Guehl D, Rotge J-Y, et al. Refractory status epilepticus: electroconvulsive therapy as a possible therapeutic strategy. Seizure 2012;21(9): 661–4. https://doi.org/10.1016/j.seizure.2012.07.010
  123. Trinka E, Brigo F. Neurostimulation in the treatment of refractory and super-refractory status epilepticus. Epilepsy Behav 2019;101(Pt B):106551. https://doi.org/10.1016/j.yebeh.2019.106551
  124. Tan HJ, Tee TY, Husin M, Khoo CS, Woon LS. A case series of super-refractory status epilepticus successfully treated with electroconvulsive therapy. Epileptic Disord 2020;22(6):828–33. https://doi.org/10.1684/epd.2020.1233
  125. García-López B, Gómez-Menéndez AI, Vázquez-Sánchez F, Pérez-Cabo E, Isidro-Mesas F, Zabalegui-Pérez A, et al. Electroconvulsive Therapy in Super Refractory Status Epilepticus: Case Series with a Defined Protocol. Int J Environ Res Public Health 2020;17(11):4023. https://doi.org/10.3390/ijerph17114023
  126. Stavropoulos I, Lim Pak H, Valentin A. Neuromodulation in Super-refractory Status Epilepticus. J Clin Neurophysiol 2021;38(6):494–502. https://doi.org/10.1097/WNP.0000000000000710
  127. Brandt C, Töllner K, Klee R, Bröer S, Löscher W. Effective termination of status epilepticus by rational polypharmacy in the lithium-pilocarpine model in rats: window of opportunity to prevent epilepsy and prediction of epilepsy by biomarkers. Neurobiol Dis 2015;75:78–90. https://doi.org/10.1016/j.nbd.2014.12.015
  128. Treiman DM, Meyers PD, Walton NY, Collins JF, Colling C, Rowan AJ, et al. A comparison of four treatments for generalized convulsive status epilepticus. Veterans Affairs Status Epilepticus Cooperative Study Group. N Engl J Med 1998;339(12):792–8. https://doi.org/10.1056/NEJM199809173391202
  129. Crippen D, Burrows D, Stocchetti N, Mayer SA, Andrews P, Bleck, et al. Ethics roundtable: ‘open-ended ICU care: can we afford it?’ Crit Care 2010;14(3):222. https://doi.org/10.1186/cc8962