Skip to main content
Erschienen in: Critical Care 1/2018

Open Access 01.12.2018 | Review

Antiepileptic drugs in critically ill patients

verfasst von: Salia Farrokh, Pouya Tahsili-Fahadan, Eva K. Ritzl, John J. Lewin III, Marek A. Mirski

Erschienen in: Critical Care | Ausgabe 1/2018

Abstract

Background

The incidence of seizures in intensive care units ranges from 3.3% to 34%. It is therefore often necessary to initiate or continue anticonvulsant drugs in this setting. When a new anticonvulsant is initiated, drug factors, such as onset of action and side effects, and patient factors, such as age, renal, and hepatic function, should be taken into account. It is important to note that the altered physiology of critically ill patients as well as pharmacological and nonpharmacological interventions such as renal replacement therapy, extracorporeal membrane oxygenation, and target temperature management may lead to therapeutic failure or toxicity. This may be even more challenging with the availability of newer antiepileptics where the evidence for their use in critically ill patients is limited.

Main body

This article reviews the pharmacokinetics and pharmacodynamics of antiepileptics as well as application of these principles when dosing antiepileptics and monitoring serum levels in critically ill patients. The selection of the most appropriate anticonvulsant to treat seizure and status epileptics as well as the prophylactic use of these agents in this setting are also discussed. Drug-drug interactions and the effect of nonpharmacological interventions such as renal replacement therapy, plasma exchange, and extracorporeal membrane oxygenation on anticonvulsant removal are also included.

Conclusion

Optimal management of antiepileptic drugs in the intensive care unit is challenging given altered physiology, polypharmacy, and nonpharmacological interventions, and requires a multidisciplinary approach where appropriate and timely assessment, diagnosis, treatment, and monitoring plans are in place.
Abkürzungen
AAN
American Academy of Neurology
AED
Antiepileptic drug
AHA
American Heart Association
cEEG
Continuous electroencephalography
CRRT
Continuous renal replacement therapy
CYP450
Cytochrome P450
ECMO
Extracorporeal membrane oxygenation
EEG
Electroencephalography
GABA
Gamma-aminobutyric acid
ICH
Intracerebral hemorrhage
ICP
Intracranial pressure
ICU
Intensive care unit
IHD
Intermittent hemodialysis
NCSE
Nonconvulsive status epilepticus
PLEX
Plasmapheresis
RRT
Renal replacement therapy
SAH
Subarachnoid hemorrhage
SE
Status epilepticus
TBI
Traumatic brain injury
TDM
Therapeutic drug monitoring

Background

The incidence of seizures in general intensive care units (ICUs) ranges from 3.3% to 34% [1]. Risk factors include common diagnoses such as brain tumor, head trauma, stroke, history of seizure, electrolyte abnormalities, hypoglycemia, infections, and drug overdose or withdrawal [2]. Thus, ICU management of antiepileptic drugs (AEDs) is routinely practiced by intensive care providers.
Selection of the most effective AED with the least adverse events, however, can often be challenging. About a third of patients with seizures fail on monotherapy, necessitating two or more AEDs [3]. Several factors commonly seen in the critical care setting, such as polypharmacy, unpredictable medication pharmacokinetics, and implementation of a variety of nonpharmacological interventions, may lead to drug-drug interactions, elevated risk for drug toxicity, and subtherapeutic drug serum levels; these are discussed in this review.

Pharmacodynamics and pharmacokinetics of AEDs

Pharmacodynamics

AEDs depress abnormal neuronal firing by various mechanisms of action including altering ion channel activity, enhancing gamma-aminobutyric acid (GABA)-mediated inhibition, or reducing glutamate-mediated excitation. While some AEDs have a single mechanism of action, others have multiple mechanisms of action and, in some, the exact mechanism of action is not yet known (Table 1).
Table 1
Mechanism of action of antiepileptic drugs [113]
Antiepileptic drug agent
Presumed mechanism of action
Brivaracetam
SV2A modulation
Carbamazepine
Na+ channel blockade
Clobazam
GABA potentiation
Clonazapam
GABA potentiation
Diazepam
GABA potentiation
Fosphenytoin/phenytoin
Na+ channel blockade
Lacosamide
Enhanced slow inactivation of voltage-gated Na+ channels
Lamotrigine
Na+ channel blockade
Levetiracetam
SV2A modulation
Lorazepam
GABA potentiation
Midazolam
GABA potentiation
Oxcarbazepine
Na+ channel blockade
Pentobarbital
GABA potentiation
Perampanel
AMPA glutamate receptor antagonist
Phenobarbital
GABA potentiation
Topiramate
Na+ channel blockade, GABA potentiation, AMPA/Kainate glutamate antagonist
Valproic acid
GABA potentiation, glutamate (NMDA) inhibition, sodium channel and T-type calcium channel blockade
Zonisamide
Na+ and Ca2+ channel blockade
AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, GABA gamma-aminobutyric acid, NMDA, N-methyl-d-aspartate, SV2A synaptic vesicle glycoprotein 2A
When using multiple AEDs, it is reasonable to select medications with different mechanisms of action.

Pharmacokinetics (absorption, distribution, metabolism, excretion)

Alterations in normal physiology and the physical properties of medications can affect the rate and extent of enteral absorption of medications in the ICU, leading to a need for parenteral administration. Deranged gastrointestinal absorption may be expected in circumstances of decreased blood flow, intestinal atrophy, dysmotility, and interactions with enteral nutrition. Pharmacokinetic parameters of selected AEDs, including their oral bioavailability, are summarized in Table 2.
Table 2
Bioavailability and pharmacokinetic data of antiepileptic drugs [113]
Antiepileptic drug agent
Bioavailability (PO formulation)
Protein binding
Half-life
Metabolism
Elimination
Brivaracetam
Almost completely absorbed
≤ 20%
~  9 h
Hydrolysis (primary route) CYP2C19
≥ 95% renally
Carbamazepine
75–85%
Total daily intravenous dose should be equivalent to 70% of previous total daily oral dose
75–95%
Range: 30–60 h
After autoinduction:
12–17 h
> 90% by CYP3A4
72% renally
Clobazam
87%
80–90%
36 to 42 h
Hepatic via CYP3A4 and to a lesser extent via CYP2C19
82% renally
Clonazapam
~ 90%
~ 85%
17–60 h
Glucuronide and sulfate conjugation
< 2% renally as unchanged drug
Diazepam
> 90%
98%
Parent drug: ~ 60 to 72 h; metabolite ~ 152 to 174 h
CYP3A4 and 2C19
Renally
Eslicarbazepine
> 90%
< 40%
13–20 h
Hydrolytic first-pass metabolism
90% renally
Fosphenytoin
100% (intramuscular formulation)
95–99%
12–28.9 h
CYP2C9 CYP2C19
Renally
Lacosamide
100%
< 15%
13 h
CYP3A4, CYP2C9, CYP2C19
95% renally
Lamotrigine
98%
55%
25–70 h
Conjugation
94% renally
Levetiracetam
100%
< 10%
6–8 h
Hydrolysis
66% renally
Lorazepam
90%
~ 91%
12–18 h
Conjugation
88% renally
Midazolam
     
Oxcarbazepine
Readily absorbed
40%
Active metabolite: 9–11 h
Glucoronidation
95% renally
Pentobarbital
45% to 70%
15–50 h
Hydroxylation and glucuronidation
< 1% renally as unchanged drug
Perampanel
Completely and rapidly absorbed
95%
~ 105 h
CYP 3A4/5 primary;
lesser extent CYP 1A2/2B6
22% renally
Phenytoin
20–90%
90–95%
7–42 h
CYP2C9, 2C19 (major) and 3A4 (minor)
< 5% renally as unchanged drug
Phenobarbital
~ 95–100%
50%
Longest half-life
46–136 h
CYP450 and UGT mediated
25–50% renally
Topiramate
80%
15–41%
IR: 21 h
ER: 31–56 h
No extensive metabolism
70% renally
Valproic acid
90%
80–90%
9–16 h
CYPs 2C9, 2C19, 2A6, UGT-glucuronidation
70–80% renally
Zonisamide
Rapid and complete absorption
40%
50–68 h
CYP 3A4
60% renally
CYP Cytochrome, ER Extended Release, IR Immediate Release, PO By mouth, UGT Uridine diphosphate-glucuronyltransferase
Changes in serum pH as well as both respiratory and renal failure affect the ionized state of many drugs, affecting their penetration across lipophilic-based membranes such as the blood-brain barrier. Bioavailability of active drugs is also affected by alterations in volume of distribution for hydrophilic medications, while hypoalbuminemia increases the unbound (free) fraction of highly albumin-bound medications.
Although drugs are commonly metabolized to less active compounds, prodrugs such as oxcarbazepine and fosphenytoin must be metabolized into their active forms carbamazepine and phenytoin, respectively. Drug metabolism generally occurs in two phases. Phase I involves nonsynthetic reactions to form a modified group. A cytochrome P450 (CYP450) enzyme is frequently involved in such oxidative reactions. Phase II involves synthetic reactions to conjugate the metabolite with an endogenous substance. Metabolism for most anticonvulsants occurs primarily in the liver and is dependent on hepatic blood flow, enzyme activity, and protein binding. Although critical illness often inhibits the CYP450 isoenzymes, drug metabolism may be enhanced over several days as in the cases of pentobarbital and phenytoin, resulting in potential subtherapeutic concentrations [46]. Induced hypothermia will also reduce the systemic clearance of many medications mediated by CYP450 (such as phenytoin or propofol), between 7 and 22% for every degree of Celsius below 37 [7].
Regardless of the route of administration, renal elimination of parent drugs or metabolites is the primary excretory pathway for most drugs. For patients on renal replacement therapy (RRT), the type of dialysis that is performed and the frequency/duration should also be considered (see section 8, special therapeutic considerations).

Dosing and therapeutic drug monitoring in the ICU setting

Dosing of AEDs should be individualized to achieve seizure control with minimal adverse effects. The 1-h postloading dose is commonly recommended as a time to measure peak serum concentration. Pharmacokinetic alterations are frequently observed in critically ill patients; hence, frequent therapeutic drug monitoring (TDM) may be required. Basic dosing and TDM recommendations for AEDs commonly used in the ICU are summarized below and in Table 3.
Table 3
Dosing and monitoring of commonly used antiepileptic drugs [8, 11, 15, 16]
Antiepileptic drug
Dose
Therapeutic drug level to treat epilepsy (μg/mL)
Monitoring
TDM clinical pearls
Phenytoin
Load: 15–20 mg/kg
Maintenance:
4–6 mg/kg/day
10–20
1 h postload or
~ 7–10 days after initiation of maintenance dose (may check earlier within 2–3 days in seizing patients to ensure their metabolism is not significantly different from average patient population)
At total concentrations > 20 μg/mL, nystagmus may occur.
In concentrations > 30 μg/mL, ataxia, slurred speech, and incoordination can be observed.
If total concentrations are above 40 μg/mL, coma is possible.
At concentrations > 50–60 μg/m drug induced seizures may occur
Valproic acid
Load: 20–40 mg/kg
Maintenance:
10–15 mg/kg/day
50–100 (levels as high as 175 are used in RSE)
1 h postload or
2–4 days after initiation of maintenance dose
At total concentrations > 75 μg/mL lethargy and ataxia may occur.
In concentrations > 100 μg/mL tremor is observed. Coma may occur if total serum concentrations are above 175 μg/mL.
Thrombocytopenia is a dose-related side effect that can be limited by reducing the dose
Phenobarbital
Load: 20 mg/kg
Maintenance: 1.5–2 mg/kg/day
(dose adjustment may be required in liver impairment due to reduced clearance)
15–40 (higher levels may be utilized in RSE)
1 h postload or
4–7 days after initiation of maintenance dose
CNS depression is a dose-related side effect. In concentrations > 60 μg/mL respiratory depression may occur
Pentobarbital
Load: 5–15 mg/kg
Maintenance: 0.5–5 mg/kg/h
1–5 (rarely used to assess clinical efficacy or toxicity)
May be used after discontinuation to monitor the residual effects of the drug
Drug levels have not been correlated with electroencephalography
CNS depression, respiratory depression, and hemodynamic instability are dose-related side effects
CNS central nervous system, RSE Refractory Status Epilepticus, TDM therapeutic drug monitoring

Phenytoin/fosphenytoin

Phenytoin has an average half-life of 24 h, but this ranges from 7 to 40 h, increasing with dose escalation due to its nonlinear kinetics [8]. Phenytoin is insoluble in water and is dissolved in a basic solution including ethylene glycol, the mixture being linked to tissue necrosis (“purple hand syndrome”) if extravasated [9]. Fosphenytoin, the prodrug of phenytoin, is water soluble and hence free of the toxic emulsion. This difference in solubility allows intramuscular and faster intravenous administration of fosphenytoin, but the need for plasma conversion to the active drug (phenytoin) results in a comparable time to peak plasma levels when compared with phenytoin administration itself [10].
Because phenytoin follows nonlinear or saturable metabolism pharmacokinetics, it is possible to attain excessive concentrations much easier than medications that follow linear pharmacokinetics. Phenytoin TDM is therefore clinically important in the critically ill and should be followed closely.
At normal serum levels, patients may experience minor central nervous system depression and adverse effects such as nystagmus, drowsiness, or fatigue. Beyond the normal target range, ataxia, slurred speech, and incoordination often occurs. Drug-induced seizure activity has been reported at concentrations over 50–60 μg/mL [8].
Typically, protein binding accounts for 90% of total plasma concentrations, hence the therapeutic range for unbound phenytoin (free) concentrations is 1–2 μg/mL. In patients suspected of having altered drug plasma protein binding, monitoring of free phenytoin serum concentration is of value.

Valproic acid

Although the accepted therapeutic range for total valproic acid concentration for seizure therapeutics is 50–100 μg/mL, levels up to 175 μg/mL have been suggested in cases of refractory status epilepticus (SE) cases. Concentration-related side effects include ataxia, lethargy, tremor, and coma [11]. The common adverse effects—thrombocytopenia and hyperammonemia (via carnitine depletion)—can often be limited by dose reduction and by carnitine replacement in the latter condition. Due to significant interpatient differences in valproic acid metabolism, there is a poor correlation between valproic acid dose and total serum concentrations [11, 12].
Valproic acid is highly (90–95%) protein bound and is saturable within the therapeutic range which results in higher unbound fractions at higher concentrations. Although not often monitored, a therapeutic free valproic acid range of 2.5–10 μg/mL can be used as an initial guide [11].

Phenobarbital

Phenobarbital has a long half-life of approximately 100 h, which limits its use when short-term AED use is desired. As with other AEDs, treatment of SE in particular may require higher than normal dosing, and this may reach upwards of even 10 mg/kg/day (serum levels of > 100 μg/mL) with solid efficacy demonstrated [13]. Concentration-related adverse effects of phenobarbital are sedation, confusion, and lethargy, with high doses leading to obtundation and respiratory depression [14]. As phenobarbital is only about 50% protein-bound, free drug monitoring is not warranted. However, in severe hepatic impairment (Child-Pugh score > 8) a decrease of 25–50% in the initial daily maintenance dose may be required [15].

Pentobarbital

Pentobarbital is often used in the ICU setting to treat SE or elevated intracranial pressure. Initiation of pentobarbital involves sequential bolus doses followed by a continuous infusion [16, 17]. The average half-life of pentobarbital in adults is reported to be about 22 h and it is 20–45% protein-bound. Serum pentobarbital TDM (reference range is 1–5 μg/mL) is of limited utility in determining treatment clinical response or toxicity; the direct measure of intracranial pressure (ICP) control or inducement of “burst suppression” by electroencephalography (EEG) are monitored instead. Serum concentrations, however, may be useful in assessing residual effects of pentobarbital-induced coma once discontinued [18].

Drug-drug interactions

AED-AED drug interactions

Administration of multiple AEDs is common in the ICU to manage refractory seizures and SE. Satisfactory seizure control is not achieved in 30–40% of patients with monotherapy, making it necessary to use two or more AEDs [19, 20]. Although synergistic anticonvulsant activity between medications may be desirable, concurrent use of multiple AEDs generally increases the risk of drug interactions. Table 4 lists common AED-AED drug-drug interactions.
Table 4
AED-AED drug interactions [113116]
 
CBZ
CNP
DZP
LAC
LAM
MDZ
OXZ
PEN
PMP
PHB
PHT
PFL
TOP
ZON
↓ZON
  
↓LAC
  
↓ZON
↓ZON
 
↓ZON
↓ZON
 
↓ZON
↑CBZ
VPA
↓VPA
? risk of absence seizures
  
↑ LAM
 
↓OXZ
 
↑VPA
↑PHB
↑PHT
↑PFL
↓VPA
↑CBZ
↓VPA
↓VPA
TOP
↓VPA
 
↑ DZP
  
↓MDZ
 
↓TOP
↓ PMP
↓TOP
↑PHT
  
↓TOP
↓TOP
PHT
↓↑CBZ
 
↓ DZP
↓LAC
↓ LAM
↓MDZ
 
↓PEN
↓ PMP
↓PHT
   
↓↑PHT
PHB
↓CBZ
 
↓ DZP
 
↓ LAM
↓MDZ
↓OXZ
      
PEN
↓CBZ
 
↓ DZP
  
↓MDZ
       
OXZ
↓CBZ
 
↑↓DZP
  
↓MDZ
       
↓OXZ
MDZ
↓MDZ
            
LAM
↓LAM
            
LAC
↓LAC
            
DZP
↓DZP
            
↑CBZ
CNP
             
BRV
↓BRV
            
↑ and ↓ indicate increased and decreased levels and/or effects, respectively
AED antiepileptic drug, CBZ carbamazepine, CNP clonazepam, GBT gabapentine, GTC generalized tonic-clonic, LAC lacosamide, LAM lamotrigine, LEV levetiracetam, OXZ oxcarbazepine, PMP perampanel, PHB phenobarbital, PHT phenytoin, PGB pregabalin, TOP topiramate, VPA valproic acid, ZON zonisamide

AED-nonAED interactions

Interactions of AEDs with other agents is seen more frequently with drugs that are metabolized in the liver, and therefore are more common with older AEDs. Tables 5 and 6 describe AED-nonAED and nonAED-AED interactions.
Table 5
Interaction of selected antiepileptic drugs with commonly used medications in the intensive care unit [113, 115, 117124]
Antiepileptic drug
Therapeutic group
Selected examples
Phenytoin, phenobarbital, carbamazepine
Psychotropic agents
↓ Amitriptyline, nortriptyline, imipramine, bupropion, paroxetine, citalopram, Haloperidol, chlorpromazine, olanzapine, risperidone, quetiapine, ziprasidone
Valproic acid
↑ Amitriptyline, nortriptyline, paroxetine
Topiramate
↑ Haloperidol
Phenytoin, phenobarbital, carbamazepine
Antimicrobials
↓ Doxycycline, metronidazole, itraconazole, retrovirals
Valproic acid
↑ Zidovudine
Phenytoin
Cardiovascular agents
↓ Amiodarone, nimodipine, diltiazem, verapamil, ticagrelor, atorvastatin, dabigatran, apixaban, rivaroxaban (↑ warfarin effects with phenytoin load, ↓ warfarin effects with maintenance dose of phenytoin)a
Lacosamide
Diltiazem, verapamil (risk of atrioventricular block/bradycardia), ↓ warfarin
Carbamazepine
↓ Nimodipine, diltiazem, verapamil, ticagrelor, atorvastatin, warfarin, dabigatran, apixaban and rivaroxaban
Phenobarbital
↓ Nimodipine, atorvastatin
Valproic acid
↑ Nimodipine, warfarin
Phenytoin, phenobarbital, carbamazepine
Analgesics
↓ Fentanyl, methadone
Phenytoin, phenobarbital, carbamazepine
Immunosuppressant
↓ Cyclosporine, sirolimus, tacrolimus, corticosteroids
↑ and ↓ indicate increased and decreased levels and/or effects, respectively
aThe interaction between phenytoin and warfarin is complicated and unpredictable. Close monitoring of the international normalized ratio (INR) as well as serum phenytoin levels are critical if this combination is clinically necessary. Upon initiation of phenytoin, there may be transient increases in the INR as a result of protein binding displacement of warfarin by phenytoin, and enhanced anticoagulant effect. This may be followed by a reduction in anticoagulant activity as result of phenytoin’s induction of warfarin metabolism [125, 126]
Table 6
Interaction of selected therapeutic classes with antiepileptic drugs [115, 117, 127132]
Therapeutic class
Selected examples
Antiepileptic drugs
Psychotropic agents
Fluoxetine, sertraline, trazodone
↑ Phenytoin
Trazodone, fluoxetine, risperidone
↑ Carbamazepine
Sertraline
↑ Valproic acid
Antimicrobials
Erythromycin, clarithromycin, ketoconazole, fluconazole
↑ Carbamazepine
Ritonavir
Sulfonamides
↑ Phenytoin
Carbapenems (imipenem, doripenem, meropenem, ertapenem)
↓ Valproic acid
Cardiovascular agents
Amiodarone
↑ Phenytoin (a dose reduction of approximately 25% is recommended)
Diltiazem
↑ Carbamazepine, phenytoin (risk of toxicity)
Clopidogrel
↑ Phenytoin
Analgesics
Acetaminophen
↓ Lamotrigine
Immunosuppressants
Methotrexate
↓ Valproic acid, carbamazepine
↑ and ↓ indicate increased and decreased levels and/or effects, respectively

Seizure prevention in the ICU

Traumatic brain injury

The incidence of post-traumatic seizures accounts for 6% of overall symptomatic epilepsy [2123] and is estimated to occur in up to 15% and 53% in civilian and military populations, respectively [24, 25]. Clinical studies have divided post-traumatic seizures into either immediate (< 24 h), early (within 1 week, with a seizure incidence of 2.1–16.9%), or late (with an incidence of 1.9% to > 30.0%) [22, 23, 26]).
Early seizures occur in about 10–15% of patients not treated with anticonvulsants, and this incidence can be reduced to about 2–3% with the use of phenytoin [27]. There is, however, little evidence that such use reduces the occurrence of late seizures or the incidence of death and neurologic disability [28]. It thus appears that anticonvulsants are suppressive during the early period following traumatic brain injury (TBI), but are not prophylactic for the time period beyond the first week. The American Academy of Neurology (AAN) [29] and the Brain Trauma Foundation [30] advocate prophylactic use of AED only during the first 7 days after a head injury [31, 32]. Phenytoin has been shown to decrease the incidence of early post-traumatic seizures [30] and to be more cost-effective than levetiracetam [33]. Although levetiracetam was shown in small early studies to be as effective as phenytoin [33, 34], there are recent data suggesting a lack of any prophylactic action in a comparative study of levetiracetam versus no prophylaxis [35]. A trial that incorporated a blinded design and the use of continuous electroencephalography (cEEG) monitoring demonstrated no difference in early or late seizure incidence between drugs, but did favor levetiracetam over phenytoin in the 6-month Glasgow Outcome score measure [36].
The use of valproic acid, as compared with phenytoin, failed to demonstrate benefit and its use correlated with higher mortality [37]. However, a recent Cochrane analysis failed to discriminate between phenytoin and other anticonvulsants with regards to reduced incidence or mortality [37].

Ischemic stroke

Seizures following ischemic stroke occur within an incidence range of 4% to 14% [3840]. According to a large recent registry from France, early seizures are not associated with higher mortality or unfavorable outcomes at 1 month or at 1 year [41]. Patients with late-onset seizures have a greater than twice the risk for subsequent stroke, although the causal relationship is unclear [42].
Given the rather low incidence of seizures following most ischemic stroke syndromes, neither the American Heart Association (AHA) nor the American Stroke Association guidelines recommend prophylactic use of anticonvulsants following ischemic stroke [43]. However, some patients may be at greater risk for seizures. An increase in seizure risk has been reported following cardioembolism [39], hyperglycemia [41], intravenous thrombolysis with alteplase [44], in patient with SE following poststroke seizures, and either large cortical or hemorrhagic infarction [38, 4548].
If “prophylaxis” is considered, then the agent used should be selected for its adverse effect profile. In recent years there has been some interest in ascribing statins as a “first-line” anticonvulsant in stroke patients, although supportive data are by association only and this type of intervention is not readily practiced [49, 50].

Intracerebral hemorrhage

In contrast to ischemic stroke, intracerebral hemorrhage (ICH) portends a somewhat higher risk of seizures, ranging from two- to seven-times that from ischemic stroke [38, 47, 51, 52]. If cEEG is performed during the acute admission period, nonconvulsive epileptiform activity is commonly observed and may affect more than 20% of ICH patients [5356]. As expected, lobar or supratentorial ICHs have the highest incidence of seizures; those in the posterior fossa have almost zero [38, 54]. Surgical evacuation confers an increased incidence, and subarachnoid or subdural blood also increases the risk for seizures [57].
Prophylactic anticonvulsant treatment in ICH patients is currently not strongly advocated. cEEG is suggested by some for ICH patients with a depressed mental status out of proportion to that expected for the severity of the stroke [58]. Levetiracetam has become the most widely prescribed agent despite a lack of evidence [59], likely due to its ease of use. Uncontrolled study data have suggested that phenytoin confers worse outcomes than others (primarily against levetiracetam) [6062], but these data are not supported by other recent studies [63, 64].

Subarachnoid hemorrhage

Early and late-onset seizures are observed in patients with subarachnoid hemorrhage (SAH). Risk factors include the severity of SAH, concomitant intracerebral or intraventricular blood, and treatment with craniotomy [65]. A recent study noted a 10% incidence of seizures, with almost half (47%) occurring within the initial 24 h [66]. Interestingly, the use of prophylactic anticonvulsants did not reduce the overall risk. As seizures dramatically increase cerebral blood flow and blood pressure, rebleeding from a recently ruptured aneurysm during seizures may pose a serious risk.
Current guidelines from the AHA state that the use of anticonvulsants for a brief period is reasonable, without specific drug recommendation [67]. Beyond the immediate posthemorrhagic period, routine use of anticonvulsants is advocated by the AHA only in patients with a high risk for delayed seizures, such as prior seizure, intracerebral hematoma, intractable hypertension, infarction, or middle cerebral artery aneurysm. The Neurocritical Care Society (NCS) specifically recommended against the routine use of phenytoin for prophylaxis [68].

Brain tumors

The incidence of seizures in cerebral malignancy broadly ranges from about 10% to as high as 90%, and is correlated with tumor type (glioma and oligodendroglioma being amongst the highest risk) and also the rate of progression, with slow growing tumors tending towards higher seizure incidence [69]. Prophylactic use of AEDs in brain tumors remains controversial. There are strikingly little data in support of seizure prophylaxis for brain tumor patients [7072], and guidelines by the AAN and the European Federation of Neurosciences do not support this practice [73, 74]. Since these guidelines were written prior to the widespread use of cEEG and prior to the introduction of newer, less toxic anticonvulsants (levetiracetam, lamotrigine, lacosamide, and so forth), some have suggested that such a recommendation should be reassessed [75].
Whenever patients require adjuvant chemotherapy, it is prudent to avoid the use of potent CYP3A4 coenzyme inducers such as carbamazepine, phenytoin, and phenobarbital because of the risks of compromising concurrent chemotherapy [74]. Based on more recent evidence, more patients are being managed with levetiracetam than phenytoin or sodium valproate [71], but evidence of superior efficacy remains absent [72].

Cerebral venous and sinus thrombosis

Seizures manifest as a common presentation, occurring in 29% to 50% of patients with cerebral venous sinus thrombosis [76, 77]. Approximately a third to one half of seizures occur early—as the presenting symptom or within 2 weeks of diagnosis, [7779]. Despite the lack of controlled studies, prophylaxis appears prudent where forthcoming seizures appear highly probable. There is some evidence that early seizures are predictive of a remote epilepsy condition [80] but, in and of themselves, seizures in the context of cerebral venous sinus thrombosis do not appear to be associated with death or 6-month worse outcomes [81].

Seizure control in the ICU

The widespread use of cEEG has made it apparent that seizures are more common in the ICU patients than previously thought, with estimates for seizures and SE ranging from 19% to 27% [82, 83]. Bauer and Trinka introduced the distinction between nonconvulsive status epilepticus (NCSE) proper and comatose NCSE [84]. This distinction is both important and useful as it acknowledges that treatment as well as prognosis are dependent on the underlying epileptic syndrome in the former, yet on the underlying cause of the coma in the latter. In the ICU patient population, the electrographic seizure patterns can be less distinct and harder to identify. Thus, a body of literature on the nature of, and the best treatment approach for, the “ictal-interictal continuum” has emerged and now dominates the literature. For recent reviews, see Sivaraju and Gilmore 2016 [85] and Rodríguez et al. 2016 [86].
As a general principle, the current consensus is that treatment of even a single ICU seizure should occur in order to prevent escalation into SE. Once airway, breathing, circulation, and “dextrose” (the “ABCDs”) have been addressed, pharmacologic seizure treatment should occur immediately. Several authors suggest an algorithm that escalates from first-line to second-line treatment in the time span of 30 min [87, 88]. Once urgent first- and second-line agents are prescribed during the first 30 min, persistent SE should be considered refractory, and this should prompt the use of intravenous anesthetic therapy. SE is a neurological emergency and swift intervention is of the essence. While convulsive and nonconvulsive SE should be initially approached in the same way, once the need for intubation arises it is generally accepted that NCSE may warrant a less aggressive and more individualized approach to treatment with anesthetic agents (third-line agents) compared with convulsive SE.
Available evidence uniformly suggests that benzodiazepines remain the drugs of choice for the immediate control of seizures of any kind. The efficacy of intravenous lorazepam was demonstrated by Treiman et al. in 1998 [89]. Lorazepam was most effective in the initial treatment of convulsive SE when compared with phenobarbital and diazepam plus phenytoin, although the latter were also effective. Other routes for the administration of benzodiazepines have successfully been explored, particularly for the therapy of seizures encountered outside the ICU. Silbergleit et al. reported that intramuscular midazolam was at least as safe and effective as intravenous lorazepam for prehospital seizure cessation [90], while McIntyre et al. demonstrated the efficacy of buccal midazolam [91].
Although an open-label use study purported to suggest equipoise between lorazepam and levetiracetam in the treatment of SE [92], a randomized, double-blind, placebo-controlled trial (SAMU Keppra Trial) failed to show benefit from adding intravenous levetiracetam to clonazepam in the treatment of generalized convulsive SE [93].
To monitor for ongoing seizure activity after convulsions may have ceased, cEEG is crucial. Second-line therapy for SE should be initiated within 30 min if first-line treatment has failed. Agents used as second-line therapy include fosphenytoin, valproic acid, phenobarbital, and levetiracetam. Evidence for the superiority of one over the other is generally lacking [94]. Nonetheless, phenytoin has been the most studied drug in SE and is therefore often listed as the preferred choice over the others. Valproic acid is notable for being the only agent that has shown a trend towards superior efficacy in some studies but this has not been confirmed in a large-scale trial [95]. Levetiracetam has a favorable side-effect profile and is therefore increasingly prescribed as a second-line drug. Phenobarbital is often avoided due to its long half-life, although data support its efficacy [89]. Lacosamide has recently shown promise in small case studies [96], while brivaracetam is currently under investigation [97].
If second-line agents are ineffective, treatment is typically escalated to anesthetic agents. For patients in NCSE, however, the decision whether or not to intubate and sedate the patient will usually be made on a case-by-case basis. There is some evidence to suggest that uncontrolled NCSE will result in brain damage over time [98]. Furthermore, DeLorenzo et al. noted that persistent NCSE after convulsive status may carry a worse prognosis than other forms of nonconvulsive status and perhaps should be treated more aggressively, although the presence of NCSE may simply be a marker for more severe brain injury [99]. Agents used for continuous infusion are midazolam, propofol, and pentobarbital, with no data suggesting the superiority of one over the other [100]. Ultimately, it is important not only to treat the patient with an anesthetic agent but to also continue maintenance therapy with appropriate AEDs to allow transition to a well-tolerated, long-term antiepileptic regimen.

Drugs lowering seizure threshold

Polypharmacy is common in the ICU and can contribute to adverse drug effects such as seizures by lowering the seizure threshold or via drug-drug interactions that may reduce the blood concentration of an AED the patient may already be receiving (see the next section). Antibiotics, psychotropic agents, and analgesics have been all associated with seizures [101, 102] (Table 7).
Table 7
Selected list of medications associated with lower seizure threshold [113]
Antibiotics
Psychotropic agents
Analgesics
Neurostimulants
Miscellaneous agents
Cefepime
Bupropion
Meperidineb
Amantadine
Baclofen
Erythromycin
Haloperidol
Tramadol
Amphetamines
Flumazenil
Imipenema
Phenothiazines
 
Bromocriptine
 
Isoniazid
SSRIs
   
Levofloxacin
TCAs
   
Linezolid
    
Meropenem
    
Metronidazole
    
Penicillins
    
SSRI selective serotonin reuptake inhibitor, TCA tricyclic antidepressant
aCarbapenems have been reported to have the highest rate of seizure among all drugs
bNormeperidine, the active metabolite of meperidine, has been associated with seizures and accumulates in renal failure

Special therapeutic considerations

Extracorporeal removal of antiepileptic drugs

Antiepileptic drug removal by renal replacement therapies

The renal route of elimination, low protein binding, low volume of distribution, and molecular weight are physiochemical factors that impact the propensity for drug removal via dialysis (Table 8). Unfortunately, kinetic data on optimal dosing of AEDs in RRT are not readily available. Clearly such patients benefit from careful TDM and a multidisciplinary approach. There are extensive expert reviews available in the medical literature [103, 104].
Table 8
Factors impacting drug removal by renal replacement therapies
Drug-related factors
RRT-related factors
• Low protein binding
• Mode of dialysis (CRRT vs. IHD vs. others)
• Low volume of distribution
• RRT filter membrane (sieving coefficient)
• Predominantly renally eliminated
• Dialysis prescription (mode and flow rates)
• Low molecular weight
• Duration/frequency of renal replacement
CRRT continuous renal replacement therapy, IHD intermittent hemodialysis, RRT renal replacement therapy
Intermittent hemodialysis
The removal of most AEDs by intermittent hemodialysis (IHD) is usually well characterized in the drug’s labeled prescribing information or other references. Two common drugs that are removed by IHD and for which data recommendations exist are levetiracetam and lacosamide. In addition, while phenobarbital is predominantly hepatically metabolized, some reports suggest partial elimination by IHD prompting several experts to suggest supplemental dosing after IHD and monitoring of serum drug levels [103, 105, 106].
Continuous renal replacement therapy
There is insufficient evidence to recommend clear dosing guidelines for AEDs in continuous renal replacement therapy (CRRT), and more research is needed to appropriately guide therapy. AEDs that are eliminated via IHD will also be eliminated by CRRT, often times even more so (depending on the duration of CRRT as well as the mode and flow rates) thereby requiring higher doses. For drugs where serum levels are available, TDM is indicated. Estimations of drug removal can be performed to estimate the removal of medications by CRRT based on flow rates, as well as drug and filter properties. The reader is referred to other resources for a more comprehensive discussion on these methods [107, 108]. A recent review of the literature proposed dosing recommendations based on AED protein binding and the extent of renal versus hepatic elimination [104]. This is a reasonable approach, although clinicians should also factor in CRRT flow rates, the clinical condition of the patient, and other factors into their clinical decision making accordingly.

Antiepileptic drug removal by plasmapheresis

In the neurocritical care unit, plasmapheresis (PLEX) is often employed to manage neuroautoimmune diseases. Unlike RRT, which removes the free fraction of a drug, PLEX removes whole plasma which includes both the free fraction of a drug as well as the protein-bound portion. Thus, a key factor in determining the whole body removal of a drug with PLEX is its volume of distribution. Other factors can include the duration/frequency of PLEX and exchange volume, as well as the rate of intercompartmental equilibration. In general, drugs with low volumes of distribution (< 0.2 L/kg) predominantly reside in the vascular compartment and PLEX would be expected to remove a significant portion of the total body stores of these drugs. As for AEDs with high volumes of distribution such as phenytoin, PLEX appears to remove only about 2.5–10% of total body phenytoin [109, 110].

Antiepileptic drug removal by extracorporeal membrane oxygenation

Extracorporeal membrane oxygenation (ECMO) can impact the pharmacokinetics of drugs in a number of ways. ECMO circuits are well known to sequester drugs given the large surface area of the tubing and membranes which can result in an increase in the volume of distribution initially [111], while later releasing the drug into the circulation resulting in an unpredictable effect [112]. Lipophilic and highly protein-bound drugs are considered to be particularly vulnerable, but other physiochemical factors such as molecular weight, protein binding, and ionization may all play a role [113]. In addition, ECMO is typically associated with reduced drug clearance as a result of alterations of end-organ perfusion. ECMO is also often combined with some form of RRT, further complicating predictability. One case report suggests ECMO has little impact on the disposition of levetiracetam (which has a low volume of distribution and low protein binding) [114]. Where available, clinicians should monitor serum levels to guide dosing in patients on AEDs.

Conclusion

Selection of the most appropriate AED in the ICU setting can be challenging for a variety of reasons. Older AEDs such as phenytoin, valproic acid, and phenobarbital are often used by clinicians due to their familiarity with these agents, intravenous formulations, and availability of evidence in certain clinical scenarios. Despite this, adverse effects of these agents, drug-drug interactions, and the need for TDM may limit their use. Newer agents such as levetiracetam and lacosamide are gaining popularity due to their relatively safe AED profile, fewer drug-drug interactions, and lack of need for TDM. The efficacy of these agents for seizure prophylaxis and as second-line treatment for SE, however, should be further evaluated in large randomized clinical trials.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Varelas PN, Spanaki MV, Mirski MA. Seizures and the neurosurgical intensive care unit. Neurosurg Clin N Am. 2013;24(3):393–406.PubMedCrossRef Varelas PN, Spanaki MV, Mirski MA. Seizures and the neurosurgical intensive care unit. Neurosurg Clin N Am. 2013;24(3):393–406.PubMedCrossRef
2.
Zurück zum Zitat Voils SA, Human T, Brophy GM. Adverse neurologic effects of medications commonly used in the intensive care unit. Crit Care Clin. 2014;30(4):795–811.PubMedCrossRef Voils SA, Human T, Brophy GM. Adverse neurologic effects of medications commonly used in the intensive care unit. Crit Care Clin. 2014;30(4):795–811.PubMedCrossRef
4.
Zurück zum Zitat Boucher BA, Hanes SD. Pharmacokinetic alterations after severe head injury. Clin Pharmacokinet. 1998;35(3):209–21.PubMedCrossRef Boucher BA, Hanes SD. Pharmacokinetic alterations after severe head injury. Clin Pharmacokinet. 1998;35(3):209–21.PubMedCrossRef
5.
Zurück zum Zitat Boucher BA, et al. Phenytoin pharmacokinetics in critically ill trauma patients. Clin Pharmacol Ther. 1988;44(6):675–83.PubMedCrossRef Boucher BA, et al. Phenytoin pharmacokinetics in critically ill trauma patients. Clin Pharmacol Ther. 1988;44(6):675–83.PubMedCrossRef
6.
Zurück zum Zitat McKindley DS, et al. Effect of acute phase response on phenytoin metabolism in neurotrauma patients. J Clin Pharmacol. 1997;37(2):129–39.PubMedCrossRef McKindley DS, et al. Effect of acute phase response on phenytoin metabolism in neurotrauma patients. J Clin Pharmacol. 1997;37(2):129–39.PubMedCrossRef
7.
Zurück zum Zitat Arpino PA, Greer DM. Practical pharmacologic aspects of therapeutic hypothermia after cardiac arrest. Pharmacotherapy. 2008;28(1):102–11.PubMedCrossRef Arpino PA, Greer DM. Practical pharmacologic aspects of therapeutic hypothermia after cardiac arrest. Pharmacotherapy. 2008;28(1):102–11.PubMedCrossRef
8.
Zurück zum Zitat Bauer LA. Phenytoin/fosphenytoin. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015. Bauer LA. Phenytoin/fosphenytoin. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015.
9.
Zurück zum Zitat Spengler RF, et al. Severe soft-tissue injury following intravenous infusion of phenytoin: Patient and drug administration risk factors. Arch Intern Med. 1988;148(6):1329–33.PubMedCrossRef Spengler RF, et al. Severe soft-tissue injury following intravenous infusion of phenytoin: Patient and drug administration risk factors. Arch Intern Med. 1988;148(6):1329–33.PubMedCrossRef
10.
Zurück zum Zitat Millares-Sipin CA, Alafris A, Cohen H. Phenytoin and fosphenytoin, in casebook. In: Cohen H, editor. Clinical pharmacokinetics and drug dosing. New York: McGraw-Hill Education; 2015. Millares-Sipin CA, Alafris A, Cohen H. Phenytoin and fosphenytoin, in casebook. In: Cohen H, editor. Clinical pharmacokinetics and drug dosing. New York: McGraw-Hill Education; 2015.
11.
Zurück zum Zitat Bauer LA. Valproic acid. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015. Bauer LA. Valproic acid. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015.
12.
Zurück zum Zitat Bauer LA, et al. Valproic acid clearance: unbound fraction and diurnal variation in young and elderly adults. Clin Pharmacol Ther. 1985;37(6):697–700.PubMedCrossRef Bauer LA, et al. Valproic acid clearance: unbound fraction and diurnal variation in young and elderly adults. Clin Pharmacol Ther. 1985;37(6):697–700.PubMedCrossRef
13.
Zurück zum Zitat Mirski MA, Williams MA, Hanley DF. Prolonged pentobarbital and phenobarbital coma for refractory generalized status epilepticus. Crit Care Med. 1995;23(2):400–4.PubMedCrossRef Mirski MA, Williams MA, Hanley DF. Prolonged pentobarbital and phenobarbital coma for refractory generalized status epilepticus. Crit Care Med. 1995;23(2):400–4.PubMedCrossRef
14.
Zurück zum Zitat Rogers SJ, Cavazos JE. Chapter 40. Epilepsy. In: DiPiro JT, et al., editors. Pharmacotherapy: a pathophysiologic approach, 9e. New York: The McGraw-Hill Companies; 2014. Rogers SJ, Cavazos JE. Chapter 40. Epilepsy. In: DiPiro JT, et al., editors. Pharmacotherapy: a pathophysiologic approach, 9e. New York: The McGraw-Hill Companies; 2014.
15.
Zurück zum Zitat Bauer LA. Phenobarbital/primidone. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015. Bauer LA. Phenobarbital/primidone. In: Applied clinical pharmacokinetics, 3e. New York: McGraw-Hill Medical; 2015.
16.
Zurück zum Zitat Eisenberg HM, et al. High-dose barbiturate control of elevated intracranial pressure in patients with severe head injury. J Neurosurg. 1988;69(1):15–23.PubMedCrossRef Eisenberg HM, et al. High-dose barbiturate control of elevated intracranial pressure in patients with severe head injury. J Neurosurg. 1988;69(1):15–23.PubMedCrossRef
17.
Zurück zum Zitat Wermeling DP, et al. Pentobarbital pharmacokinetics in patients with severe head injury. Drug Intell Clin Pharm. 1987;21(5):459–63.PubMedCrossRef Wermeling DP, et al. Pentobarbital pharmacokinetics in patients with severe head injury. Drug Intell Clin Pharm. 1987;21(5):459–63.PubMedCrossRef
18.
Zurück zum Zitat Huynh F, Mabasa VH, Ensom MH. A critical review: does thiopental continuous infusion warrant therapeutic drug monitoring in the critical care population? Ther Drug Monit. 2009;31(2):153–69.PubMedCrossRef Huynh F, Mabasa VH, Ensom MH. A critical review: does thiopental continuous infusion warrant therapeutic drug monitoring in the critical care population? Ther Drug Monit. 2009;31(2):153–69.PubMedCrossRef
19.
20.
Zurück zum Zitat Kwan P, Brodie MJ. Effectiveness of first antiepileptic drug. Epilepsia. 2001;42(10):1255–60.PubMedCrossRef Kwan P, Brodie MJ. Effectiveness of first antiepileptic drug. Epilepsia. 2001;42(10):1255–60.PubMedCrossRef
21.
Zurück zum Zitat Temkin NR. Preventing and treating posttraumatic seizures: the human experience. Epilepsia. 2009;50(s2):10–3.PubMedCrossRef Temkin NR. Preventing and treating posttraumatic seizures: the human experience. Epilepsia. 2009;50(s2):10–3.PubMedCrossRef
22.
Zurück zum Zitat Herman ST. Epilepsy after brain insult targeting epileptogenesis. Neurology. 2002;59(9 suppl 5):S21–6.PubMedCrossRef Herman ST. Epilepsy after brain insult targeting epileptogenesis. Neurology. 2002;59(9 suppl 5):S21–6.PubMedCrossRef
23.
Zurück zum Zitat Pitkänen A, Bolkvadze T. Head trauma and epilepsy. Epilepsia. 2010;51(s5):31.CrossRef Pitkänen A, Bolkvadze T. Head trauma and epilepsy. Epilepsia. 2010;51(s5):31.CrossRef
25.
Zurück zum Zitat Lee S-T, et al. Early seizures after moderate closed head injury. Acta Neurochir. 1995;137(3–4):151–4.PubMedCrossRef Lee S-T, et al. Early seizures after moderate closed head injury. Acta Neurochir. 1995;137(3–4):151–4.PubMedCrossRef
26.
Zurück zum Zitat Frey LC. Epidemiology of posttraumatic epilepsy: a critical review. Epilepsia. 2003;44(s10):11–7.PubMedCrossRef Frey LC. Epidemiology of posttraumatic epilepsy: a critical review. Epilepsia. 2003;44(s10):11–7.PubMedCrossRef
27.
Zurück zum Zitat Temkin NR, et al. A randomized, double-blind study of phenytoin for the prevention of post-traumatic seizures. N Engl J Med. 1990;323(8):497–502.PubMedCrossRef Temkin NR, et al. A randomized, double-blind study of phenytoin for the prevention of post-traumatic seizures. N Engl J Med. 1990;323(8):497–502.PubMedCrossRef
28.
Zurück zum Zitat Schierhout G, Roberts I. Prophylactic antiepileptic agents after head injury: a systematic review. J Neurol Neurosurg Psychiatry. 1998;64(1):108–12.PubMedPubMedCentralCrossRef Schierhout G, Roberts I. Prophylactic antiepileptic agents after head injury: a systematic review. J Neurol Neurosurg Psychiatry. 1998;64(1):108–12.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Chang BS, Lowenstein DH. Antiepileptic drug prophylaxis in severe traumatic brain injury. Neurology. 2003;61(8):1162.CrossRef Chang BS, Lowenstein DH. Antiepileptic drug prophylaxis in severe traumatic brain injury. Neurology. 2003;61(8):1162.CrossRef
30.
Zurück zum Zitat Bratton S, et al. Guidelines for the management of severe traumatic brain injury. XIII Antiseizure prophylaxis. J Neurotrauma. 2006;24:S83–6.CrossRef Bratton S, et al. Guidelines for the management of severe traumatic brain injury. XIII Antiseizure prophylaxis. J Neurotrauma. 2006;24:S83–6.CrossRef
31.
Zurück zum Zitat Sundararajan K, et al. Anti-seizure prophylaxis in critically ill patients with traumatic brain injury in an intensive care unit. Anaesth Intensive Care. 2015;43(5):646–51.PubMed Sundararajan K, et al. Anti-seizure prophylaxis in critically ill patients with traumatic brain injury in an intensive care unit. Anaesth Intensive Care. 2015;43(5):646–51.PubMed
32.
Zurück zum Zitat Cranley MR, Craner M, McGilloway E. Antiepileptic prophylaxis following severe traumatic brain injury within a military cohort. J R Army Med Corps. 2015;162:109–14.PubMedCrossRef Cranley MR, Craner M, McGilloway E. Antiepileptic prophylaxis following severe traumatic brain injury within a military cohort. J R Army Med Corps. 2015;162:109–14.PubMedCrossRef
33.
Zurück zum Zitat Cotton BA, et al. Cost-utility analysis of levetiracetam and phenytoin for posttraumatic seizure prophylaxis. J Trauma Acute Care Surg. 2011;71(2):375–9.CrossRef Cotton BA, et al. Cost-utility analysis of levetiracetam and phenytoin for posttraumatic seizure prophylaxis. J Trauma Acute Care Surg. 2011;71(2):375–9.CrossRef
34.
35.
Zurück zum Zitat Zangbar B, et al. Levetiracetam prophylaxis for post-traumatic brain injury seizures is ineffective: a propensity score analysis. World J Surg. 2016;40(11):2667–72.PubMedCrossRef Zangbar B, et al. Levetiracetam prophylaxis for post-traumatic brain injury seizures is ineffective: a propensity score analysis. World J Surg. 2016;40(11):2667–72.PubMedCrossRef
36.
Zurück zum Zitat Szaflarski JP, et al. Prospective, randomized, single-blinded comparative trial of intravenous levetiracetam versus phenytoin for seizure prophylaxis. Neurocrit Care. 2010;12(2):165–72.PubMedCrossRef Szaflarski JP, et al. Prospective, randomized, single-blinded comparative trial of intravenous levetiracetam versus phenytoin for seizure prophylaxis. Neurocrit Care. 2010;12(2):165–72.PubMedCrossRef
37.
Zurück zum Zitat Temkin NR, et al. Valproate therapy for prevention of posttraumatic seizures: a randomized trial. J Neurosurg. 1999;91(4):593–600.PubMedCrossRef Temkin NR, et al. Valproate therapy for prevention of posttraumatic seizures: a randomized trial. J Neurosurg. 1999;91(4):593–600.PubMedCrossRef
38.
Zurück zum Zitat Bladin CF, et al. Seizures after stroke: a prospective multicenter study. Arch Neurol. 2000;57(11):1617–22.PubMedCrossRef Bladin CF, et al. Seizures after stroke: a prospective multicenter study. Arch Neurol. 2000;57(11):1617–22.PubMedCrossRef
40.
Zurück zum Zitat Bryndziar T, et al. Seizures following ischemic stroke: frequency of occurrence and impact on outcome in a long-term population-based study. J Stroke Cerebrovasc Dis. 2016;25(1):150–6.PubMedCrossRef Bryndziar T, et al. Seizures following ischemic stroke: frequency of occurrence and impact on outcome in a long-term population-based study. J Stroke Cerebrovasc Dis. 2016;25(1):150–6.PubMedCrossRef
41.
Zurück zum Zitat Hamidou B, et al. Prognostic value of early epileptic seizures on mortality and functional disability in acute stroke: the Dijon Stroke Registry (1985–2010). J Neurol. 2013;260(4):1043–51.PubMedCrossRef Hamidou B, et al. Prognostic value of early epileptic seizures on mortality and functional disability in acute stroke: the Dijon Stroke Registry (1985–2010). J Neurol. 2013;260(4):1043–51.PubMedCrossRef
42.
Zurück zum Zitat Cleary P, Shorvon S, Tallis R. Late-onset seizures as a predictor of subsequent stroke. Lancet. 2004;363(9416):1184–6.PubMedCrossRef Cleary P, Shorvon S, Tallis R. Late-onset seizures as a predictor of subsequent stroke. Lancet. 2004;363(9416):1184–6.PubMedCrossRef
43.
Zurück zum Zitat Jauch EC, et al. Guidelines for the early management of patients with acute ischemic stroke. Stroke. 2013;44(3):870–947.PubMedCrossRef Jauch EC, et al. Guidelines for the early management of patients with acute ischemic stroke. Stroke. 2013;44(3):870–947.PubMedCrossRef
44.
Zurück zum Zitat Alvarez V, et al. Acute seizures in acute ischemic stroke: does thrombolysis have a role to play? J Neurol. 2013;260(1):55–61.PubMedCrossRef Alvarez V, et al. Acute seizures in acute ischemic stroke: does thrombolysis have a role to play? J Neurol. 2013;260(1):55–61.PubMedCrossRef
45.
Zurück zum Zitat Reith J, et al. Seizures in acute stroke: predictors and prognostic significance. The Copenhagen Stroke Study. 1997;28(8):1585–9. Reith J, et al. Seizures in acute stroke: predictors and prognostic significance. The Copenhagen Stroke Study. 1997;28(8):1585–9.
46.
Zurück zum Zitat Lamy C, et al. Early and late seizures after cryptogenic ischemic stroke in young adults. Neurology. 2003;60(3):400–4.PubMedCrossRef Lamy C, et al. Early and late seizures after cryptogenic ischemic stroke in young adults. Neurology. 2003;60(3):400–4.PubMedCrossRef
47.
Zurück zum Zitat Beghi E, et al. Incidence and predictors of acute symptomatic seizures after stroke. Neurology. 2011;77(20):1785–93.PubMedCrossRef Beghi E, et al. Incidence and predictors of acute symptomatic seizures after stroke. Neurology. 2011;77(20):1785–93.PubMedCrossRef
48.
Zurück zum Zitat Rumbach L, et al. Status epilepticus in stroke: report on a hospital-based stroke cohort. Neurology. 2000;54(2):350.PubMedCrossRef Rumbach L, et al. Status epilepticus in stroke: report on a hospital-based stroke cohort. Neurology. 2000;54(2):350.PubMedCrossRef
49.
Zurück zum Zitat Guo J, et al. Statin treatment reduces the risk of poststroke seizures. Neurology. 2015;85(8):701–7.PubMedCrossRef Guo J, et al. Statin treatment reduces the risk of poststroke seizures. Neurology. 2015;85(8):701–7.PubMedCrossRef
50.
Zurück zum Zitat Siniscalchi A, Mintzer S. Statins for poststroke seizures: the first antiepileptogenic agent? Neurology. 2015;85:661–2.PubMedCrossRef Siniscalchi A, Mintzer S. Statins for poststroke seizures: the first antiepileptogenic agent? Neurology. 2015;85:661–2.PubMedCrossRef
51.
Zurück zum Zitat Lahti A-M, et al. Poststroke epilepsy in long-term survivors of primary intracerebral hemorrhage. Neurology. 2017;88(23):2169–75.PubMedCrossRef Lahti A-M, et al. Poststroke epilepsy in long-term survivors of primary intracerebral hemorrhage. Neurology. 2017;88(23):2169–75.PubMedCrossRef
52.
Zurück zum Zitat Giroud M, et al. Early seizures after acute stroke: a study of 1,640 cases. Epilepsia. 1994;35(5):959–64.PubMedCrossRef Giroud M, et al. Early seizures after acute stroke: a study of 1,640 cases. Epilepsia. 1994;35(5):959–64.PubMedCrossRef
53.
Zurück zum Zitat Vespa PM, et al. Acute seizures after intracerebral hemorrhage: a factor in progressive midline shift and outcome. Neurology. 2003;60(9):1441–6.PubMedCrossRef Vespa PM, et al. Acute seizures after intracerebral hemorrhage: a factor in progressive midline shift and outcome. Neurology. 2003;60(9):1441–6.PubMedCrossRef
54.
Zurück zum Zitat Faught E, et al. Seizures after primary intracerebral hemorrhage. Neurology. 1989;39(8):1089.PubMedCrossRef Faught E, et al. Seizures after primary intracerebral hemorrhage. Neurology. 1989;39(8):1089.PubMedCrossRef
55.
Zurück zum Zitat Tatu L, et al. Primary intracerebral hemorrhages in the Besancon stroke registry. Eur Neurol. 2000;43(4):209–14.PubMedCrossRef Tatu L, et al. Primary intracerebral hemorrhages in the Besancon stroke registry. Eur Neurol. 2000;43(4):209–14.PubMedCrossRef
56.
Zurück zum Zitat Claassen J, et al. Electrographic seizures and periodic discharges after intracerebral hemorrhage. Neurology. 2007;69(13):1356–65.PubMedCrossRef Claassen J, et al. Electrographic seizures and periodic discharges after intracerebral hemorrhage. Neurology. 2007;69(13):1356–65.PubMedCrossRef
57.
Zurück zum Zitat Garrett MC, et al. Predictors of seizure onset after intracerebral hemorrhage and the role of long-term antiepileptic therapy. J Crit Care. 2009;24(3):335–9.PubMedCrossRef Garrett MC, et al. Predictors of seizure onset after intracerebral hemorrhage and the role of long-term antiepileptic therapy. J Crit Care. 2009;24(3):335–9.PubMedCrossRef
58.
Zurück zum Zitat Morgenstern LB, et al. Guidelines for the management of spontaneous intracerebral hemorrhage. A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2010;41:2108–29.PubMedPubMedCentralCrossRef Morgenstern LB, et al. Guidelines for the management of spontaneous intracerebral hemorrhage. A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2010;41:2108–29.PubMedPubMedCentralCrossRef
59.
60.
Zurück zum Zitat Naidech AM, et al. Anticonvulsant use and outcomes after intracerebral hemorrhage. Stroke. 2009;40(12):3810–5.PubMedCrossRef Naidech AM, et al. Anticonvulsant use and outcomes after intracerebral hemorrhage. Stroke. 2009;40(12):3810–5.PubMedCrossRef
61.
Zurück zum Zitat Messé SR, et al. Prophylactic antiepileptic drug use is associated with poor outcome following ICH. Neurocrit Care. 2009;11(1):38–44.PubMedCrossRef Messé SR, et al. Prophylactic antiepileptic drug use is associated with poor outcome following ICH. Neurocrit Care. 2009;11(1):38–44.PubMedCrossRef
62.
Zurück zum Zitat Gilmore EJ, et al. Review of the utility of prophylactic anticonvulsant use in critically ill patients with intracerebral hemorrhage. Stroke. 2016;47(10):2666–72.PubMedCrossRef Gilmore EJ, et al. Review of the utility of prophylactic anticonvulsant use in critically ill patients with intracerebral hemorrhage. Stroke. 2016;47(10):2666–72.PubMedCrossRef
63.
Zurück zum Zitat Sheth KN, et al. Prophylactic antiepileptic drug use and outcome in the ethnic/racial variations of intracerebral hemorrhage study. Stroke. 2015;46(12):3532–5.PubMedPubMedCentralCrossRef Sheth KN, et al. Prophylactic antiepileptic drug use and outcome in the ethnic/racial variations of intracerebral hemorrhage study. Stroke. 2015;46(12):3532–5.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Zandieh A, et al. Prophylactic use of antiepileptic drugs in patients with spontaneous intracerebral hemorrhage. J Stroke Cerebrovasc Dis. 2016;25(9):2159–66.PubMedPubMedCentralCrossRef Zandieh A, et al. Prophylactic use of antiepileptic drugs in patients with spontaneous intracerebral hemorrhage. J Stroke Cerebrovasc Dis. 2016;25(9):2159–66.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Maciel CB, Gilmore EJ. Seizures and epileptiform patterns in SAH and their relation to outcomes. J Clin Neurophysiol. 2016;33(3):183–95.PubMedCrossRef Maciel CB, Gilmore EJ. Seizures and epileptiform patterns in SAH and their relation to outcomes. J Clin Neurophysiol. 2016;33(3):183–95.PubMedCrossRef
66.
67.
Zurück zum Zitat Connolly ES, et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage. Stroke. 2012;43:1711–37.PubMedCrossRef Connolly ES, et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage. Stroke. 2012;43:1711–37.PubMedCrossRef
68.
Zurück zum Zitat Diringer MN, et al. Critical care management of patients following aneurysmal subarachnoid hemorrhage: recommendations from the Neurocritical Care Society’s Multidisciplinary Consensus Conference. Neurocrit Care. 2011;15(2):211.PubMedCrossRef Diringer MN, et al. Critical care management of patients following aneurysmal subarachnoid hemorrhage: recommendations from the Neurocritical Care Society’s Multidisciplinary Consensus Conference. Neurocrit Care. 2011;15(2):211.PubMedCrossRef
69.
Zurück zum Zitat Bauer R, et al. Treatment of epileptic seizures in brain tumors: a critical review. Neurosurg Rev. 2014;37(3):381–8.PubMedCrossRef Bauer R, et al. Treatment of epileptic seizures in brain tumors: a critical review. Neurosurg Rev. 2014;37(3):381–8.PubMedCrossRef
70.
Zurück zum Zitat Garbossa D, et al. A retrospective two-center study of antiepileptic prophylaxis in patients with surgically treated high-grade gliomas. Neurol India. 2013;61(2):131.PubMedCrossRef Garbossa D, et al. A retrospective two-center study of antiepileptic prophylaxis in patients with surgically treated high-grade gliomas. Neurol India. 2013;61(2):131.PubMedCrossRef
71.
Zurück zum Zitat Kruer RM, et al. Changing trends in the use of seizure prophylaxis after traumatic brain injury: a shift from phenytoin to levetiracetam. J Crit Care. 2013;28(5):883. e9-883. e13PubMedCrossRef Kruer RM, et al. Changing trends in the use of seizure prophylaxis after traumatic brain injury: a shift from phenytoin to levetiracetam. J Crit Care. 2013;28(5):883. e9-883. e13PubMedCrossRef
72.
73.
Zurück zum Zitat Glantz M, et al. Practice parameter: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000;54(10):1886–93.PubMedCrossRef Glantz M, et al. Practice parameter: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000;54(10):1886–93.PubMedCrossRef
74.
Zurück zum Zitat Soffietti R, et al. Guidelines on management of low-grade gliomas: report of an EFNS–EANO Task Force. Eur J Neurol. 2010;17(9):1124–33.PubMedCrossRef Soffietti R, et al. Guidelines on management of low-grade gliomas: report of an EFNS–EANO Task Force. Eur J Neurol. 2010;17(9):1124–33.PubMedCrossRef
75.
Zurück zum Zitat Wychowski T, et al. Considerations in prophylaxis for tumor-associated epilepsy: prevention of status epilepticus and tolerability of newer generation AEDs. Clin Neurol Neurosurg. 2013;115(11):2365–9.PubMedCrossRef Wychowski T, et al. Considerations in prophylaxis for tumor-associated epilepsy: prevention of status epilepticus and tolerability of newer generation AEDs. Clin Neurol Neurosurg. 2013;115(11):2365–9.PubMedCrossRef
76.
Zurück zum Zitat Masuhr F, et al. Risk and predictors of early epileptic seizures in acute cerebral venous and sinus thrombosis. Eur J Neurol. 2006;13(8):852–6.PubMedCrossRef Masuhr F, et al. Risk and predictors of early epileptic seizures in acute cerebral venous and sinus thrombosis. Eur J Neurol. 2006;13(8):852–6.PubMedCrossRef
77.
Zurück zum Zitat Ferro J, et al. Seizures in cerebral vein and dural sinus thrombosis. Cerebrovasc Dis. 2003;15:78–83.PubMedCrossRef Ferro J, et al. Seizures in cerebral vein and dural sinus thrombosis. Cerebrovasc Dis. 2003;15:78–83.PubMedCrossRef
78.
Zurück zum Zitat Ferro JM, et al. Early seizures in cerebral vein and dural sinus thrombosis. Stroke. 2008;39(4):1152–8.PubMedCrossRef Ferro JM, et al. Early seizures in cerebral vein and dural sinus thrombosis. Stroke. 2008;39(4):1152–8.PubMedCrossRef
79.
Zurück zum Zitat Mahale R, et al. Acute seizures in cerebral venous sinus thrombosis: what predicts it? Epilepsy Res. 2016;123:1–5.PubMedCrossRef Mahale R, et al. Acute seizures in cerebral venous sinus thrombosis: what predicts it? Epilepsy Res. 2016;123:1–5.PubMedCrossRef
80.
Zurück zum Zitat Davoudi V, Saadatnia M. Risk factors for remote seizure development in patients with cerebral vein and dural sinus thrombosis. Seizure. 2014;23(2):135–9.PubMedCrossRef Davoudi V, Saadatnia M. Risk factors for remote seizure development in patients with cerebral vein and dural sinus thrombosis. Seizure. 2014;23(2):135–9.PubMedCrossRef
81.
Zurück zum Zitat Kalita J, Chandra S, Misra UK. Significance of seizure in cerebral venous sinus thrombosis. Seizure. 2012;21(8):639–42.PubMedCrossRef Kalita J, Chandra S, Misra UK. Significance of seizure in cerebral venous sinus thrombosis. Seizure. 2012;21(8):639–42.PubMedCrossRef
82.
Zurück zum Zitat Claassen J, et al. Detection of electrographic seizures with continuous EEG monitoring in critically ill patients. Neurology. 2004;62(10):1743–8.PubMedCrossRef Claassen J, et al. Detection of electrographic seizures with continuous EEG monitoring in critically ill patients. Neurology. 2004;62(10):1743–8.PubMedCrossRef
83.
Zurück zum Zitat Westover MB, et al. The probability of seizures during EEG monitoring in critically ill adults. Clin Neurophysiol. 2015;126(3):463–71.PubMedCrossRef Westover MB, et al. The probability of seizures during EEG monitoring in critically ill adults. Clin Neurophysiol. 2015;126(3):463–71.PubMedCrossRef
84.
Zurück zum Zitat Bauer G, Trinka E. Nonconvulsive status epilepticus and coma. Epilepsia. 2010;51(2):177–90.PubMedCrossRef Bauer G, Trinka E. Nonconvulsive status epilepticus and coma. Epilepsia. 2010;51(2):177–90.PubMedCrossRef
85.
Zurück zum Zitat Sivaraju A, Gilmore EJ. Understanding and managing the ictal-interictal continuum in neurocritical care. Curr Treat Options Neurol. 2016;18(2):8.PubMedCrossRef Sivaraju A, Gilmore EJ. Understanding and managing the ictal-interictal continuum in neurocritical care. Curr Treat Options Neurol. 2016;18(2):8.PubMedCrossRef
86.
Zurück zum Zitat Rodríguez V, Rodden MF, LaRoche SM. Ictal–interictal continuum: a proposed treatment algorithm. Clin Neurophysiol. 2016;127(4):2056–64.PubMedCrossRef Rodríguez V, Rodden MF, LaRoche SM. Ictal–interictal continuum: a proposed treatment algorithm. Clin Neurophysiol. 2016;127(4):2056–64.PubMedCrossRef
87.
Zurück zum Zitat Costello DJ, Cole AJ. Treatment of acute seizures and status epilepticus. J Intensive Care Med. 2007;22(6):319–47.PubMedCrossRef Costello DJ, Cole AJ. Treatment of acute seizures and status epilepticus. J Intensive Care Med. 2007;22(6):319–47.PubMedCrossRef
88.
Zurück zum Zitat Billington M, Kandalaft OR, Aisiku IP. Adult status epilepticus: a review of the prehospital and emergency department management. J Clin Med. 2016;5(9):74.PubMedCentralCrossRef Billington M, Kandalaft OR, Aisiku IP. Adult status epilepticus: a review of the prehospital and emergency department management. J Clin Med. 2016;5(9):74.PubMedCentralCrossRef
89.
Zurück zum Zitat Treiman DM, et al. A comparison of four treatments for generalized convulsive status epilepticus. N Engl J Med. 1998;339(12):792–8.PubMedCrossRef Treiman DM, et al. A comparison of four treatments for generalized convulsive status epilepticus. N Engl J Med. 1998;339(12):792–8.PubMedCrossRef
90.
91.
Zurück zum Zitat McIntyre J, et al. Safety and efficacy of buccal midazolam versus rectal diazepam for emergency treatment of seizures in children: a randomised controlled trial. Lancet. 2005;366(9481):205–10.PubMedCrossRef McIntyre J, et al. Safety and efficacy of buccal midazolam versus rectal diazepam for emergency treatment of seizures in children: a randomised controlled trial. Lancet. 2005;366(9481):205–10.PubMedCrossRef
92.
Zurück zum Zitat Misra U, Kalita J, Maurya P. Levetiracetam versus lorazepam in status epilepticus: a randomized, open labeled pilot study. J Neurol. 2012;259(4):645–8.PubMedCrossRef Misra U, Kalita J, Maurya P. Levetiracetam versus lorazepam in status epilepticus: a randomized, open labeled pilot study. J Neurol. 2012;259(4):645–8.PubMedCrossRef
93.
Zurück zum Zitat Navarro V, et al. Prehospital treatment with levetiracetam plus clonazepam or placebo plus clonazepam in status epilepticus (SAMUKeppra): a randomised, double-blind, phase 3 trial. Lancet Neurol. 2016;15(1):47–55.PubMedCrossRef Navarro V, et al. Prehospital treatment with levetiracetam plus clonazepam or placebo plus clonazepam in status epilepticus (SAMUKeppra): a randomised, double-blind, phase 3 trial. Lancet Neurol. 2016;15(1):47–55.PubMedCrossRef
94.
Zurück zum Zitat Brophy G, et al. Neurocritical Care Society Status Epilepticus Guideline Neurocrit Care Writing Committee. Guidelines for the evaluation and management of status epilepticus. Neurocrit Care. 2012;17:3–23.PubMedCrossRef Brophy G, et al. Neurocritical Care Society Status Epilepticus Guideline Neurocrit Care Writing Committee. Guidelines for the evaluation and management of status epilepticus. Neurocrit Care. 2012;17:3–23.PubMedCrossRef
95.
Zurück zum Zitat Misra U, Kalita J, Patel R. Sodium valproate vs phenytoin in status epilepticus: a pilot study. Neurology. 2006;67(2):340–2.PubMedCrossRef Misra U, Kalita J, Patel R. Sodium valproate vs phenytoin in status epilepticus: a pilot study. Neurology. 2006;67(2):340–2.PubMedCrossRef
96.
Zurück zum Zitat Höfler J, Trinka E. Lacosamide as a new treatment option in status epilepticus. Epilepsia. 2013;54(3):393–404.PubMedCrossRef Höfler J, Trinka E. Lacosamide as a new treatment option in status epilepticus. Epilepsia. 2013;54(3):393–404.PubMedCrossRef
97.
Zurück zum Zitat Strzelczyk A, et al. Treatment of refractory and super-refractory status epilepticus with brivaracetam: A cohort study from two German university hospitals. Epilepsy Behav. 2017;70:177–81.PubMedCrossRef Strzelczyk A, et al. Treatment of refractory and super-refractory status epilepticus with brivaracetam: A cohort study from two German university hospitals. Epilepsy Behav. 2017;70:177–81.PubMedCrossRef
98.
Zurück zum Zitat Hirsch LJ, Gaspard N. Status epilepticus CONTINUUM: lifelong learning in neurology. Continuum. 2013;19(3, Epilepsy):767–94. Hirsch LJ, Gaspard N. Status epilepticus CONTINUUM: lifelong learning in neurology. Continuum. 2013;19(3, Epilepsy):767–94.
99.
Zurück zum Zitat DeLorenzo R, et al. Persistent nonconvulsive status epilepticus after the control of convulsive status epilepticus. Epilepsia. 1998;39(8):833–40.PubMedCrossRef DeLorenzo R, et al. Persistent nonconvulsive status epilepticus after the control of convulsive status epilepticus. Epilepsia. 1998;39(8):833–40.PubMedCrossRef
100.
Zurück zum Zitat Rossetti AO, Lowenstein DH. Management of refractory status epilepticus in adults: still more questions than answers. Lancet Neurol. 2011;10(10):922–30.PubMedPubMedCentralCrossRef Rossetti AO, Lowenstein DH. Management of refractory status epilepticus in adults: still more questions than answers. Lancet Neurol. 2011;10(10):922–30.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Miller AD, et al. Epileptogenic potential of carbapenem agents: mechanism of action, seizure rates, and clinical considerations. Pharmacotherapy. 2011;31(4):408–23.PubMedCrossRef Miller AD, et al. Epileptogenic potential of carbapenem agents: mechanism of action, seizure rates, and clinical considerations. Pharmacotherapy. 2011;31(4):408–23.PubMedCrossRef
102.
Zurück zum Zitat Pisani F, et al. Effects of psychotropic drugs on seizure threshold. Drug Saf. 2002;25(2):91–110.PubMedCrossRef Pisani F, et al. Effects of psychotropic drugs on seizure threshold. Drug Saf. 2002;25(2):91–110.PubMedCrossRef
103.
Zurück zum Zitat Asconape JJ. Use of antiepileptic drugs in hepatic and renal disease. In: Miller J, Ferro JM, editors. Handbook of Clinical Neurology. 2014;119:417–32.PubMedCrossRef Asconape JJ. Use of antiepileptic drugs in hepatic and renal disease. In: Miller J, Ferro JM, editors. Handbook of Clinical Neurology. 2014;119:417–32.PubMedCrossRef
104.
Zurück zum Zitat Mahmoud SH. Antiepileptic drug removal by continuous renal replacement therapy: a review of the literature. Clin Drug Investig. 2017;37(1):7–23.PubMedCrossRef Mahmoud SH. Antiepileptic drug removal by continuous renal replacement therapy: a review of the literature. Clin Drug Investig. 2017;37(1):7–23.PubMedCrossRef
105.
Zurück zum Zitat Porto I, John EG, Heilliczer J. Removal of phenobarbital during continuous cycling peritoneal dialysis in a child. Pharmacotherapy. 1997;17(4):832–5.PubMed Porto I, John EG, Heilliczer J. Removal of phenobarbital during continuous cycling peritoneal dialysis in a child. Pharmacotherapy. 1997;17(4):832–5.PubMed
106.
Zurück zum Zitat Palmer BF. Effectiveness of hemodialysis in the extracorporeal therapy of phenobarbital overdose. Am J Kidney Dis. 2000;36(3):640–3.PubMedCrossRef Palmer BF. Effectiveness of hemodialysis in the extracorporeal therapy of phenobarbital overdose. Am J Kidney Dis. 2000;36(3):640–3.PubMedCrossRef
107.
Zurück zum Zitat Schetz M, et al. Pharmacokinetics of continuous renal replacement therapy. Intensive Care Med. 1995;21(7):612–20.PubMedCrossRef Schetz M, et al. Pharmacokinetics of continuous renal replacement therapy. Intensive Care Med. 1995;21(7):612–20.PubMedCrossRef
108.
Zurück zum Zitat Reetze-Bonorden P, Bohler J, Keller E. Drug dosage in patients during continuous renal replacement therapy. Pharmacokinetic and therapeutic considerations. Clin Pharmacokinet. 1993;24(5):362–79.PubMedCrossRef Reetze-Bonorden P, Bohler J, Keller E. Drug dosage in patients during continuous renal replacement therapy. Pharmacokinetic and therapeutic considerations. Clin Pharmacokinet. 1993;24(5):362–79.PubMedCrossRef
109.
Zurück zum Zitat Silberstein LE, Shaw LM. Effect of plasma exchange on phenytoin plasma concentration. Ther Drug Monit. 1986;8(2):172–6.PubMedCrossRef Silberstein LE, Shaw LM. Effect of plasma exchange on phenytoin plasma concentration. Ther Drug Monit. 1986;8(2):172–6.PubMedCrossRef
110.
Zurück zum Zitat Ibrahim RB, et al. Drug removal by plasmapheresis: an evidence-based review. Pharmacotherapy. 2007;27(11):1529–49.PubMedCrossRef Ibrahim RB, et al. Drug removal by plasmapheresis: an evidence-based review. Pharmacotherapy. 2007;27(11):1529–49.PubMedCrossRef
111.
Zurück zum Zitat Elliott ES, Buck ML. Phenobarbital dosing and pharmacokinetics in a neonate receiving extracorporeal membrane oxygenation. Ann Pharmacother. 1999;33(4):419–22.PubMedCrossRef Elliott ES, Buck ML. Phenobarbital dosing and pharmacokinetics in a neonate receiving extracorporeal membrane oxygenation. Ann Pharmacother. 1999;33(4):419–22.PubMedCrossRef
112.
Zurück zum Zitat Shekar K, et al. Pharmacokinetic changes in patients receiving extracorporeal membrane oxygenation. J Crit Care. 2012;27(6):741. e9–18PubMedCrossRef Shekar K, et al. Pharmacokinetic changes in patients receiving extracorporeal membrane oxygenation. J Crit Care. 2012;27(6):741. e9–18PubMedCrossRef
113.
Zurück zum Zitat Shekar K, et al. Protein-bound drugs are prone to sequestration in the extracorporeal membrane oxygenation circuit: results from an ex vivo study. Crit Care. 2015;19:164.PubMedPubMedCentralCrossRef Shekar K, et al. Protein-bound drugs are prone to sequestration in the extracorporeal membrane oxygenation circuit: results from an ex vivo study. Crit Care. 2015;19:164.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Nei SD, et al. Levetiracetam pharmacokinetics in a patient receiving continuous venovenous hemofiltration and venoarterial extracorporeal membrane oxygenation. Pharmacotherapy. 2015;35(8):e127–30.PubMedCrossRef Nei SD, et al. Levetiracetam pharmacokinetics in a patient receiving continuous venovenous hemofiltration and venoarterial extracorporeal membrane oxygenation. Pharmacotherapy. 2015;35(8):e127–30.PubMedCrossRef
115.
Zurück zum Zitat Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs)—part 2: pharmacokinetic and pharmacodynamic interactions between AEDs and drugs used to treat non-epilepsy disorders. Clinical pharmacokinetics. 2013;52(12):1045–1061. Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs)—part 2: pharmacokinetic and pharmacodynamic interactions between AEDs and drugs used to treat non-epilepsy disorders. Clinical pharmacokinetics. 2013;52(12):1045–1061.
116.
Zurück zum Zitat Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: general features and interactions between antiepileptic drugs. The Lancet Neurology. 2003;2(6):347–356. Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: general features and interactions between antiepileptic drugs. The Lancet Neurology. 2003;2(6):347–356.
117.
Zurück zum Zitat Sancho J, Serratosa J. Antiepileptic drug interactions. The neurologist. 2008;14(6):S55–S65. Sancho J, Serratosa J. Antiepileptic drug interactions. The neurologist. 2008;14(6):S55–S65.
118.
Zurück zum Zitat Schmidt D, Schachter SC. Drug treatment of epilepsy in adults. Bmj. 2014;348:g254. Schmidt D, Schachter SC. Drug treatment of epilepsy in adults. Bmj. 2014;348:g254.
119.
Zurück zum Zitat Perucca E. Clinically relevant drug interactions with antiepileptic drugs. British journal of clinical pharmacology. 2006;61(3):246–255. Perucca E. Clinically relevant drug interactions with antiepileptic drugs. British journal of clinical pharmacology. 2006;61(3):246–255.
120.
Zurück zum Zitat French J, et al. Efficacy and tolerability of the new antiepileptic drugs I: Treatment of new onset epilepsy Report of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62(8):1252–1260. French J, et al. Efficacy and tolerability of the new antiepileptic drugs I: Treatment of new onset epilepsy Report of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62(8):1252–1260.
121.
Zurück zum Zitat Food U and D Administration. VIMPAT®(lacosamide) Prescribing Information. Accessed July, 2017. Food U and D Administration. VIMPAT®(lacosamide) Prescribing Information. Accessed July, 2017.
122.
Zurück zum Zitat Ingelheim B. Pradaxa (dabigatran) package insert. Ridgefield, CT. 2012. Ingelheim B. Pradaxa (dabigatran) package insert. Ridgefield, CT. 2012.
123.
Zurück zum Zitat Healthcare B. Xarelto (rivaroxaban) package insert. 2012. Healthcare B. Xarelto (rivaroxaban) package insert. 2012.
124.
Zurück zum Zitat Squibb B.-M. Eliquis (apixaban) package insert. 2015. Squibb B.-M. Eliquis (apixaban) package insert. 2015.
125.
Zurück zum Zitat Nappi JM. Warfarin and phenytoin interaction. Ann Intern Med. 1979;90(5):852. Nappi JM. Warfarin and phenytoin interaction. Ann Intern Med. 1979;90(5):852.
126.
Zurück zum Zitat Levine M, Sheppard I. Biphasic interaction of phenytoin with warfarin. Clin Pharm. 1984;3(2):200–3. Levine M, Sheppard I. Biphasic interaction of phenytoin with warfarin. Clin Pharm. 1984;3(2):200–3.
127.
Zurück zum Zitat Food U and D. Administration, Cordarone (Amiodarone HCL) Tablets: Package Insert. US Food and Drug Administration, Maryland. 2010. Food U and D. Administration, Cordarone (Amiodarone HCL) Tablets: Package Insert. US Food and Drug Administration, Maryland. 2010.
128.
Zurück zum Zitat Macphee GA, et al. Verapamil potentiates carbamazepine neurotoxicity: a clinically important inhibitory interaction. The Lancet. 1986;327(8483):700–703. Macphee GA, et al. Verapamil potentiates carbamazepine neurotoxicity: a clinically important inhibitory interaction. The Lancet. 1986;327(8483):700–703.
129.
Zurück zum Zitat Maoz E, et al. Carbamazepine neurotoxic reaction after administration of diltiazem. Archives of internal medicine. 1992;152(12):2503–2504. Maoz E, et al. Carbamazepine neurotoxic reaction after administration of diltiazem. Archives of internal medicine. 1992;152(12):2503–2504.
130.
Zurück zum Zitat Bahls FH, Ozuna J, Ritchie DE. Interactions between calcium channel blockers and the anticonvulsants carbamazepine and phenytoin. Neurology. 1991;41(5):740–742. Bahls FH, Ozuna J, Ritchie DE. Interactions between calcium channel blockers and the anticonvulsants carbamazepine and phenytoin. Neurology. 1991;41(5):740–742.
131.
Zurück zum Zitat I Johannessen, S. and C. Johannessen Landmark, Antiepileptic drug interactions-principles and clinical implications. Current neuropharmacology. 2010;8(3):254–267. I Johannessen, S. and C. Johannessen Landmark, Antiepileptic drug interactions-principles and clinical implications. Current neuropharmacology. 2010;8(3):254–267.
132.
Zurück zum Zitat Park MK, et al. Reduced valproic acid serum concentrations due to drug interactions with carbapenem antibiotics: overview of 6 cases. Therapeutic drug monitoring. 2012;34(5):599–603. Park MK, et al. Reduced valproic acid serum concentrations due to drug interactions with carbapenem antibiotics: overview of 6 cases. Therapeutic drug monitoring. 2012;34(5):599–603.
Metadaten
Titel
Antiepileptic drugs in critically ill patients
verfasst von
Salia Farrokh
Pouya Tahsili-Fahadan
Eva K. Ritzl
John J. Lewin III
Marek A. Mirski
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2018
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-018-2066-1

Weitere Artikel der Ausgabe 1/2018

Critical Care 1/2018 Zur Ausgabe

Update AINS

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