Critical Care Considerations



Fig. 1
cEEG demonstrating NCSE only detected with depth electrodes (D leads) in a 59-year-old woman with refractory status epilepticus despite pentobarbital, midazolam, levetiracetam, valproate, and phenytoin. Note the relative lack of activity at the surface electrodes



A328697_1_En_24_Fig2_HTML.gif


Fig. 2
cEEG from the same patient as in Fig. 1 demonstrating NCSE on scalp electrodes with depth electrode correlate (D leads)


A328697_1_En_24_Fig3_HTML.gif


Fig. 3
cEEG demonstrating NCSE only detected with depth electrode (D leads in blue) in a 64-year-old man with nontraumatic SAH. Note the relative lack of activity at the surface electrodes


A328697_1_En_24_Fig4_HTML.gif


Fig. 4
cEEG demonstrating electrographic seizure captured only with depth electrode in a 74-year-old woman with nontraumatic SAH (D leads). Note the relative lack of activity at the surface electrodes


A328697_1_En_24_Fig5_HTML.gif


Fig. 5
cEEG in a 66-year-old woman with a right frontal inflammatory lesion, demonstrating NCSE in the right frontal depth electrode (D leads, blue), with only a subtle correlate on the scalp electrodes


A328697_1_En_24_Fig6_HTML.gif


Fig. 6
cEEG from the same patient as in Fig. 5, expanded view showing electrographic seizures on the depth electrode (D leads, blue) with subtle scalp correlate





Neurocritical Care ICU Considerations



Status Epilepticus in the Intensive Care Unit


Status epilepticus is common in all types of acute brain injury and is not restricted to patients with a previous history of epilepsy or those admitted for seizures. It is estimated that approximately 150,000 cases of generalized convulsive status epilepticus (GCSE) occur annually in the United States [4]. Most seizures occurring in the ICU setting are nonconvulsive and will remain undetectable unless EEG monitoring is employed. The astute clinician may notice some subtle signs that may raise the suspicion for NCSz such as face and limb myoclonus, nystagmus, eye deviation, pupillary abnormalities, and autonomic instability, while many patients will have purely electrographic seizures without any overt signs [3, 5]. cEEG is therefore necessary to make the diagnosis of NCSE. As more ICUs employ cEEG, the epidemiology of seizures is becoming better understood. Generalized seizures complicate about 8 % of general ICU admissions with another 10 % having electrographic seizures. NCSz are seen in 48 % and NCSE in 14 % following clinical control of GCSE with benzodiazepines. In the neurocritical ICU setting, up to 34 % will have NCSz, and up to 76 % of those will go into NCSE [6, 7]. The subset of patients undergoing hypothermic protocols for coma after cardiac arrest have a seizure frequency of 20–30 % (excluding clinical myoclonus), most of which are NCSE [4, 8].

The recognition of NCSE has increased exponentially in the past 40 years [9]. The underlying etiologies for GCSE and NCSE are usually similar and include structural lesions, infections, metabolic derangements, toxins, withdrawal, intake of psychotropic drugs, and epilepsy, all of which are commonly encountered in the ICU setting either on their own or in conjunction with other medical problems.


Diagnostic Considerations


A single self-limited seizure in the ICU should prompt a diagnostic workup although it may not always require anticonvulsant therapy to prevent recurrence. For example, drug withdrawal, intoxication, and electrolyte disturbances are known causes of seizures for which the treatment is to address the primary underlying etiology. Renal failure, hepatic failure, as well as CNS infections are other common causes of seizures in the ICU which may or may not require anticonvulsant therapy. Toxicity from beta-lactam antibiotics is a commonly overlooked cause of seizures, especially in patients with renal failure. Hyposmolarity has the potential to exacerbate conditions which may cause seizures but should only be accepted as the sole cause of seizures when it develops acutely (over the course of hours), in which case it usually also produces intracranial hypertension. Chronic hyposmolarity causes weakness, fatigue, and confusion, but not usually seizures unless concomitant conditions are present [10].

Hypo- and hyperglycemia, hyponatremia, hypocalcemia, uremia, liver dysfunction, hypertensive encephalopathy, and sepsis have all been associated with NCSE; the incidence of which has been shown to vary from 5 to 22 %. Acute renal failure and sepsis have especially been linked to increased electrographic seizures [10]. Certain periodic discharges, such as those with triphasic morphology, are more closely related to underlying systemic metabolic abnormalities (e.g., triphasic waves in hepatic encephalopathy) and are not considered to be epileptiform, while the significance of lateralized periodic discharges (LPDs, formerly known as PLEDs or periodic lateralized epileptiform discharges) remains controversial. At times a benzodiazepine trial may be warranted to attempt to differentiate ictal from non-ictal EEG patterns in critically ill patients (Fig. 7). In a benzodiazepine trial, a bolus of a fast-acting benzodiazepine is administered to a comatose patient with an EEG pattern suspicious for NCSE. However, almost all periodic discharges, including those with triphasic morphology, are attenuated by benzodiazepines; therefore, a benzodiazepine trial is nondiagnostic unless accompanied by a clinical improvement.

A328697_1_En_24_Fig7_HTML.gif


Fig. 7
cEEG demonstrating NCSE before and after benzodiazepine trial (red arrow) in a 59-year-old liver transplant patient with sepsis. The improvement in the EEG background was associated with a dramatic clinical improvement


Seizure Prophylaxis in the Neurocritical ICU


Prophylactic antiepileptic treatment should be started in the ICU patient that has had one provoked or unprovoked seizure if even one more seizure would adversely affect the patient’s condition. For example, the acute hypertension that accompanies most generalized convulsions could prove detrimental for a patient suffering from raised intracranial pressure, and therefore antiepileptic drug (AED) treatment should be initiated after the first seizure. In other patients, it may be wise to hold off on initiating AED treatment after the first seizure when taking into consideration possible side effects, drug interactions, and sedative effects of those medications.

Most clinicians will choose to start an AED after the second seizure. In recent years, there has been a proliferation of IV AEDs available to the intensivists, so that options other than the traditional fosphenytoin can be considered, including IV valproate, levetiracetam, and lacosamide. Benefits of choosing phenytoin and valproate include monitoring serum concentrations, as they are readily available. However, ICU patients often have very low serum albumin concentrations and require therapies that compete for protein-binding sites, so unbound (free) concentrations may be needed to guide therapy [11]. Concomitant conditions must be taken into consideration. Valproate should be avoided in patients with liver failure, for example. Levetiracetam and lacosamide have fewer interactions than phenytoin and valproate with other hepatically metabolized medications but must be carefully dosed in patients with renal failure.


Management of Status Epilepticus


The emergent and potentially fatal nature of status epilepticus makes the initiation of treatment to terminate the seizures mandatory before the clinician can investigate their etiology. First and foremost, patients presenting in GCSE need attention to the basics of life support. While the best way to manage airway problems in GCSE is to terminate the seizures pharmacologically, often endotracheal intubation will be required due to the sedating nature of the pharmacological agents used to terminate the seizures [11]. In these cases, the drugs typically used for sedation – such as propofol and etomidate – will often terminate seizures briefly. If neuromuscular junction (NMJ) blockade is used, the sedative and anticonvulsant effect will usually wear off prior to the NMJ blockade, and therefore, patients may go back to having seizures while still paralyzed and without any overt signs except perhaps pupillary dilation. These patients should be treated as if they are still in GCSE and placed on cEEG to detect ongoing seizure activity [11, 12].

Most patients will be hypertensive in the first 30 to 60 min of GCSE; however antihypertensives are not recommended as almost all of the parenteral anticonvulsants (except ketamine) will lower blood pressure. Sedative drugs used for intubation (except etomidate) and positive-pressure ventilation will decrease preload and further cause hypotension as well. Conversely, if the patient is found with low blood pressure on presentation, this is suggestive of GCSE or NCSE ongoing for more than 60 min, unless a concomitant condition is causing hypotension. Saline resuscitation or vasopressor support should be considered at this point [11, 12].

NCSE in the ICU setting is associated with high morbidity and mortality, although experimental models and pathologic studies showing neuronal damage from status epilepticus were performed on convulsing patients. No randomized controlled study has conclusively proven that treating NCSz or NCSE alters the amount of neuronal damage; therefore, it is technically unclear if treating this phenomenon is beneficial. There is overwhelming evidence, however, in the form of elevated neuron-specific enolase (NSE) [13], that NCSz and NCSE have the potential to damage the brain. Elevations in NSE can also be seen after stroke, global cerebral ischemia, and coma. Elevated brain interstitial glutamate, lactate-pyruvate ratio, elevated intracranial pressure in NCSE lasting greater than 96 h, brain tissue hypoxia, increasing mass effect, and hippocampal atrophy on follow-up MRI are all evidence that NCSz and NCSE cause brain injury [14].

It can take a substantial amount of time to note clinical improvement in patients who have had NCSE once it is aborted. Therefore, lack of clinical improvement immediately after the resolution of the suspected electrographic pattern does not exclude NCSE. The general approach to the patient in SE should focus on the following: (1) terminating SE, (2) preventing its recurrence, (3) treating its complications, and (4) determining and managing its etiology.


Termination of Status Epilepticus


Studies have compared lorazepam, phenobarbital, diazepam, and phenytoin to phenytoin alone and determined that lorazepam was most likely to terminate convulsive SE. It was statistically significantly more likely to terminate SE than phenytoin although not statistically superior to the other agents [15]. It has emerged as the drug of choice for the initial treatment of SE because it is also faster and more convenient to administer than the other drugs tested. The dose of lorazepam studied was 0.1 mg/kg; lower doses may be efficacious but have not been systematically studied. At all time points studied, patients randomized to receive lorazepam were more likely to have stopped clinical seizure activity than those who received diazepam (or placebo) [15]. Confounding these studies, however, is the use of EEG. Further investigations in these studies went on to show that many of the patients who were no longer clinically seizing after “successful treatment” of SE were in fact still in NCSE. Thus, it is likely that studies only looking at clinical symptoms overestimate the efficacy of treatment. Recent studies have compared prehospital administration of IM midazolam with IV lorazepam, testing the theory that a rapidly absorbed IM drug would yield the same success rates as an IV drug because the IM drug could be administered more rapidly. The dose of midazolam was 10 mg and for lorazepam was 4 mg, and if patients weighed between 13 and 40 kg, the doses were halved. The study showed that the more rapidly administered midazolam was superior at terminating SE than lorazepam, likely because of the ease with which it could be given [16]. Most clinicians still agree, however, that if an IV line is already in place (such as in the inpatient setting), then IV lorazepam should be administered.

One of the most important confounding factors in assessing the efficacy of treatment of SE is the latency from seizure onset to treatment. Studies have investigated this based on the EEG pattern at the time treatment was initiated [11]. Table 1 shows the likelihood of terminating SE based on the initial EEG pattern, while Figs. 8, 9, and 10 demonstrate different SE patterns. While it is difficult to know the time of seizure onset, the progression from one EEG pattern to the next is well established and provides an estimate of the latency to treatment. For example, patients with a burst suppression pattern likely have been seizing for longer than those with a waxing-and-waning pattern. Patients classified as having subtle SE most likely have been experiencing seizures for a long period of time or have a catastrophic underlying condition, which helps to explain their poor response rates to any of the conventional antiepileptic drugs [11].


Table 1
Likelihood of successful overt status epilepticus termination in relation to initial EEG pattern

























Pattern

% treated successfully

Discrete seizures

75

Waxing and waning

30

Continuous (invariant) pattern

24

Brief suppressions

8

Burst suppression

7


A328697_1_En_24_Fig8_HTML.gif


Fig. 8
cEEG demonstrating SE, waxing-and-waning pattern


A328697_1_En_24_Fig9_HTML.gif


Fig. 9
cEEG demonstrating SE, invariant pattern


A328697_1_En_24_Fig10_HTML.gif


Fig. 10
cEEG demonstrating NCSE, burst suppression pattern in both the surface and depth electrodes (D leads, blue)

Available data shows that only the first anticonvulsant has a reasonable chance of terminating SE (Table 2) and does not have any implications on preventing recurrence [15]. While fosphenytoin is the only AED to be approved by the US Food and Drug Administration (USFDA) for the termination of SE, it is not superior to phenytoin as a first- or second-line agent. It is used because it carries a lower risk of complications related to intravenous infusion, although complications like bradycardia and hypotension are still possible. Other second-line agents with published data include valproate, levetiracetam, lacosamide, and topiramate (given enterally as no parenteral form is currently available). There is insufficient data to recommend one over the other at this time. However, the clinician should be familiar with the reasons to choose one agent over another, e.g., side effects or interactions with other medications.


Table 2
Response rates for subsequent antiepileptic drugs (overt status epilepticus patients only)




















































Initial agent

Drug

Response rate (%)

Lorazepam

Lorazepam

64.9

Phenytoin

7.2

Phenobarbital

2.1

Phenobarbital

Phenobarbital

58.2

Phenytoin

3.3

Lorazepam

2.2

Diazepam plus phenytoin

Diazepam and phenytoin

55.8

Lorazepam

3.2

Phenobarbital

2.1

Phenytoin

Phenytoin

43.5

Lorazepam

13.9

Phenobarbital

3

Regarding second-line agents, many believe that if SE has lasted through the time that it took to administer and assess the efficacy of the first-line AED, a more definitive treatment should be employed for the second-line treatment – usually an anesthetic agent (summarized in Table 3). Endotracheal intubation and mechanical ventilation should be started prior to the anesthetic agent. At this point, cEEG is necessary because these agents will usually terminate all movements before abolishing GCSE, and the only way for the intensivist to titrate the anesthetic agent to resolution of status epilepticus is via cEEG monitoring. Vasopressor and inotropic support is almost always necessary, as is support of core body temperature since anesthetic agents will often lead to poikilothermia. Enteral feeding may be possible, but parenteral nutrition may be required if an ileus develops. Infections are common, at least in part due to immunosuppression.


Table 3
Conventional second-line agents for terminating status epilepticus























































Agent

IV loading dose

Maintenance

Adverse effects

Comments

Valproate

20 mg/kg to 40 mg/kg at 5 mg/kg/min

4 mg/kg to 6 mg/kg every 6 h

Hepatic toxicity, thrombocytopenia, pancreatitis, induction of autoimmunity

Avoid in pregnancy or after head trauma; numerous drug interactions

Levetiracetam

1 g to 6 g at 2 mg/kg/min to 5 mg/kg/min

10 mg/kg to 15 mg/kg every 12 h

Accumulates when creatinine clearance is diminished

Minimal drug interactions

Lacosamide

200 mg to 400 mg over 15 min to 30 min

200 mg every 12 h

Somnolence, atrial fibrillation

Interactions with antiretroviral rugs

Topiramate

Not available for IV use; 400 mg enterally every 3 h to 4 h up to 2 g

300 mg every 6 h

Sedation, metabolic acidosis

Numerous drug interactions

Midazolam

0.2 mg/kg over 5 min

0.2 mg/kg/h to 2.0 mg/kg/h

Hypoventilation, hypotension

Tachyphylaxis occurs rapidly

Propofol

1 mg/kg to 5 mg/kg (depending on blood pressure and other drugs used) over 5 min to 10 min

Up to 15 mg/kg/h (increasing risk of propofol infusion syndrome above 5 mg/kg/h)

Propofol infusion syndrome (acidosis, rhabdomyolysis), hypotension, immunosuppression

Lipid vehicle is a substantial calorie source

Pentobarbital

5 mg/kg to 10 mg/kg at 50 mg/min; slow infusion for hypotension

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 12, 2017 | Posted by in NEUROLOGY | Comments Off on Critical Care Considerations

Full access? Get Clinical Tree

Get Clinical Tree app for offline access