You are on page 1of 12

Epilepsia, 54(3):393–404, 2013

doi: 10.1111/epi.12058

CRITICAL REVIEW AND INVITED COMMENTARY

Lacosamide as a new treatment option in status epilepticus


Julia H€
ofler and Eugen Trinka

Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University of Salzburg, Salzburg, Austria

epilepticus) treated with lacosamide. The most often used


SUMMARY
bolus dose was 200–400 mg over 3–5 min. The overall suc-
Status epilepticus is among the most common neurologic cess rate was 56% (76/136). Adverse events (AEs) were
emergencies, with a mortality rate of up to 20%. The most reported in 25% (34/136) of patients: mild sedation in 25
important therapeutic goal is fast, effective, and well- cases, 1 patient with possible angioedema, 2 with allergic
tolerated cessation of status epilepticus. Intravenous phe- skin reaction, 4 with hypotension, and 1 with pruritus. One
nytoin/fosphenytoin, phenobarbital, or valproate is the patient developed a third-degree atrioventricular (AV)
current standard treatment after failure of benzodiaze- block and paroxysmal asystole. Overall, the rate of AEs
pines. Lacosamide as a new antiepileptic drug has been was low. Current evidence on the use of intravenous laco-
available as an intravenous solution since 2009. To date, samide in acute seizures and status epilepticus is restricted
PubMed lists 19 studies (10 single case reports and 9 case to retrospective case reports and case series (class IV). Fur-
series), reporting a total of 136 episodes of refractory ther prospective studies to inform clinicians are necessary.
status epilepticus (50% nonconvulsive status epilepticus, KEY WORDS: Lacosamide, Acute seizures, Seizure
31% focal status epilepticus, and 19% convulsive status emergency, Treatment algorithm, Antiepileptic drug.

Status epilepticus (SE) is a life-threatening condition that Based on experimental evidence (St€ohr et al., 2007;
requires urgent and rigorous treatment with antiepileptic Wasterlain et al., 2011), safety studies in healthy individuals
drugs (AEDs). More than 60% of generalized convulsive and patients (Ben-Menachem et al., 2007; Biton et al., 2008;
SE will be controlled by first-line treatment with intra- Krauss et al., 2010), as well as retrospective case series with
venous (IV) lorazepam (LZP) (Leppik et al., 1983; Treiman SE and seizure clusters in humans (Kellinghaus et al., 2009;
et al., 1998; Alldredge et al., 2001) or intramuscular Tilz et al., 2010; Turpin-Fenoll et al., 2010; Albers et al.,
midazolam (MDZ) (Silbergleit et al., 2012), irrespective of 2011; Chen et al., 2011; Goodwin et al., 2011; Granda-
its cause. At least 40% of all patients will need further treat- Mendez et al., 2011; H€ofler et al., 2011; Kellinghaus et al.,
ment with IV AEDs. Despite its known disadvantages (such 2011; Koubeissi et al., 2011; Krause et al., 2011; LaRoche
as cardiac arrhythmias or hypotension), IV phenytoin & Shivdat-Nanhoe, 2011; Parkerson et al., 2011; Rantsch
(PHE) is frequently used to control the seizures, assuming et al., 2011; Shiloh-Malawsky et al., 2011; Torres-Cano
that treatment benefits outweigh the well-known risks et al., 2011; Cherry et al., 2012; Jain & Harvey, 2012; Mnat-
(Brophy et al., 2012). sakanyan et al., 2012), IV LCM may be a useful alternative
Lacosamide (LCM) became available in Europe in Sep- in emergency treatment of seizures with IV AEDs.
tember 2009 and in the United States in October 2009 as an The aim of the present review is to analyze all published
IV solution based on bioequivalence to the oral formulation studies on IV LCM in the treatment of SE and acute recur-
(Biton et al., 2008; Krauss et al., 2010). LCM is a function- rent seizures.
alized amino acid with anticonvulsant properties. Its mecha-
nism of action is the selective enhancement of sodium
channel slow inactivation (Beyreuther et al., 2007). LCM
Methods
was effective in different rodent seizure models for general- Identification of studies and search strategy
ized and complex partial seizures as well as for SE (Beyreu- All 19 studies were identified by electronic search of Pub-
ther et al., 2007; St€
ohr et al., 2007). Med (January 2009–May 2012) using the search terms:
“lacosamide” AND “intravenous” AND “status epilepti-
cus.” Only studies published as a full article were eligible if
Accepted October 25, 2012; Early View publication January 7, 2013. they reported the use of IV LCM in the treatment of SE.
Address correspondence to Eugen Trinka, Universit€atsklinik f€ur Neuro-
logie, Christian-Doppler-Klinik, Paracelsus Medical University, Ignaz-
Harrer-Str. 79, A-5020 Salzburg, Austria. E-mail: e.trinka@salk.at Mechanism of action of LCM
Wiley Periodicals, Inc. LCM is a functionalized amino acid with anticonvulsive
© 2013 International League Against Epilepsy properties (Andurkar et al., 1999; Hovinga, 2003; Duncan &

393
394
J. H€
ofler and E. Trinka

Kohn, 2005). LCM selectively enhances slow inactivation of Another multicenter double-blind, double-dummy, ran-
sodium channels, with no effect on fast inactivation (Erring- domized, inpatient trial supported the safety and utility of
ton et al., 2006). This mechanism is in contrast to other AEDs IV LCM as replacement for oral LCM. Intravenous LCM
such as carbamazepine (CBZ) or PHE, which do not enhance was administered as 60- or 30-min twice-daily infusions
slow inactivation. LCM shifts the voltage dependence of and showed a safety and tolerability profile similar to that of
inactivation to more hyperpolarized membrane potentials and oral LCM. Treatment-emergent AEs were reported by 16 of
thereby promotes the transition of sodium channels into the 60 patients—including dizziness, headache, back pain, som-
slow inactivated state (Beyreuther et al., 2007). nolence, and injection-site pain. All AEs were considered
mild or moderate in intensity, and no AE led to discontinua-
Pharmacokinetics of LCM tion (Biton et al., 2008).
Oral LCM is rapidly absorbed, and plasma concentra- In a safety and tolerability trial with 25 subjects, IV LCM
tions are dose proportional. With an elimination half-life of was applied in three progressively increasing doses (200,
13 h, a twice-daily dosing is possible. Steady state is 300, and 400 mg). Optimal tolerance was found with IV
reached within 2–3 days (Horstmann et al., 2002; Hovinga, loading doses of 200 and 300 mg administered over
2003; Cawello et al., 2004). LCM has a low protein binding 15 min; a higher frequency of dose-related AEs (dizziness,
(<15% bound) and no effect on the plasma concentrations somnolence, nausea, and diplopia) was observed with a
of concomitantly administered AEDs (Kropeit et al., 2004; loading dose of 400 mg (Fountain et al., 2011).
Jatuzis et al., 2005). A small proportion of LCM is metabo-
lized to an O-desmethyl metabolite that is not pharmacolog- Experimental basis for the use of LCM in SE
ically active in in vivo models (Doty et al., 2007; Cross & In an experimental model of limbic self-sustaining SE
Curran, 2009). LCM and its major metabolites are elimi- induced by perforant path stimulation, rats were treated with
nated renally. IV LCM (3, 10, 30, or 50 mg/kg) either after 10 min (early)
Because of its high water solubility, LCM was also devel- or after 40 min (late). Compared with the late treatment,
oped as an IV solution, in parallel with the oral formula- LCM given after 10 min showed a significant dose-dependent
tions. The isotonic solution contains 10 mg/ml and has a pH reduction of acute SE seizure activity (Wasterlain et al.,
of 3.5–5. The solution is stable at room temperature and can 2011). In other models, such as maximal electroshock test
be administered without dilution (Biton et al., 2008). in mice, LCM protected mice and rats against tonic-
However, it can also be diluted in sodium chloride 0.9%, extension seizures and prevented seizure spread (Swinyard
dextrose 5%, or lactated Ringers solution, if needed. In a et al., 1952; Borowicz et al., 1997). In the 6-Hz psychomo-
phase 1 trial with 24 healthy volunteers, bioequivalence of tor seizure test, LCM demonstrated full efficacy in contrast
short-time infusion was demonstrated as a replacement ther- to other AEDs such as LTG, PHE, and CBZ (Barton et al.,
apy for oral LCM at doses of 200 mg twice daily and IV 2001). However, LCM was not effective in chemoconvul-
infusions at 30 and 60 min (Kropeit et al., 2004). sant-induced seizures induced by the subcutaneous bolus
Krauss et al. (2010) reported a multicenter, open-label, injection of pentylenetetrazole (St€ohr et al., 2007).
inpatient, dose-escalation trial with 160 patients from ongo-
ing open-label, long-term trials who were taking stable Clinical studies in SE
doses of oral LCM and up to three concomitant AEDs. The There are 19 published articles on the use of IV LCM in
patients received IV LCM dosed over progressively shorter SE and acute repetitive seizures, including 10 single case
infusion durations: 30, 15, and 10 min for 2–5 days. Most reports (Kellinghaus et al., 2009; Tilz et al., 2010; Turpin-
patients received IV LCM 200–600 mg/day; around 4% (7/ Fenoll et al., 2010; Chen et al., 2011; Granda-Mendez
160) received 700–800 mg/day in 15 min. et al., 2011; Krause et al., 2011; LaRoche & Shivdat-
The adverse event (AE) rate was low, the most common Nanhoe, 2011; Parkerson et al., 2011; Shiloh-Malawsky
(10% or less) being headache, dizziness, diplopia, and som- et al., 2011; Torres-Cano et al., 2011; see Table 1) and 9
nolence. The occurrence of AEs did not correlate with retrospective case series (Albers et al., 2011; Goodwin
shorter infusion time, but with higher doses of LCM. Two et al., 2011; H€ofler et al., 2011; Kellinghaus et al., 2011;
patients had cardiac AEs: one patient treated with beta- Koubeissi et al., 2011; Rantsch et al., 2011; Cherry et al.,
blockers had a reversible bradycardia of 26 bpm during a 2012; Jain & Harvey, 2012; Mnatsakanyan et al., 2012; see
15-min infusion on day 2 of treatment with 300 mg/day IV Table 2).
LCM; the second patient had a nonrecurring prolonged
QTcB interval, without clinical symptoms or signs, on day 4 Single case reports
of treatment with 100 mg/day IV LCM by 15-min infusion. Kellinghaus et al. (2009) published a case of a 42-
It should be noted that all patients were long-term respond- year-old woman who had preexisting epilepsy after a severe
ers to LCM in the open-label extension trials, which cardioembolic stroke in the territory of the left middle cere-
enriches the study population with those patients who toler- bral artery in 1993. After SE in 2006 with vomiting, worsen-
ate LCM well (Krauss et al., 2010). ing of her preexisting aphasia, hemiparesis, and nystagmus,
Epilepsia, 54(3):393–404, 2013
doi: 10.1111/epi.12058
Table 1. Overview of all case reports of SE treated with IV LCM
Why? Indication? When? Conditions? How?
Time from
SE onset to
SE specific AEDs before Order LCM LCM LCM Adverse
Study SE (n) Age (y) SE type SE etiology etiology LCM (mg) BDZ of LCM initiation dose route Outcome events
Kellinghaus 1 42 NCSE Remote Stroke IV DZP (2.5); Y 3rd  1.5 h 200 mg IV Responder Possible skin
et al. symptomatic IV LZP (6) (within after 5 itching and
(2009) 3–5 min) min mild
erythema
Tilz et al. 1 38 CSE Remote Perinatal hypoxia IV DZP (22.5); Y 6th (with Not 300 mg PEG- Responder N
(2010) symptomatic with resulting IV etomidate (12.5); etomidate) reported tube after 30
infantile cerebral IV MDZ (5); enteral (Syrup) min
palsy LZP (4); enteral LEV
(1,500)
Turpin-Fenoll 1 72 NCSE Remote Stroke LEV (3,000), VPA, Y 5th Not 25 mg PO Responder N
et al. (2010) symptomatic CLN, OXC (dose reported twice daily 1 week
and route not after
reported) LCM
was
started
Parkerson 17 75 Focal SE Remote GBM Not reported Y Not Not 100 mg IV Responder N
et al. (2011) symptomatic reported reported twice daily
Chen et al. 1 57 Focal SE/EPC Remote Traumatic LZP (2); FOS (1,000); Y 4th 4 days 50 mg PO Responder N
(2011) symptomatic brain injury LEV (3,000); LZP (2); twice per
VPA (loading day for 5
20 mg/kg, maintain days, then
10 mg/kg every 6 h); uptitration
LEV daily dose to 400
increased to mg/day
4,000 mg, PHE within
administration 8 days
within a serum level
of 20–30 lg/ml
Krause et al. 1 89 NCSE Acute Not reported IV LZP (2); Day 1: Y 3rd Not Two 400-mg IV Responder Third-degree
(2011) symptomatic IV LEV (2,000) reported bolus AV block
Day 2: IV LEV doses and asystolia
(3,000) within 6 h
Granda- 1 81 NCSE Acute Stroke IV CLN (2); IV VPA Y 3rd Not 100 mg/day IV Responder N
Mendez symptomatic (40 mg/kg) and VPA reported
et al. (2011) 800 mg/day over
continuous infusion

Continued
IV Lacosamide in Seizure Emergencies

Epilepsia, 54(3):393–404, 2013


395

doi: 10.1111/epi.12058
396
J. H€

doi: 10.1111/epi.12058
Epilepsia, 54(3):393–404, 2013
Table 1. Continued.
ofler and E. Trinka

Why? Indication? When? Conditions? How?


Time from
SE onset to
SE specific AEDs before Order LCM LCM LCM Adverse
Study SE (n) Age (y) SE type SE etiology etiology LCM (mg) BDZ of LCM initiation dose route Outcome events
LaRoche & 1 61 NCSE Acute SESA LZP; FOS; VPA; Y 4th Not Not IV Responder N
Shivdat- symptomatic doses not reported reported reported
Nanhoe
(2011)
Torres- 1 49 NCSE Remote Polymicrogyria DZP (5); VPA Y 7th 15 days 200 mg/12 h IV Responder N
Cano symptomatic and nodular (500 + 500 mg/12 h);
et al. heterotopia LEV (1,000);
(2011) CLN (1 g/12 h);
LTG (200 mg/2 h);
propofol and MDZ
per perfusor; PHE
Shiloh- 1 8 CSE?NCSE Acute Unclear LZP; PHE; PB; VPA; Y 8th 10 weeks 25 mg Enteral Responder; N
Malawsky symptomatic LEV; FBM; TPM; twice 5 days
et al. (2011) propofol; daily after
corticosteroids; IVIG; LCM
plasmapheresis was
started,
SE was
resolved

AED, antiepileptic drug; BDZ, benzodiazepines; CLN, clonazepam; CSE, convulsive status epilepticus; DZP, diazepam; EPC, epilepsia partialis continua; FBM, felbamate; FOS, fosphenytoin; GBM, glioblastoma multi-
forme; IV, intravenous; IVIG, intravenous immunoglobulin; LCM, lacosamide; LEV, levetiracetam; LTG, lamotrigine; LZP, lorazepam; MDZ, midazolam; NCSE, nonconvulsive status epilepticus; OXC, oxcarbazepine; PB,
phenobarbital; PEG tube, Percutaneous gastrostomy tube; PHE, phenytoin; PO, orally; RSE, resistant status epilepticus; SE, status epilepticus; SESA, subacute encephalopathy and seizures in alcoholics; TPM, topiramate;
VPA, valproic acid.
Table 2. Overview of all case series of SE treated with IV LCM
Why? Indication? When? Conditions? HOW?

Time from
Episodes SE onset to
Episodes of SE SE etiology, SE specific AEDs before BDZ, Order LCM LCM dose LCM Outcome, Adverse
Study of SE (n) Age (y) by type (n) % (n) pts etiology, n/N LCM, % (n) pts % (n) pts of LCM initiation (mg) route % (n) events (n)

Kellinghaus 39 Median 62 6 CSE 26% (10/39) Acute Not reported 95% (37/39) 95% (37/39) 13% (5/39) Median latency Median bolus IV 100% (5/5) LCM as 1 Allergic skin
et al. (2011) (range, 16 NCSE symptomatic; BDZ; 85% (33/39) pts LCM 30 (range, dose 400 1st/2nd; 95% reaction; 25
18–90) 17 focal SE 23% (9/39) LEV; 36% (14/39) as 1st/2nd; 0.5–1,440) h (range, (18/19) LCM as Sedation; 4
Remote PHE 13% (5/39) 49% (19/39) 200–400; 3rd; 73%(11/15) Hypotension
symptomatic other AEDs; 10% pts LCM as mean dose LCM as 4th/later
(new-onset (4/39) Anesthesia 3rd; 38% 1st day 424
epilepsy); (15/39) (range,
49% (19/39) pts LCM 200–600)
Remote as 4th/later
symptomatic
(preexisting
epilepsy);
3% (1/39)
Others/NK
Koubeissi 4 Median 65 4/4 NCSE 75% (3/4) Acute 2/4 Hemorrhage 100% (4/4) LZP; 50% 100% (4/4) Median4 Median 34 Median initial IV 100% (4/4) N
et al. (2011) (range, symptomatic; 1/4 Meningioma (2/4) FOS; 100% (range, 3–5) (range, dose 100 (range,
53–79) 25% (1/4) 1/4 Temporal (4/4) LEV; 25% 3–50) h 50–100)
Preexisting encephalomalacia (1/4) PGB; 25%
epilepsy (1/4) VPA
Goodwin 9 Median 63 6/9 NCSE 4 Preexisting 2/9 rapid 100% (9/9) LEV, 100% (9/9) Median 3 Median 2 Median loading IV 100% (9/9) 2 Episodes of
et al. (2011) (range, 3/9 epilepsy neurodegenerative 78% (7/9) PHE, (range, 2–5) (range, dose 200 (range, Nonresponder angioedema;
47–89) CSE 5 Acute disease; 1/9 Rosai– 22% (2/9) 0–14) days 100–300)
symptomatic Dorfman disease; Pentobarbital coma
1/9 GBM and stroke;
1/9 Aneurysms;
1/9 Stroke;
1/9 Meningioma;
1/9 VPS infection;
1/9 No underlying
neurologic disease
Albers et al. 7 Median 63 7/7 focal SE 42% (3/7) Acute 3/7 SDH; 1/7 History 29% (2/7) VPA; 42% (3/7) Median 4 Not Median initial IV 100% (7/7) N
(2011) (range, symptomatic; of stroke; 1/7 100% (7/7) (range, 2–5) reported dose 400 Responder
33–83) 29% (2/7) Alcohol abuse and LEV; 4/7 propofol; (range, 400)
Remote hypertension; 14% (1/7) PHE;
symptomatic; 1/7 ICH; 1/7 42% (3/7) TPM;
29% (2/7) Infantile 42% (3/7) MDZ
Pre-existing brain damage
epilepsy

Continued
IV Lacosamide in Seizure Emergencies

Epilepsia, 54(3):393–404, 2013


397

doi: 10.1111/epi.12058
398
J. H€

Table 2. Continued.
Why? Indication? When? Conditions? HOW?

Time from

doi: 10.1111/epi.12058
Episodes SE onset to
Episodes of SE SE etiology, SE specific AEDs before BDZ, Order LCM LCM dose LCM Outcome, Adverse

Epilepsia, 54(3):393–404, 2013


Study of SE (n) Age (y) by type (n) % (n) pts etiology, n/N LCM, % (n) pts % (n) pts of LCM initiation (mg) route % (n) events (n)
ofler and E. Trinka

H€
ofler et al. 31 Median 67 11/31 CSE; 19% (6/31) Acute 1/31 NK; 5/31 68% (21/31) LEV; 94% (29/31) 7% (2/31) Not listed Median 200 IV Overall In group SE no
(2011) (range, 10/31 symptomatic; Traumatic 94% (29/31) BDZ; pts LCM (range, 81% AEs
22–95) NCSE; 35% (11/31) lesions; 1/31 10% (3/31) PHE; 1st; 19% 200–400) (25/31)
10/31 Remote Cortical dysplasia; 16% (5/31) VPA; (6/31) pts Responders
focal SE symptomatic; 9/31 Vascular 3% (1/31) LCM 2nd; LCM 1st 2/2,
46% (14/31) lesions; 3/31 Anesthesia 48% (15/31) LCM 2nd 6/6,
Preexisting Tumor; 1/31 pts LCM 3rd; LCM 3rd
epilepsy Encephalitis, 26% (8/31) 11/15, LCM
11/31 Others pts LCM 4th or
4th or later later 6/8
Cherry et al. 13 Median 51.1 7/13 NCSE; 9/13 Preexisting Not reported 92% (12/13) LZP; 92% (12/13) Mean 3.2 Mean 39.5 Mean 180.8 IV 38% (5/13) 3/13 Episodes
(2012) (range, 4/13 epilepsy 85%(11/13) (range, 1–7) (range (range, Complete of significant
24–80) focal SE; LEV; 69% (9/13) 6.5–74) h 100–400); seizure hypotension;
2/13 FOS; 31% (4/13) Infusion rate, cessation; 54% 1/13 Fever
CSE VPA; 31% mean 3.33 (7/13)  50% without
(4/13) PB (range, Reduction source;
1.67–6.67) in seizure 1/13 Elevated
mg/min frequency liver function
tests
Jain & Harvey 3 Median 16 3/3 refractory 3/3 Pre-existing 1/3 Lissencephaly; 3/3 LEV; 2/3 VPA; 100% (3/3) Median 6 Median Median 100 IV 100% (3/3) 1/3 Chorea; 1/3
(2012) (range, tonic SE epilepsy 1/3 Rett syndrome; 3/3 CLZ; 1/3 PB (range, 5–7) 28 (range, (range, 50–200) Oculogyric
12–17) 1/3 Generalized 8–29) h crisis
epilepsy
Mnatsakanyan 10 Median 60.5 10/10 NCSE 60% (6/10) 1/10 NK; 2/10 40% (4/10) BDZ; 40% (4/10) Median 4 Not Median loading IV 70% (7/10) N
et al. (2012) (range, Acute Asystole; 2/10 80% (8/8) PHE; (range, 2–8) reported dose 200–300 Responder
16–90) symptomatic; Tumor; 1/10 HSV 90% (9/10) LEV; within 30 min
40% (4/10) encephalitis; 30% (3/10) VPA;
Preexisting 1/10 Frontal 10% (1/10)
epilepsy AVM resection; Propofol
1/10 Metastases;
1/10 Stroke;
1/10 Posterior
reversible
encephalopathy
syndrome

AED, antiepileptic drug; AVM, arteriovenous malformation; BDZ, benzodiazepines; CLN, clonazepam; CSE, convulsive status epilepticus; EPC, epilepsia partialis continua; FOS, fosphenytoin; GBM, glioblastoma multi-
forme; HSV, herpes simplex virus; ICH, intracerebral hemorrhage; LCM, lacosamide; LEV, levetiracetam; LZP, lorazepam; MDZ, midazolam; NCSE, nonconvulsive status epilepticus; NK, not known; PB, phenobarbital;
PGB, pregabalin; PHE, phenytoin; SDH, subdural hematoma; SE, status epilepticus; TPM, topiramate; VPA, valproic acid; VPS, ventriculoperitoneal shunt.
399
IV Lacosamide in Seizure Emergencies

she received levetiracetam (LEV) 2,000 mg/day. Under this particular, no change of PR intervals prior to LCM or at the
therapy, she developed psychotic symptoms; therapy was end of infusion.
therefore changed, first to lamotrigine (LTG) and then to Chen et al. (2011) reported a patient with epilepsia parti-
gabapentin (GBP). Due to noncompliance with AED dos- alis continua: a 57-year-old man with right-sided hemipare-
ing, four further episodes of SE followed. In October 2008, sis with cognitive and language impairment after a brain
the patient had her sixth SE with her habitual clinical semi- injury at age 25, who was admitted because of involuntary
ology. Previous medication was LTG 200 mg and GBP jerking of the right arm and leg. EEG showed a continuous
1,200 mg/day. In the emergency situation, she received IV left posterior slowing and epileptiform rhythms, with hyper-
diazepam (DZP) 2.5 mg. Because of rhythmic slowing and intense areas in diffusion-weighted imaging and fluid-
spiking on the left posterior temporal area, IV LZP 6 mg attenuated inversion recovery (FLAIR) sequences observed
was administered, but without success, and the patient on magnetic resonance imaging (MRI). The patient received
developed respiratory problems. Because of the enzyme- IV LZP 2 mg, IV fosphenytoin (FOS) 1,000 mg, and IV
inducing effect of valproic acid (VPA) and previous treat- LEV 3,000 mg, but clinical status and EEG remained
ment with LTG, VPA was not used; LEV was also avoided, unchanged. Furthermore, IV LZP 2 mg, IV VPA (loading
because of her previous psychiatric episode. Instead, IV dose 20 mg/kg), increase of IV LEV daily dose from 3,000
LCM was administered as a bolus of 200 mg within 3– to 4,000 mg, and PHE administration within a serum level
5 min; epileptic activity on electroencephalography (EEG) of 20–30 lg/ml achieved no seizure control. After 4 days of
ceased about 5 min later. After 6 days, the patient devel- refractory SE, an oral dose of LCM 100 mg/day was started
oped mild pruritus. and all other AEDs except LEV were tapered down. On day
Tilz et al. (2010) described a 38-year-old man who had 4, the SE was stopped clinically and electrographically. This
preexisting epilepsy with infantile cerebral palsy. Diagnosis low dose of LCM was claimed to be effective, but of course
on admission was a series of complex partial seizures, which SE may have resolved unrelated to LCM.
evolved into generalized tonic–clonic SE. PHE and VPA Krause et al. (2011) described an 89-year-old woman
had been ineffective in the past. Initially, the patient who was admitted to the hospital with an NCSE. She had a
received IV DZP 22.5 mg, IV etomidate 12.5 mg, and IV history of arterial hypertension, heart insufficiency, and
MDZ 5 mg within 45 min. Because of ongoing SE, LZP hypothyroidism, and had been treated with a beta-blocker
4 mg and LEV 1,500 mg were administered via percutane- and amlodipine. Laboratory tests showed a potassium level
ous gastric fistula within 30 min. Because of the persistent of 2.5 mEq/L, so supplementation was started. For SE, the
SE, treatment with a 150-mg LCM minced tablet powder patient initially received IV LZP 2 mg; SE continued, and
was given via percutaneous gastrostomy tube, following she developed respiratory problems. On day 1, IV LEV
which the seizures were interrupted; a second dose of 2,000 mg was administered as a bolus within 60 min, fol-
150 mg LCM syrup stopped SE after 30 min. Lower doses lowed by 3,000 mg on day 2. Because of persistent SE, IV
of AEDs were used because of the patients low body LCM was administered in two 400-mg bolus doses within
weight. 6 h, following which the patient developed third-degree AV
Turpin-Fenoll et al. (2010) presented a 72-year-old block and asystole three times, with duration of  10 s.
patient with a nonconvulsive SE (NCSE) 2 weeks after his After infusion 30 min later with 500 ml hydroxyethyl
first tonic–clonic seizure. As PHE had previously caused starch, sinus rhythm with first-degree AV block returned;
bradycardia and respiratory failure, it was avoided as first- the PQ interval normalized over the course of the next day.
line therapy. The patient received IV LCM after failed ther- Possible reasons for the occurrence of AV block included
apy with LEV (3,000 mg/day), oxcarbazepine, VPA, and the low potassium level, negative effects of premedication
clobazam. One week after NCSE had started, the seizures on AV conduction, and the low glomerular filtration rate.
were stopped with a dose of LCM 50 mg/day. Because of The authors concluded that high doses of LCM should be
the low dose of LCM, the authors discussed a remote effect avoided in patients at risk for AV conduction delay.
of previously given drugs. One 61-year-old patient with NCSE during the course of
Parkerson et al. (2011) reported on 17 patients who subacute encephalopathy with seizures was fully controlled
received LCM (13 intravenously) who were undergoing by IV LCM after failure of LZP, FOS, and VPA. Dose and
prolonged video-EEG monitoring. Three patients had no duration of SE before LCM were not reported. No AEs were
seizures, but periodic lateralized epileptiform discharges noted (LaRoche & Shivdat-Nanhoe, 2011).
plus (PLEDs-plus) or generalized periodic discharges. Nine Shiloh-Malawsky et al. (2011) reported an 8-year-old
patients had repetitive focal seizures and EEG changes (like boy with convulsive seizures 2 days after a febrile illness.
PLEDs or PLEDs-plus). One 75-year-old male patient had Because of frequent focal seizures, he received PHE, pheno-
focal SE resulting from a glioblastoma multiforme and was barbital (PB), VPA, and LEV, but without success; thus he
taking LEV and PHE at admission. He received a benzodi- had up to 300 focal-onset electrographic seizures per
azepine as first-line therapy, then IV LCM 200 mg/day, and day under a combination of six AEDs (PHE, LEV, PB,
the focal SE was stopped. There were no AEs reported; in topiramate [TPM], felbamate [FBM], and VPA). He also
Epilepsia, 54(3):393–404, 2013
doi: 10.1111/epi.12058
400
J. H€
ofler and E. Trinka

received propofol, pentobarbital, MDZ, and ketamine, until Rantsch et al. (2011) reported on nine patients with 10
a burst-suppression EEG pattern, for up to 1 week and a ke- episodes of SE (eight NCSE and two focal SE). The etiol-
togenic diet for 6 weeks. Doses for AEDs and anesthetics ogy was remote in six patients, and acute symptomatic in
were not reported. Because of a possible autoimmune etiol- three. Median administration of LCM was as the sixth drug
ogy, the patient was treated with high-dose corticosteroids, (range 4th–12th), with an initial dose of 50–100 mg. In 2
IV immunoglobulin, and plasmapheresis, but again without (20%) of 10 episodes of SE, LCM was the last drug. AEs
effect. Ten weeks after refractory SE (RSE), LCM 25 mg were not reported. Three patients died during the observa-
was administered enterally twice daily; seizure frequency tion period due to complications of their underlying dis-
decreased significantly within 3 days, with seizures ceasing ease. In this study, the investigators found a highly
altogether after 5 days. It is possible that the response after significant (r = 0.94, p < 0.00005) correlation between age
3 days of therapy with low dose LCM may have been noth- and outcome: patients aged <60 years returned to baseline,
ing to do with the LCM. whereas those aged >75 years died. However, this is an
Granda-Mendez et al. (2011) presented the case of an 81- uncontrolled small series and these data must be interpreted
year-old woman with an acute symptomatic SE after an with caution.
ischemic infarction of the left/right posterior cerebral artery. Goodwin et al. (2011) described nine patients with RSE,
Initially she received IV clonazepam (CLN) 2 mg, followed which was defined as seizure lasting >30 min, or multiple
by IV VPA at a dose of 40 mg/kg, and then 800 mg/day seizures without return of consciousness, and a failure of at
continuous infusion. Because of comorbidities (chronic least two medications to abort SE. Six of their patients had
renal failure), PHE and LEV were avoided and the patient NCSE; three had CSE. Four had a preexisting epilepsy and
received IV LCM 100 mg/day. No AEs were reported. five were acute symptomatic. Patients had three (range 2–5)
Torres-Cano et al. (2011) reported a 49-year-old woman AEDs prior to LCM, and all received benzodiazepines as
with an NCSE. She had symptomatic epilepsy due to poly- first-line treatment. Response to LCM was defined for the
microgyria and nodular heterotopia. The premedication was patients in burst suppression as an absence of electrographic
VPA 500 mg twice daily, CLN 1 mg twice daily, and LEV seizure activity for 24 h; for patients not in burst suppres-
500 mg twice daily. In the emergency department she sion, response was defined as electrographic seizure resolu-
received IV DZP 5 mg, IV VPA 500 mg, and IV LEV tion  4 h after LCM administration. None of their patients
500 mg before being admitted to the intensive care unit, had their SE controlled with IV LCM, and only one patient
where she received another IV LEV 500 mg over 12 h, IV had an improvement of his EEG, without clinical improve-
CLN (1 g/12 h), and IV VPA (500 mg/12 h). Because of ment. Two patients had a decrease of seizure frequency with
generalized convulsions, the patient received propofol and PHE and LCM. Two patients died during the observation
MDZ by perfusor. After 10 days, the SE persisted; treat- period due to their underlying disease. The median initial
ment was IV CLN 1 g/12 h, IV LTG 200 mg/12 h, and dose was 200 mg (range 100–300). Two patients had an-
PHE. On day 15, IV LCM 200 mg/12 h was added, and SE gioedema during LCM treatment.
resolved at day 17. Koubeissi et al. (2011) reported on four patients with
NCSE. Etiology was acute symptomatic in three patients;
Retrospective case series one had a preexisting epilepsy. One patient had a temporal
Kellinghaus et al. (2011) reported the pooled experience encephalomalacia, one had a meningioma, and two had an
of four centers in Germany, Austria, and Switzerland. Alto- intracerebral hemorrhage; they all received benzodiaze-
gether, 39 patients receiving IV LEV for the treatment of pines as first-line drugs. LCM was on median the fourth
SE (six convulsive SE [CSE], 17 NCSE, and 16 focal SE) AED (range 3rd–5th) and the initial dose was in the range of
refractory to benzodiazepines were included. Acute symp- 50–100 mg. All patients were responders and no AEs were
tomatic etiology was found in 10 patients (26%), remote observed. Positive response or “seizure freedom” was
symptomatic in 9 (23%), and preexisting epilepsy in 19 defined as a reduction of seizure frequency from two to
(49%). Specific etiologies were not reported for individual three seizures per hour to one seizure every 2 h.
patients. LCM was successfully used as first or second Another retrospective study (Albers et al., 2011)
drug in 3 of 5 patients, as third drug in 11 of 19 patients, described seven patients treated with IV LCM for focal SE.
and as fourth or later drug in 3 of 15 patients. Median bolus Three had acute symptomatic, two remote symptomatic,
dose was 400 mg (range 200–400). In 44% of patients and two a preexisting epilepsy. Three patients had intracere-
(17/39), SE was controlled by LCM; only mild AEs were bral subdural hematomas; one had a history of ischemic
reported (1 allergic skin reaction, 25 sedation, 4 hypoten- stroke, one patient a history of infantile brain damage, and
sion). Of the 39 patients, 37 received benzodiazepines as one patient a history of intracerebral bleeding. One patient
first-line treatment. When LCM was administered early (as had alcohol abuse and hypertension. LCM was used on
first, second, or third drug) it was successful in 60%, median as fourth drug (range 2nd–5th). Initial loading was
whereas only 20% were treated successfully in later stages 400 mg/day. Response to LCM was defined as resolution of
of SE. EEG status within 24 h. In all patients, SE was resolved and
Epilepsia, 54(3):393–404, 2013
doi: 10.1111/epi.12058
401
IV Lacosamide in Seizure Emergencies

no serious AEs were reported. Four of the seven patients patients, there was an immediate clinical response to LCM;
had no initial treatment with benzodiazepines, whereas one the other had resolution of SE after the second dose.
received this only during the course of treatment.
Cherry et al. (2012) reported on 24 patients: 10 with 13
episodes of RSE (defined as SE with failed response to
Route of Administration of LCM
 2 drugs), and 14 with isolated seizures. Specific etiolo- Single case reports
gies for SE were not reported in detail. Among 13 episodes The route of administration was oral in one patient (Chen
of RSE, 7 were nonconvulsive, 4 focal, and 2 convulsive. et al., 2011), enteral in three (Tilz et al., 2010; Turpin-
The median use of LCM was as third drug (range 1st–7th). Fenoll et al., 2010; Shiloh-Malawsky et al., 2011), and IV
Concurrent AEDs were LZP in 92% (12/13), LEV in 85% in the remaining six patients (Granda-Mendez et al., 2011;
(11/13), FOS in 69% (9/13), VPA in 31% (4/13), PB in Kellinghaus et al., 2011; Krause et al., 2011; LaRoche &
22% (6/13), “other” in 19% (5/13), and sedatives or anes- Shivdat-Nanhoe, 2011; Parkerson et al., 2011; Torres-Cano
thetics in 33% (9/13). Complete seizure cessation was et al., 2011).
achieved in 5 of 13 episodes (38%) of SE, and seizure
reduction of >50% in 7 of 13 episodes (54%). The median Case series
loading dose was 180.8 mg (range 100–400 mg) and mean We divided the case series into two subgroups, according
maintenance dose 361.5 mg/day. Three episodes of signifi- to the number of included patients: Group I included all
cant hypotension, and one fever with unknown origin, were studies with a study population of 3–29 patients (Albers
observed. One patient had elevated liver function test lead- et al., 2011; Goodwin et al., 2011; Koubeissi et al., 2011;
ing to discontinuation of LCM. Rantsch et al., 2011; Cherry et al., 2012; Jain & Harvey,
Our own group (H€ ofler et al., 2011) reported on 48 2012; Mnatsakanyan et al., 2012), whereas Group II
patients: 17 (35%) with seizure clusters (SCs) and 31 (65%) included studies with >30 patients (H€ofler et al., 2011;
with SE. The etiology was acute symptomatic in six (19%) Kellinghaus et al., 2011).
and remote symptomatic in 11 (36%) patients, whereas 14 In both groups, all episodes (126/126) of SE were treated
patients (45%) had a preexisting epilepsy. Altogether, 32% with IV LCM (Albers et al., 2011; Goodwin et al., 2011;
had NCSE, 36% had a convulsive, and 32% had a focal SE. H€ofler et al., 2011; Kellinghaus et al., 2011; Koubeissi
Median initial bolus dose was 200 mg (range 200–400 mg). et al., 2011; Rantsch et al., 2011; Cherry et al., 2012; Jain
LCM was used as first drug in two patients (7%); in both, & Harvey, 2012; Mnatsakanyan et al., 2012).
SE was controlled. In six patients (19%), LCM was used as
second drug; again, all of these patients responded. In 15 Type and Outcome of SE; Dose
patients (48%), LCM was administered as the third drug,
and in 11 (73%) of these SE was stopped. Eight patients
and Order of LCM
(26%) received LCM as fourth or later drug, and six of these Single case reports
(75%) were responders. Overall, we observed cessation of Seven of 10 reported SE were NCSE (Kellinghaus et al.,
SE in 81% (25/31). No serious AEs were identified. 2009; Turpin-Fenoll et al., 2010; Granda-Mendez et al.,
Mnatsakanyan et al. (2012) identified 10 patients with 2011; Krause et al., 2011; LaRoche & Shivdat-Nanhoe,
NCSE, of whom 6 had an acute symptomatic SE. Specific 2011; Shiloh-Malawsky et al., 2011; Torres-Cano et al.,
etiologies were found in 9 of 10 patients: glioma resection, 2011), one a CSE (Tilz et al., 2010), and two a focal SE
herpes simplex virus encephalitis, frontal arteriovenous (Chen et al., 2011; Parkerson et al., 2011). All 10 SE
malformation, multifocal glioma, brain metastases, stroke, reported by single case reports stopped after LCM treatment
posterior reversible encephalopathy syndrome, and anoxic (Kellinghaus et al., 2009; Tilz et al., 2010; Turpin-Fenoll
brain injury due to asystole. The median use of LCM was as et al., 2010; Chen et al., 2011; Granda-Mendez et al., 2011;
fifth drug (range 3rd–9th), including initial treatment with Krause et al., 2011; LaRoche & Shivdat-Nanhoe, 2011;
LZP. The median loading dose was 200–300 mg within Parkerson et al., 2011; Shiloh-Malawsky et al., 2011; Tor-
30 min, with a maintenance dose in the range of 100– res-Cano et al., 2011). In one of 10 case reports, the initial
200 mg every 12 h. SE resolved in seven patients; AEs dose was not specified (LaRoche & Shivdat-Nanhoe, 2011);
were not reported. In two of three nonresponders, the spe- the median initial dose of the other nine case reports was
cific etiology was anoxic brain injury. 100 mg (range 50–400 mg). The lowest doses of 25 mg
Jain & Harvey (2012) described three patients (ages 12–17 twice daily were applied by enteral route for a 72-year-old
years) with symptomatic generalized epilepsy with intel- patient and an 8-year-old patient (Turpin-Fenoll et al.,
lectual disability and refractory tonic SE. LCM bolus doses 2010; Shiloh-Malawsky et al., 2011).
(2–2.5 mg/kg; 50–200 mg in the first 2 h) were used after Order of LCM in the treatment algorithm: median use of
three or more standard AEDs had been ineffective. In two LCM was as fourth drug (range 2nd–8th) (Kellinghaus
patients, side effects were reported (chorea and oculogyric et al., 2009; Tilz et al., 2010; Turpin-Fenoll et al., 2010;
crisis). Duration of SE prior to LCM was 8–29 h. In two Chen et al., 2011; Granda-Mendez et al., 2011; Krause
Epilepsia, 54(3):393–404, 2013
doi: 10.1111/epi.12058
402
J. H€
ofler and E. Trinka

et al., 2011; Shiloh-Malawsky et al., 2011; Torres-Cano The loading dose of LCM was higher in group II (200–
et al., 2011). In 2 of 10 case reports this information was 400 mg) than in group I (50–400 mg).
missing (LaRoche & Shivdat-Nanhoe, 2011; Parkerson
et al., 2011). One 8-year-old patient received LCM as Titration Time
eighth drug, following treatment with LZP, PHE, PB, VPA,
LEV, FBM, TPM, and other therapies (propofol, corticos- Given the retrospective nature of the studies and varia-
teroids, IV immunoglobulin, and plasmapheresis). One tions in reporting strategies, it is difficult to evaluate titra-
patient received LCM as the second drug (Kellinghaus tion times: this information was supplied only in case
et al., 2009). reports, not in case series.

Case series Single case reports


In group I, there were 63% (35/56) NCSE, 23% (13/56) Chen et al. (2011) used a titration time of 8 days to reach
focal SE, and 14% (9/56) CSE. Half (28/56) were responders: a daily dose of 400 mg in a case of partial SE. In the case of
among these, 43% (12/28) had NCSE, 29% (8/28) focal SE, the 8-year-old patient (Shiloh-Malawsky et al., 2011),
and 11% (3/28) CSE. Comparing the different types, there LCM was administered at a dose of 25 mg twice daily, with
was more focal SE in the responder group (eight patients, vs. resolution of SE after 5 days. In the case reported by Kel-
one nonresponder) but no notable differences for NCSE (12 linghaus et al. (2009) IV LCM was administered as a 200-
vs. 16 responders/nonresponders) or CSE (three responders, mg bolus within 3–5 min, followed by oral LCM 100 mg
three nonresponders). In one study of 13 patients, the type of twice daily from day 2. In the study described by Torres-
SE was not specified (Cherry et al., 2012). Cano et al. (2011), LCM was given at a dose of 200 mg/
In group I the outcome in terms of seizure control varied 12 h. All patients reported by case reports were responders
enormously, between 0% and 100%: 0% (1/7 studies: nine to IV LCM.
patients with NCSE; Goodwin et al., 2011), 20% (1/7 stud-
ies: 2/10 patients; one NCSE, one focal SE; Rantsch et al., Duration of SE until LCM
2011), 38% (1/7 studies: 5/13 patients; types of SE of non-
responders were not reported; Cherry et al., 2012), 70%
Therapy
(1/7 studies: 7/10 patients with NCSE; Mnatsakanyan et al., Single case reports
2012), and up to 100% (3/7 studies: four patients with Duration of SE until LCM application was detailed in 5
NCSE; Koubeissi et al., 2011; and seven patients with par- of 10 case reports (Kellinghaus et al., 2009; Chen et al.,
tial SE; Albers et al., 2011). In the studies with 100% 2011; Krause et al., 2011; Shiloh-Malawsky et al., 2011;
responders, the loading dose ranged from 50 mg (Koubeissi Torres-Cano et al., 2011). The longest duration of SE—
et al., 2011; median 100 mg [range 50–100 mg]; four 10 weeks (CSE evolving in a NCSE) until LCM was used—
NCSE) to 400 mg (Albers et al., 2011: four had focal SE). was described by Shiloh-Malawsky et al. (2011). SE
In the remaining four studies, the initial dose of LCM varied appeared to resolve with LCM treatment. The shortest dura-
from 50 mg (Rantsch et al., 2011: 50–100 mg; eight NCSE, tion of SE before LCM was 1.5 h (Kellinghaus et al., 2009).
two focal SE) to a maximum of 400 mg (Cherry et al., All reported patients were responders.
2012; mean initial dose 180.8 [range 100–400]; Mnatsaka-
nyan et al., 2012: 200 mg LCM/12 h). In the study with Case series
100% nonresponders to LCM (six NCSE, three CSE), the In group I, duration of SE until LCM therapy was
median initial dose was 200 mg (range 100–300 mg) reported in four of seven case series (Goodwin et al., 2011;
(Goodwin et al., 2011). Overall, the initial dose of LCM Koubeissi et al., 2011; Cherry et al., 2012; Jain & Harvey,
responders (Albers et al., 2011; Koubeissi et al., 2011; Jain 2012). Goodwin et al. had a median time span of 48 h
& Harvey, 2012; Mnatsakanyan et al., 2012) versus nonre- (range 0–336 h); Koubeissi et al., 3–50 h. Cherry et al.
sponders (Goodwin et al., 2011; Rantsch et al., 2011; had a median duration of 39.5 h (range 6.5–74 h), whereas
Cherry et al., 2012) did not differ. Jain & Harvey reported 8–29 h duration of SE prior to
In group II, there were 39% (27/70) NCSE, 37% (26/70) LCM.
focal SE, and 24% (17/70) CSE. The responder rate varied In group II, duration of SE until LCM therapy was
from 44% (17/39) (Kellinghaus et al., 2011) to 81% (25/31) reported only in one case series (Kellinghaus et al.,
(H€ofler et al., 2011) of patients with SE. In the collaborative 2011). Patients had a median latency of 30 h (range 0.5–
study of Germany, Austria, and Switzerland (Kellinghaus 1,440 h) in SE before they received LCM. However, both
et al., 2011), the median initial dose of LCM was 400 mg studies in group II could demonstrate a clear order effect:
(range 200–400 mg). In our groups study, the median initial if LCM was administered early, it was successful in 60%
dose of LCM was 200 mg (range 200–400 mg). In both of cases, whereas the success rate was only 20% in more
studies, median use of LCM was as third drug (H€ofler et al., refractory patients (H€ofler et al., 2011; Kellinghaus et al.,
2011; Kellinghaus et al., 2011). 2011).

Epilepsia, 54(3):393–404, 2013


doi: 10.1111/epi.12058
403
IV Lacosamide in Seizure Emergencies

have a potential as second-line alternative to standard


Definition of Resistant SE anticonvulsants in treatment of established SE, in case of
All contemporary protocols use a staged approach for contraindications to phenytoin or valproic acid. The avail-
treatment of SE (Trinka, 2007; Kurthen et al., 2008; Shor- able evidence of efficacy is limited and randomized con-
von et al., 2008; Meierkord et al., 2010). Treatment in stage trolled data are needed.
1 (“early SE”) includes benzodiazepines. After the failure of
benzodiazepines, the next level will be stage 2 (“established
SE”), the most commonly used therapies being IV AEDs
Acknowledgments
such as PHE, PB, or VPA. If seizures continue despite this The authors thank Alison Terry for editorial work.
treatment, the SE is classed as stage 3 (“refractory SE”), at
which point general anesthesia is recommended (Trinka, Disclosure
2007; Shorvon et al., 2008).
J. H€ofler has received speakers honoraria from UCB, and travel grants
from UCB, Eisai, and Gerot E. Trinka has acted as a paid consultant to
Single case reports Eisai, Biogen Idec, Medtronics, Bial, and UCB. He has received research
All reported 10 patients with SE treated with LCM ini- funding from UCB, Biogen-Idec, Sanofi-Aventis, Red-Bull and speakers’
tially received benzodiazepines (Kellinghaus et al., 2009; honoraria from Bial, Cyberonics, Desitin Pharma, Eisai, Gerot, B€ohringer-
Ingelheim, Sanofi, Medis, and UCB. We confirm that we have read the
Tilz et al., 2010; Turpin-Fenoll et al., 2010; Chen et al., Journals position on issues involved in ethical publication and affirm that
2011; Granda-Mendez et al., 2011; Krause et al., 2011; this report is consistent with those guidelines.
LaRoche & Shivdat-Nanhoe, 2011; Shiloh-Malawsky et al.,
2011; Torres-Cano et al., 2011). References
Case series Albers JM, M€oddel G, Dittrich R, Steidl C, Suntrup S, Ringelstein EB,
Dziewas R. (2011) Intravenous lacosamide – an effective add-on
In 16% (13/126) of episodes of SE, benzodiazepines were treatment of refractory status epilepticus. Seizure 20:428–430.
not given before initiation of LCM therapy (Albers et al., Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich S,
2011; H€ ofler et al., 2011; Kellinghaus et al., 2011; Rantsch Gottwald MD, ONeil N, Neuhaus JM, Segal MR, Lowenstein DH.
(2001) A comparison of lorazepam, diazepam, and placebo for the
et al., 2011; Cherry et al., 2012); in 20% (16/126) of epi- treatment of out-of-hospital status epilepticus. N Engl J Med 345:
sodes of SE, LCM was given immediately after benzodiaze- 631–637. Erratum in: N Engl J Med 2001;345:1860.
pines (Kellinghaus et al., 2009; H€ ofler et al., 2011). Andurkar SV, Stables JP, Kohn H. (1999) The anticonvulsant activities of
N-benzyl 3-methoxypropionamides. Bioorg Med Chem 7:2381–2389.
Because dose information was missing regarding AEDs Barton ME, Klein BD, Wolf HH, White HS. (2001) Pharmacological
received before LCM, a detailed analysis of prior treatment characterization of the 6 Hz psychomotor seizure model of partial
was not possible. epilepsy. Epilepsy Res 47:217–227.
Ben-Menachem E, Biton V, Jatuzis D, Abou-Khalil B, Doty P, Rudd GD.
(2007) Efficacy and safety of oral lacosamide as adjunctive therapy in
adults with partial-onset seizures. Epilepsia 48:1308–1317.
Conclusion Beyreuther BK, Heers C, Krebsf€anger N, Scharfenecker U, St€ohr T. (2007)
Lacosamide: a review of preclinical properties. CNS Drug Rev 13:
In summary, 136 episodes of SE treated with LCM were 21–42.
reported. The type of SE was an NCSE in 50% (69/136 epi- Biton V, Rosenfeld WE, Whitesides J, Fountain NB, Vaiciene N, Rudd
sodes), focal in 31% (40/135), and convulsive in 19% (26/ GD. (2008) Intravenous lacosamide as replacement for oral lacosamide
in patients with partial-onset seizures. Epilepsia 49:418–424.
135). Fifty-one percent (70/136) had preexisting epilepsy. Borowicz KK, Gasior M, Kleinrok Z, Czuczwar SJ. (1997) Influence
The overall success rate was 56% of SE (76/136). AEs were of isradipine, niguldipine and dantrolene on the anticonvulsive
reported in 25% (34/136) of patients: sedation in 25 cases, 1 action of conventional antiepileptics in mice. Eur J Pharmacol 323:
45–51.
patient with possible angioedema, 2 with allergic skin reac- Brophy GM, Bell R, Claassen J, Alldredge B, Bleck TP, Glauser T,
tion, 4 with hypotension, and 1 with pruritus. One patient Laroche SM, Riviello JJ Jr, Shutter L, Sperling MR, Treiman DM,
developed a third-degree AV block and paroxysmal asystole Vespa PM; Neurocritical Care Society Status Epilepticus Guideline
Writing Committee. (2012) Guidelines for the evaluation and
(Krause et al., 2011). Overall, the rate of AEs was low. management of status epilepticus. Neurocrit Care Apr 24; doi:
None of the published studies had a prospective design, 10.1007/s12028-012-9695-z. [Epub ahead of print].
and all were uncontrolled. Study design, heterogeneity of Cawello W, Kropeit D, Schiltmeyer B, Hammes W, Horstmann R. (2004)
Food does not affect the pharmacokinetics of SPM 927. Epilepsia 45
population, and variations in the definition of SE may have (Suppl. 7):307 [Poster 2.342].
contributed to bias, in addition to the publication bias. The Chen LL, Haneef Z, Dorsch A, Keselman I, Stern JM. (2011) Successful
varying—and sometimes missing—definition of response treatment of refractory simple motor status epilepticus with lacosamide
and levetiracetam. Seizure 20:263–265.
to IV LCM also weakens the conclusions. Although LCM is Cherry S, Judd L, Muniz JC, Elzawahry H, LaRoche S. (2012) Safety and
a promising drug in the treatment of acute seizures and SE, efficacy of lacosamide in the intensive care unit. Neurocrit Care 16:
several issues remain to be clarified: What is the right dose? 294–298.
Cross SA, Curran MP. (2009) Lacosamide: in partial-onset seizures. Drugs
What is the maximum tolerated rate of infusion? How does 69:449–459.
LCM penetrate across the blood–brain barrier in patients Doty P, Rudd GD, Stoehr T, Thomas D. (2007) Lacosamide.
with SE? Based on the current limited evidence LCM may Neurotherapeutics 4:145–148.

Epilepsia, 54(3):393–404, 2013


doi: 10.1111/epi.12058
404
J. H€
ofler and E. Trinka

Duncan GE, Kohn H. (2005) The novel antiepileptic drug lacosamide Leppik IE, Derivan AT, Homan RW, Walker J, Ramsay RE, Patrick B.
blocks behavioural and brain metabolic manifestations of seizure (1983) Double-blind study of lorazepam and diazepam in status
activity in the 6 Hz psychomotor seizure model. Epilepsy Res 67:81–87. epilepticus. JAMA 249:1452–1454.
Errington AC, Coyne L, Stohr T, Selve N, Lees G. (2006) Seeking a Meierkord H, Boon P, Engelsen B, G€ocke K, Shorvon S, Tinuper P,
mechanism of action for the novel anticonvulsant lacosamide. Holtkamp M; European Federation of Neurological Societies. (2010)
Neuropharmacology 50:1016–1029. EFNS guideline on the management of status epilepticus in adults. Eur
Fountain NB, Krauss G, Isojarvi J, Dilley D, Doty P, Rudd GD. (2011) J Neurol 17:348–355.
Followed by oral maintenance as adjunctive therapy in patients with Mnatsakanyan L, Chung JM, Tsimerinov EI, Eliashiv DS. (2012)
partial-onset seizures. Epilepsia 52(Suppl. 8):72–85 [Poster 24]. Intravenous lacosamide in refractory nonconvulsive status epilepticus.
Goodwin H, Hinson HE, Shermock KM, Karanjia N, Lewin JJ 3rd. (2011) Seizure 21:198–201.
The use of lacosamide in refractory status epilepticus. Neurocrit Care Parkerson KA, Reinsberger C, Chou SH, Dworetzky BA, Lee JW. (2011)
14:348–353. Lacosamide in the treatment of acute recurrent seizures and periodic
Granda-Mendez J, Toledo M, Flores-Flores A, Santamarina E, Sarria S, epileptiform patterns in critically ill patients. Epilepsy Behav 20:
Salas-Puig J. (2011) Intravenous lacosamide in monotherapy in a case 48–51.
of complex partial epileptic status. Rev Neurol 53:61–63. Rantsch K, Walter U, Wittstock M, Benecke R, R€osche J. (2011) Efficacy
H€ofler J, Unterberger I, Dobesberger J, Kuchukhidze G, Walser G, Trinka of intravenous lacosamide in refractory nonconvulsive status
E. (2011) Intravenous lacosamide in status epilepticus and seizure epilepticus and simple partial status epilepticus. Seizure 20:529–532.
clusters. Epilepsia 52:148–152. Shiloh-Malawsky Y, Fan Z, Greenwood R, Tennison M. (2011) Successful
Horstmann R, Bonn R, Cawello W, Doty P, Rudd D. (2002) Basic clinical treatment of childhood prolonged refractory status epilepticus with
pharmacological investigations of the new antiepileptic drug SPM 927. lacosamide. Seizure 20:586–588.
Epilepsia 43(Suppl. 7): 188 [Poster 2.174]. Shorvon S, Baulac M, Cross H, Trinka E, Walker M; Task Force on Status
Hovinga CA. (2003) SPM-927 (Schwarz Pharma). IDrugs 6:479–485. Epilepticus of the ILAE Commission for European Affairs. (2008) The
Jain V, Harvey SA. (2012) Treatment of refractory tonic status epilepticus drug treatment of status epilepticus in Europe: consensus document
with intravenous lacosamide. Epilepsia 53:761–762. from a workshop at the first London Colloquium on Status Epilepticus.
Jatuzis D, Biton V, Ben-Menachem E, Abou-Khalil B, Doty P, Rudd GD; Epilepsia 49:1277–1285.
SP667 Study Group. (2005) Evaluation of the effect of oral lacosamide Silbergleit R, Durkalski V, Lowenstein D, Conwit R, Pancioli A, Palesch
on concomitant AED plasma concentrations in patients with partial Y, Barsan W; NETT Investigators. (2012) Intramuscular versus
seizures. Epilepsia 46(Suppl. 8):170 [Poster 2.236]. intravenous therapy for prehospital status epilepticus. N Engl J Med
Kellinghaus C, Berning S, Besselmann M. (2009) Intravenous lacosamide 366:591–600.
as successful treatment for nonconvulsive status epilepticus after St€ohr T, Kupferberg HJ, Stables JP, Choi D, Harris RH, Kohn H, Walton N,
failure of first-line therapy. Epilepsy Behav 14:429–431. White HS. (2007) Lacosamide, a novel anti-convulsant drug, shows
Kellinghaus C, Berning S, Immisch I, Larch J, Rosenow F, Rossetti AO, efficacy with a wide safety margin in rodent models for epilepsy.
Tilz C, Trinka E. (2011) Intravenous lacosamide for treatment of status Epilepsy Res 74:147–154.
epilepticus. Acta Neurol Scand 123:137–141. Epub 2010 Sep 26. Swinyard EA, Brown WC, Goodman LS. (1952) Comparative assays of
Koubeissi MZ, Mayor CL, Estephan B, Rashid S, Azar NJ. (2011) Efficacy antiepileptic drugs in mice and rats. J Pharmacol Exp Ther 106:
and safety of intravenous lacosamide in refractory nonconvulsive status 319–330.
epilepticus. Acta Neurol Scand 123:142–146. Tilz C, Resch R, Hofer T, Eggers C. (2010) Successful treatment for
Krause LU, Brodowski KO, Kellinghaus C. (2011) Atrioventricular block refractory convulsive status epilepticus by non-parenteral lacosamide.
following lacosamide intoxication. Epilepsy Behav 20:725–727. Epilepsia 51:316–317.
Krauss G, Ben-Menachem E, Mameniskiene R, Vaiciene-Magistris N, Torres-Cano N, Giner Bayarri P, Chilet-Chilet MR, Ruiz-Marquez L,
Brock M, Whitesides JG, Johnson ME; SP757 Study Group. (2010) Rodrigo-Sanbartolome A, Mazzillo-Ricaurte A, Moliner-Ibanez J.
Intravenous lacosamide as short-term replacement for oral lacosamide (2011) Refractory nonconvulsive status epilepticus. Rev Neurol 53:
in partial-onset seizures. Epilepsia 51:951–957. 255–256.
Kropeit D, Schildtmeyer B, Cawello W, Hammes W, Horstmann R. (2004) Treiman DM, Meyers PD, Walton NY, Collins JF, Colling C, Rowan AJ,
Bioequivalence of short-time infusions compared to oral administration Handforth A, Faught E, Calabrese VP, Uthman BM, Ramsay RE,
of SPM 927. Epilepsia 45(Suppl. 7):123–124 [Poster 1.323]. Mamdani MB. (1998) A comparison of four treatments for generalized
Kurthen M, Janzen R, Lerche H, Schmidt D, Schneider D, Stefan H, convulsive status epilepticus. Veterans Affairs Status Epilepticus
Steinhoff B, Trinka E, Wieser H. (2008) Status epilepticus im Cooperative Study Group. N Engl J Med 339:792–798.
Erwachsenenalter. In Diener HC, Putzki N, Berlit P, Deuschl G, Elger Trinka E. (2007) Notf€alle im Bereich der Epilepsien – Was tun und was
C, Gold R, Hacke W, Hufschmidt A, Mattle H, Meier U, Oertel WH, lassen? Nervenheilkunde 26:981–992.
Reichmann H, Schmutzhard E, Wallesch CW, Weller M (Eds) Turpin-Fenoll L, Millan-Pascual J, Navarro-Munoz S, del Saz-Saucedo P,
Leitlinien f€ur Diagnostik und Therapie in der Neurologie. 3. Auflage. Rueda-Medina I. (2010) Uso de lacosamida oral en un paciente con
Thieme, Stuttgart, pp. 17–28. estado epileptico parcial refractario. Rev Neurol 50:603–606.
LaRoche SM, Shivdat-Nanhoe RS. (2011) Subacute encephalopathy and Wasterlain CG, St€ohr T, Matagne A. (2011) The acute and chronic effects
seizures in alcoholics (SESA) presenting with non-convulsive status of the novel anticonvulsant lacosamide in an experimental model of
epilepticus. Seizure 20:505–508. status epilepticus. Epilepsy Res 94:10–17.

Epilepsia, 54(3):393–404, 2013


doi: 10.1111/epi.12058

You might also like