Adjunctive lacosamide for partial-onset seizures ... - Wiley Online Library

30 downloads 1878 Views 494KB Size Report
Oct 7, 2008 - benzyl-3-methoxypropionamide, is a novel functionalized amino acid ..... lacosamide 400 mg/day dose group at 7.4 lg/ml was approximately ...
Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

FULL-LENGTH ORIGINAL RESEARCH

Adjunctive lacosamide for partial-onset seizures: Efficacy and safety results from a randomized controlled trial *Pe´ter Hala´sz, yReetta Ka¨lvia¨inen, zMaria Mazurkiewicz-Beldzin´ska, xFelix Rosenow, {Pamela Doty, {David Hebert, and {Timothy Sullivan on behalf of the SP755 Study Group *National Institute of Psychiatry and Neurology, Budapest, Hungary; yKuopio Epilepsy Center, Kuopio University Hospital, Kuopio, Finland; zMedical University of Gdansk, Gdansk, Poland; xPhilipps University Marburg, Marburg, Germany; and {Schwarz Biosciences, Inc., a member of the UCB Group, Research Triangle Park, North Carolina, U.S.A.

SUMMARY Purpose: To evaluate the efficacy and safety of lacosamide (200 and 400 mg/day) when added to one to three concomitant antiepileptic drugs (AEDs) in patients with uncontrolled partial-onset seizures. Methods: This multicenter, double-blind, placebocontrolled trial randomized patients (age 16–70 years) with partial-onset seizures with or without secondary generalization to placebo, lacosamide 200, or lacosamide 400 mg/day. The trial consisted of an 8-week baseline, a 4-week titration, and a 12-week maintenance period. Results: Four hundred eighty-five patients were randomized and received trial medication. Among these, 87% were taking two or more concomitant AEDs. Median percent reduction in seizure frequency per 28 days from baseline to maintenance period (intent-to-treat, ITT) was 20.5% for placebo, 35.3% for lacosamide 200 mg/day (p = 0.02), and 36.4% for 400 mg/day (p = 0.03). In the per

The goal of pharmacologic therapy with antiepileptic drugs (AEDs) is to reduce the frequency of seizures and achieve a seizure-free state with minimal side effects (Shneker & Fountain, 2003). However, only half of patients achieve seizure freedom with the first administered AED, and about 60–70% of patients become seizure-free with the first or second AED tried (Brodie & Accepted October 7, 2008; Early View publication January 22, 2009. Address correspondence to Pter Halsz, M.D., Ph.D., 1026 Lotz K. u. 18 Budapest, Hungary. E-mail: [email protected] ClinicalTrials.gov Identifier: NCT00220415 Wiley Periodicals, Inc. ª 2009 International League Against Epilepsy

protocol population, the reductions were 35.3% for lacosamide 200 mg/day (p = 0.04) and 44.9% for 400 mg/day (p = 0.01) compared to placebo (25.4%). The 50% responder rate for lacosamide 400 mg/day (40.5%) was significant (p = 0.01) over placebo (25.8%), but was not for 200 mg/day (35.0%). In the per protocol population, the 50% responder rate for lacosamide 400 mg/day (46.3%) was significant (p < 0.01) compared with the placebo responder rate (27.5%). Dose-related adverse events (AEs) included dizziness, nausea, and vomiting. Clinically relevant changes in the mean plasma concentrations of commonly used AEDs were not observed. Discussion: Results of this trial demonstrated the efficacy and tolerability of adjunctive lacosamide 200 and 400 mg/day and support that lacosamide may be an advantageous option for the treatment of partial-onset seizures in patients with epilepsy. KEY WORDS: Epilepsy, Partial-onset seizures, Antiepileptic drugs, Lacosamide, Randomized controlled trial.

Kwan, 2002). Approximately 30% of patients treated with available AEDs continue to experience uncontrolled seizures (Kwan & Brodie, 2000). Despite the availability of a new generation of AEDs, many patients are unable to achieve seizure control, whereas others experience intolerable side effects; therefore, the need remains for the development of effective and well-tolerated new AEDs (Bialer, 2002). Lacosamide, the R-enantiomer of 2-acetamido-Nbenzyl-3-methoxypropionamide, is a novel functionalized amino acid that was synthesized as a potential anticonvulsant compound (Andurkar et al., 1999; Hovinga, 2003). Lacosamide was effective in a range of animal models of

443

444 P. Hala´sz et al. epilepsy (Brandt et al., 2006; Beyreuther et al., 2007; Stçhr et al., 2007). Based on in vitro studies, lacosamide appears to have a dual mode of action. It selectively enhances slow inactivation of voltage-gated sodium channels without affecting fast inactivation (Errington et al., 2006, 2008; Sheets et al., 2008). In addition, lacosamide binds to collapsin-response mediator protein 2 (CRMP-2), a protein that is involved in neuroplastic processes such as neuronal outgrowth or the modulation of N-methyl-daspartate (NMDA) receptor subunit NR2B (Beyreuther et al., 2007; Beyreuther et al., 2006). Oral lacosamide is absorbed completely and rapidly from the gastrointestinal tract with negligible first-pass metabolism (Thomas et al., 2007). Administration with food does not alter the rate or extent of lacosamide absorption (Cawello et al., 2004; Doty et al., 2007). Lacosamide plasma concentrations have shown a proportional relationship for oral doses ranging from 100–800 mg/day (Horstmann et al., 2002). Peak plasma levels occur within 1–4 h of an oral lacosamide dose with a 13-h elimination half-life that is independent of dose or duration of administration (Horstmann et al., 2002; Hovinga, 2003; Bialer et al., 2007). Lacosamide is eliminated primarily by renal excretion and biotransformation. Approximately 40% of the drug is eliminated unchanged in the urine. Lacosamide has demonstrated low plasma protein binding (£15%) as well as a low potential for pharmacokinetic drug–drug interactions (Thomas et al., 2007). No pharmacokinetic interactions were evident between lacosamide and carbamazepine, valproic acid, metformin, digoxin, oral contraceptives (ethinyl estradiol/levonorgestrel), or omeprazole in dedicated studies (Horstmann et al., 2003; Kropeit et al., 2006; Doty et al., 2007). Lacosamide has shown no or low potential to inhibit or to induce CYP isoforms (Kropeit et al., 2006). A recent multicenter, randomized, double-blind, placebo-controlled trial evaluated lacosamide 200 mg/ day, 400 mg/day, and 600 mg/day when added to one or two AEDs in 418 adult patients with uncontrolled partialonset seizures (SP667; Ben-Menachem et al., 2007). Reductions in seizure frequency and the proportion of patients with at least a 50% reduction in seizure frequency (50% responder rate) with lacosamide 400 and 600 mg/day were statistically significant compared with placebo in the primary intent-to-treat (ITT) population. However, in the per protocol population, reductions in seizure frequency and in the 50% responder rate were significant compared with placebo with all three doses of lacosamide (200, 400, and 600 mg/day). Adverse events (AEs) that appeared dose-related included dizziness, nausea, fatigue, ataxia, abnormal vision, diplopia, and nystagmus. In addition, lacosamide did not affect mean plasma concentrations of concomitantly administered AEDs. The primary objective of this trial (SP755) was to evaluate the efficacy and safety of lacosamide doses of Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

200 mg/day and 400 mg/day when added to one to three marketed concomitant AEDs in patients with uncontrolled partial-onset seizures, with or without additional vagus nerve stimulation (VNS). Dose proportionality of lacosamide and the stability of concomitant AED concentrations were assessed by determination of steady-state plasma concentrations of these drugs. This trial was designed to complement the previous trial (SP667; Ben-Menachem et al., 2007) in order to establish the efficacy of adjunctive lacosamide.

Materials and Methods This international, multicenter, double-blind, randomized, placebo-controlled, parallel group trial (SP755; NCT00220415) was conducted between June 2004 and January 2006 at 75 enrolling sites in Australia, Croatia, Czech Republic, Finland, France, Germany, Hungary, Lithuania, Poland, Russia, Spain, Sweden, and the United Kingdom. The trial was conducted in accordance with the International Conference on Harmonisation (ICH) Good Clinical Practice (GCP) guidelines, the Declaration of Helsinki, and the local laws of the countries involved, as well as the ICH Tripartite Guideline, Guideline for GCP, January 1997. The trial protocol, amendments, and informed consents were reviewed by the applicable national regulatory authority in each country and relevant ethics committees for each site. All patients or their legal representatives gave written informed consent before trial participation. Patients The study included patients between the ages of 16 and 70 years with a diagnosis of partial-onset seizures with or without secondary generalizations (ILAE, 1981). Patients must have had an electroencephalography (EEG) study and magnetic resonance imaging (MRI) or computed tomography (CT) scan exhibiting results consistent with this diagnosis and without evidence of progressive structural lesion in the central nervous system (CNS) or progressive encephalopathy. Enrollment criteria required patients to have had partial-onset seizures for at least the preceding 2 years despite prior therapy with at least two AEDs. Patients were to have had at least four partial-onset seizures per 28 days on average, with no seizure-free period longer than 21 days during the 8-week period before enrollment as well as during the 8-week baseline period. In addition, patients were to have been taking a stable dosage regimen of one to three AEDs, with or without VNS, in the 4 weeks before enrollment and during the baseline period. Female patients were excluded from participation if they were pregnant, breast-feeding, or of childbearing potential and not using approved methods of contraception. Patients were excluded if they had previously

445 Lacosamide for Partial-Onset Seizures received lacosamide or had participated in any other investigational trial within the last 2 months. Patients were also excluded if they had a history of chronic alcohol or drug abuse; any medical condition that might jeopardize the patient’s health or compromise the patient’s ability to participate in this trial; liver function test results of at least two times the upper limit of normal; creatinine clearance 99%) and approximately one-half were female. Across all treatment groups, the mean number of years since diagnosis of epilepsy was 22.3 € 12.56 years. In the ITT population, 69% of patients had tried at least four AEDs in their lifetimes, with 37% having tried at least seven (Table 2). A total of 87% of patients were taking at least two AEDs, with 37% of these taking three AEDs in addition to their assigned trial medication (Table 2). Despite treatment with one to three concomitant AEDs, the median seizure frequency across all treatment groups ranged from 9.9–11.5 seizures per 28 days during the 8-week baseline period (Table 2). The five concomitant AEDs taken most commonly by patients enrolled in this trial were carbamazepine (47.8%), valproate (32.8%), lamotrigine (30.5%), topiramate (28.2%), and levetiracetam (19.8%). Efficacy The median percent reduction in seizure frequency per 28 days from baseline to the maintenance period was 20.5% for placebo, 35.3% for lacosamide 200 mg/day, and 36.4% for lacosamide 400 mg/day (ITT; Fig. 3).

Figure 2. Patient disposition. Epilepsia ILAE Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

448 P. Hala´sz et al. Table 1. Demographic characteristics of patients receiving trial medication (safety set) Lacosamide Characteristic Age, year Mean ± SD Range Sex, n (%) Male Female Race, n (%) Caucasian Black Asian Weight, kg (mean ± SD) BMI, kg/m2 (mean ± SD) Mean time since diagnosis, year (mean ± SD) Seizure classification, n (%) Simple partial-onset seizures Complex partial-onset seizures Partial-onset seizures with secondary generalization

Placebo (n = 163)

200 mg/day (n = 163)

400 mg/day (n = 159)

Total (n = 485)

38.5 ± 10.93 17–63

36.9 ± 11.70 16–66

37.9 ± 12.96 16–70

37.8 ± 11.88 16–70

91 (55.8) 72 (44.2)

90 (55.2) 73 (44.8)

69 (43.4) 90 (56.6)

250 (51.5) 235 (48.5)

162 (99.4) 0 (0) 1 (0.6) 74.7 ± 17.06 25.9 ± 5.01 21.1 ± 12.23

162 (99.4) 1 (0.6) 0 (0) 74.9 ± 16.93 25.2 ± 4.79 22.9 ± 12.30

157 (98.7) 0 (0) 2 (1.3) 72.2 ± 16.90 25.3 ± 5.09 22.8 ± 13.15

481 (99.2) 1 (0.2) 3 (0.6) 74.0 ± 16.97 25.4 ± 4.96 22.3 ± 12.56

61 (37.4) 138 (84.7) 130 (79.8)

67 (41.1) 142 (87.1) 125 (76.7)

58 (36.5) 146 (91.8) 127 (79.9)

186 (38.4) 426 (87.8) 382 (78.8)

BMI, body mass index; SD, standard deviation.

Table 2. ITT population characteristics Lacosamide Characteristic Lifetime AEDs, n (%) 1–3 AEDs 4–6 AEDs ‡7 AEDs Missing data Concomitant AEDs, n (%) 1 AED 2 AEDs 3 AEDs VNS use n (%) Median baseline partial-onset seizure frequency per 28 days

Placebo (n = 159)

200 mg/day (n = 160)

400 mg/day (n = 158)

Total (n = 477)

49 (30.8) 53 (33.3) 56 (35.2) 1 (0.6)

46 (28.8) 54 (33.8) 56 (35.0) 4 (2.5)

47 (29.7) 49 (31.0) 62 (39.2) 0

142 (29.8) 156 (32.7) 174 (36.5) 5 (1.0)

21 (13.2) 82 (51.6) 56 (35.2) 14 (8.8) 9.9

17 (10.6) 77 (48.1) 66 (41.3) 12 (7.5) 11.5

25 (15.8) 79 (50.0) 54 (34.2) 11 (7.0) 10.3

63 (13.2) 238 (49.9) 176 (36.9) 37 (7.8) –

AEDs, antiepileptic drugs; ITT, intent-to-treat; VNS, vagus nerve stimulation.

Based on the ANCOVA model used, statistically significant percent reductions over placebo in seizure frequency were observed for both the lacosamide 200 mg/day group [14.4%; 95% confidence interval (CI), 2.2–25.1; p = 0.02] and the 400 mg/day group (15.0%; CI, 1.4– 26.8; p = 0.03). In the per protocol population, the median percent reductions in seizure frequency per 28 days from baseline to the maintenance period in comparison with placebo (25.4%) were 35.3% for lacosamide 200 mg/day (p = 0.04) and 44.9% for lacosamide 400 mg/day (p = 0.01). The 50% responder rate for lacosamide 400 mg/day (40.5%) was statistically significant (p = 0.01) over plaEpilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

cebo (25.8%). Although not statistically significant (p = 0.07), the 50% responder rate for lacosamide 200 mg/day (35.0%) was numerically higher than placebo (ITT; Fig. 4). In the per protocol population, compared with placebo (27.5%), the 50% responder rates were 35.0% for lacosamide 200 mg/day (p = 0.19) and 46.3% for lacosamide 400 mg/day (p < 0.01). Among patients completing the maintenance period, 5 (3.6%) of 137 patients in the lacosamide 200 mg/day group and 3 (2.4%) of 123 patients in the lacosamide 400 mg/day group were seizure-free throughout the 12-week maintenance period compared with 3 (2.1%) of 143 in the placebo group. A statistically significant

449 Lacosamide for Partial-Onset Seizures

Figure 3. Median percent reduction in seizure frequency per 28 days from baseline to the maintenance period (intent-to-treat, ITT). Epilepsia ILAE

Figure 4. 50% responder rate from baseline to maintenance period (intent- to-treat, ITT). Epilepsia ILAE

increase of 5% in the percentage of seizure-free days over placebo during the maintenance period was observed for lacosamide 400 mg/day (p = 0.01; 95% CI 1.5, 8.5). Safety and tolerability The most common treatment-emergent AEs (TEAEs; ‡5% in any lacosamide treatment group) were dizziness, headache, diplopia, nausea, vertigo, fatigue, nasopharyngitis, coordination abnormal, and vomiting (Table 3). Most events were reported as being mild or moderate in intensity. The most clearly dose-related TEAEs included dizziness, nausea, and vomiting. Of note, the incidence of somnolence was low for patients randomized to lacosamide, at 4.3% in the lacosamide 200 mg/day group and 3.8% in the lacosamide 400 mg/day group, and similar to the occurrence among patients randomized to placebo (3.7%). Overall, 42 patients (8.7%) discontinued participation in the trial during the treatment period because of AEs: 8

patients (4.9%) randomized to placebo, 10 (6.1%) to lacosamide 200 mg/day, and 24 (15.1%) to 400 mg/day. Four patients randomized to receive lacosamide 200 mg/day experienced AEs leading to withdrawal during the titration phase prior to receiving lacosamide (i.e., while still taking placebo). AEs leading to discontinuation from the trial in at least 1% of patients across lacosamide treatment groups were diplopia (2.2%), vertigo (1.6%), vomiting (1.2%), and convulsion (1.2%). The overall percentage of patients experiencing serious AEs (SAEs) during the treatment period was greater in the lacosamide treatment groups compared to placebo (3.7%, 8.0%, and 9.4% of patients in the placebo, lacosamide 200 mg/day, and 400 mg/day treatment groups, respectively). Across all treatment groups, the most frequently reported SAEs were convulsion (one patient in each treatment group), epilepsy (reported term epileptic seizure; one patient in the placebo group and two in the lacosamide Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

450 P. Hala´sz et al. Table 3. Incidence of treatment-emergent adverse events occurring in at least 5% of patients in any treatment group during the treatment period (titration plus maintenance periods) Lacosamide

Adverse event Dizziness Headache Diplopia Nausea Vertigo Fatigue Nasopharyngitis Coordination abnormal Vomiting

Placebo (n = 163) n (%)

200 mg/day (n = 163) n (%)

400 mg/day (n = 159) n (%)

Total (n = 322) n (%)

8 (4.9) 12 (7.4) 2 (1.2) 2 (1.2) 3 (1.8) 6 (3.7) 6 (3.7) 1 (0.6)

17 (10.4) 18 (11.0) 13 (8.0) 9 (5.5) 11 (6.7) 8 (4.9) 8 (4.9) 7 (4.3)

25 (15.7) 13 (8.2) 16 (10.1) 13 (8.2) 10 (6.3) 10 (6.3) 10 (6.3) 10 (6.3)

42 (13.0) 31 (9.6) 29 (9.0) 22 (6.8) 21 (6.5) 18 (5.6) 18 (5.6) 17 (5.3)

3 (1.8)

5 (3.1)

9 (5.7)

14 (4.3)

200 mg/day treatment group), grand mal convulsion (two patients in the lacosamide 400 mg/day group), and psychotic disorder (two patients in the lacosamide 400 mg/ day group). All other treatment-emergent SAEs occurred in only one patient each. There were no deaths among patients participating in the trial. Comparison of on-treatment versus baseline ECG results did not demonstrate any tendency for lacosamide to prolong the QT/QTc interval or QRS duration. Lacosamide was associated with a dose-related increase in mean PR interval (4.6 ms at the end of maintenance for the 400 mg/day dose). This increase in PR interval did not affect the AE profile, as only one lacosamide patient had ECG PR prolongation reported as an AE. Comprehensive chemistry, hematology, and urinalysis laboratory evaluations as well as periodic physical and neurologic examinations and assessments of vital signs did not reveal any abnormalities that appeared to be associated with administration of lacosamide. No patient was reported to have an alanine aminotransferase (ALT) level of at least three times the upper limit of normal and a bilirubin value of at least 2 mg/dl concurrently. Lacosamide did not affect the body weight. The mean change from baseline to the end of the 12-week maintenance period was 0.0 kg for placebo, 0.2 kg for lacosamide 200 mg/day, and )0.2 kg for 400 mg/day. Pharmacokinetics Plasma concentrations of lacosamide showed dose proportionality. At the end of the 12-week maintenance period, the mean lacosamide plasma concentration for the lacosamide 400 mg/day dose group at 7.4 lg/ml was approximately twice the mean plasma concentrations for the 200 mg/day group at 3.8 lg/ml. Mean plasma concenEpilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

trations of lacosamide remained stable during the maintenance period. Mean plasma concentrations for the common concomitant AEDs generally were not affected by concomitant intake of 200 mg/day and 400 mg/day lacosamide, other than a decrease (4.2 € 7.53 lg/ml) in the mean concentration of MHD in the lacosamide 400 mg/day group (Table 4).

Discussion Lacosamide 200 mg/day and 400 mg/day significantly reduced seizure frequency in patients with uncontrolled partial-onset seizures when added to one to three concomitant AEDs. These results are encouraging given the characteristics of the population studied in this trial. Trial patients had been experiencing a median of 9.9– 11.5 seizures per 28 days at baseline, with 87% taking two to three other AEDs, including newer AEDs such as lamotrigine, topiramate, and levetiracetam. These patients had a mean duration of epilepsy of over 22 years, and more than one-third of these patients had tried at least seven AEDs in their lifetime. The secondary efficacy analyses in the per protocol population were consistent with the primary ITT analyses and showed a greater seizure reduction with the lacosamide 400 mg/day dose compared with placebo. Compared to placebo, lacosamide was generally well tolerated, and it did not produce clinically meaningful changes in plasma concentrations of commonly used concomitant AEDs. The apparent small decrease in the MHD of oxcarbazepine concentration in this trial was not observed in two other clinical trials with lacosamide in patients with partial-onset seizures (Ben-Menachem et al., 2007; Thomas et al., 2007). Results from this trial confirm and extend those from a previously completed randomized, placebo-controlled trial (SP667; Ben-Menachem et al., 2007) that evaluated the efficacy and safety of lacosamide as adjunctive therapy for patients with uncontrolled partial-onset seizures. In the earlier trial (Ben-Menachem et al., 2007) as well as in the present trial, lacosamide 400 mg/day produced a significant reduction in seizure frequency and achieved a significantly higher 50% responder rate compared with placebo. The 50% responder rate for lacosamide 200 mg/day (35.0%) was numerically greater than placebo, similar to the 200 mg/day responder rate (33%) in the SP667 trial, and was within the range reported for marketed AEDs (14–45 %; Cramer et al., 2001). Comparative trials between old and new AEDs have suggested that newer AEDs might be better tolerated than older ones, with a lower incidence of side effects (Sander, 1998; Sander & Bell, 2004). In the present trial, the incidence of TEAEs was generally low and most events were considered mild or moderate in intensity. The most

451 Lacosamide for Partial-Onset Seizures Table 4. Mean change from baseline to the end of maintenance in plasma concentration of concomitant antiepileptic drugs Lacosamide (n) Antiepileptic drug Carbamazepine, lg/ml Baseline Change at end of maintenance Carbamazepine epoxide, lg/ml Baseline Change at end of maintenance Valproic acid, lg/ml Baseline Change at end of maintenance Lamotrigine, lg/ml Baseline Change at end of maintenance Topiramate, lg/ml Baseline Change at end of maintenance Levetiracetam, lg/ml Baseline Change at end of maintenance MHD, lg/ml Baseline Change at end of maintenance Clonazepam, ng/ml Baseline Change at end of maintenance Phenytoin, lg/ml Baseline Change at end of maintenance

Placebo (n)

200 mg/day

400 mg/day

8.0 ± 2.82 (71) 0.5 ± 2.08 (61)

8.0 ± 2.47 (85) 0.1 ± 2.31 (72)

8.0 ± 3.05 (72) 0.5 ± 1.91 (52)

1.7 ± 1.12 (71) 0.1 ± 0.92 (61)

1.7 ± 0.97 (85) 0.1 ± 0.93 (72)

1.6 ± 0.77 (72) 0.2 ± 0.53 (52)

58.0 ± 21.19 (58) 2.8 ± 26.41 (53)

64.4 ± 30.73 (50) )2.2 ± 19.54 (43)

71.1 ± 28.97 (50) )4.5 ± 20.45 (41)

6.7 ± 3.79 (51) )0.1 ± 1.48 (43)

6.7 ± 4.38 (44) )0.1 ± 1.68 (34)

6.6 ± 4.22 (51) 0.0 ± 1.57 (35)

7.0 ± 5.15 (43) )0.6 ± 3.09 (39)

6.3 ± 3.85 (48) 0.0 ± 3.78 (42)

7.2 ± 4.81 (43) 0.1 ± 1.74 (34)

33.8 ± 21.70 (29) 3.6 ± 12.95 (24)

41.0 ± 21.74 (29) )0.3 ± 17.31 (26)

33.7 ± 17.34 (37) 0.2 ± 10.32 (28)

25.4 ± 11.99 (26) )0.8 ± 8.22 (24)

26.4 ± 12.71 (26) )0.4 ± 6.48 (20)

28.3 ± 16.56 (23) )4.2 ± 7.53 (19)

20.8 ± 20.16 (19) 2.3 ± 21.92 (16)

23.0 ± 17.45 (19) 1.2 ± 3.93 (13)

25.1 ± 24.21 (15) )1.0 ± 4.41 (13)

11.5 ± 8.30 (15) 2.4 ± 4.17 (14)

8.5 ± 5.88 (13) 1.6 ± 1.61 (11)

11.9 ± 9.92 (11) )0.5 ± 3.37 (11)

Baseline, mean baseline value ± SD; change at end of maintenance, mean change from baseline value to the end of maintenance; MHD, monohydroxy derivative of oxcarbazepine.

commonly reported TEAEs were associated with the nervous and gastrointestinal systems. Adverse events that appeared to be dose related included dizziness, nausea, and vomiting. The incidence of somnolence in patients on lacosamide was low and similar to that in the placebo group. Discontinuations because of treatment-emergent adverse events were relatively low and were similar between placebo and lacosamide 200 mg/day. The higher rate of discontinuation among patients who received lacosamide 400 mg/day was primarily because of nervous system and gastrointestinal-related adverse events. The overall AE profile was comparable in quality and frequency to that reported earlier with lacosamide (SP667; Ben-Menachem et al., 2007). Based on the findings in this trial, the tolerability profile of lacosamide compares favorably with marketed AEDs (Marson et al., 1996, 1997; Bialer et al., 2002). Lacosamide generally had no clinically important effects on laboratory values, vital sign measurements, body weight, or ECG variables. Lacosamide produced a small, dose-related increase in PR interval that was not of clinical consequence.

Several characteristics of lacosamide contribute to its ease of use. Lacosamide dose escalation of 100 mg/day per week resulted in an effective dose (200 mg/day) being achieved after 1 week of treatment. Oral lacosamide is administered twice daily and has a low potential for pharmacokinetic drug interactions. Clinical pharmacokinetic studies in healthy patients have demonstrated that lacosamide does not affect the plasma concentrations of concomitant AEDs (valproic acid and carbamazepine) or other medications, including metformin, oral contraceptive agents (ethinyl estradiol and levonorgestrel), and digoxin (Kropeit et al., 2006; Bialer et al., 2007; Thomas et al., 2007). The development of an intravenous formulation of lacosamide as short-term replacement for oral lacosamide further contributes to the ease of use of lacosamide (Krauss et al., 2007; Biton et al., 2008). For new AEDs currently under development, important characteristics include demonstrated fewer side effects, improved ease of use, lack of drug interactions, and ability to prevent epileptogenesis or potential for disease modification, as well as broad use in other CNS disorders (Bialer, 2002). Results of the current trial and other trials Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

452 P. Hala´sz et al. indicate that lacosamide demonstrates most of these characteristics. Although results from preclinical studies indicate that lacosamide may have the potential to retard kindling-induced epileptogenesis, further research is needed to evaluate whether lacosamide possesses antiepileptogenic or disease-modifying effects (Brandt et al., 2006). Results from this trial confirm the efficacy of lacosamide 200 mg/day or 400 mg/day, as adjunctive treatment for partial-onset seizures in patients with epilepsy. A high proportion of patients with uncontrolled partial-onset seizures responded well to treatment with adjunctive lacosamide. Lacosamide was generally well tolerated. With twice-daily dosing, low potential for drug–drug interactions, demonstrated efficacy, a favorable tolerability profile, and the potential availability of an intravenous formulation, adjunctive lacosamide may provide an advantageous treatment option for patients with partial-onset seizures.

Acknowledgments The authors wish to acknowledge the contribution made by all of the members of the SP755 Study Group who have contributed materially to the contents of this manuscript by their participation in taking care of patients according to the approved protocol and contributing to the total data generated for this trial. Australia: S. Berkovic, D. Reutens (Victoria); M. Kiley (Adelaide); T. O’Brien (Parkville); M. Robinson (Woodville); G. Schapel (Maroochydore); E. Somerville (Randwick) Croatia: T. Babic, V. Demarin, S. Hajnsek, Z. Poljakovic (Zagreb). Czech Republic: M. Bar (Ostrava); J. Buresova (Olomouc); H. Hojdikova (Hradec Kralove); P. Marusic (Prague); E. Nespor, J. Zarubova (Prague); I. Rektor (Brno); H. Vacovska (Plzen). Finland: T. Kernen (Tampere); J. Korpelainen (Oulu); R. Klviinen (Kuopio); S. Lamusuo, T. Nylund (Helsinki). France: A. Crespel (Montpellier); P. Derambure (Lille); E. Hirsch (Strasbourg); P. Ryvlin (Bron); F. Semah (Paris); L. Valton (Toulouse). Germany: S. Arnold (Munich); H. Lerche (Ulm); H. Meencke, B. Schmitz (Berlin); F. Rosenow (Marburg); C. Siebold (Starnberg); H. Stefan (Erlangen); H. Straub (Bernau); F. Tergau (Gçttingen); K. Werhahn (Mainz). Hungary: A. Balogh, P. Halsz, A. Solyom (Budapest); A. Horvath (Szombathely Markusovsky); I. Kondakor (Ret); L. Vecsei (Szeged). Lithuania: M. Endziniene, V. Liesiene (Kaunas); R. Mameniskiene, A. Skaringa (Vilnius). Poland: A. Czlonkowska (Warsaw); W. Fryze, M. Mazurkiewicz-Beldzin´ska (Gdansk); J. Kochanowicz (Bialystok); R. Motyl (Krakow). Russia: A. Fedin, A. Guekht, V. Karlov, S. Kotov, A. Petrukhin, (Moscow); V. Guzeva, A. Skoromets (St. Petersburg). Spain: A. Gil-Nagel, M. Moro, J. Serratosa (Madrid); J. Llerda (Zaragoza); A. Molins (Girona); J. Sanchez (Granada). Sweden: E. Ben-Menachem (Gçteborg); P. Lindstrçm (Stockholm); B. Sçderfeldt (Linkoping). United Kingdom: O. Cockerell (Essex); P. Cooper (Salford); R. Roberts (Dundee); J. Sander (London); M. Brodie (Glasgow); I. Sawhney (Swansea); P. Smith (Cardiff). UCB Group Monheim, Germany, provided the trial supplies and UCB Group, Research Triangle Park, NC, USA, sponsored and funded the trial. Central laboratory facilities were provided by Quintiles Laboratories, Europe (West Lothian, Scotland, U.K.), for the European sites and their affiliate in Singapore, Malaysia, for the Australian sites. Plasma samples were analyzed by UCB, Monheim, Germany and plasma concentrations of selected concomitant antiepileptic drugs were determined by MedTox Laboratories, Inc. (St. Paul, MN, U.S.A.). Electrocardiograms for all sites were centrally read by Covance Central Diagnostics, Inc. (Reno, NV, U.S.A.). The authors would like to express their appreciation to Bengt Hoepken, PhD, and Maria Hackenson, clinical trial managers, UCB, and Jyoti Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x

Nandi MD, PhD, for writing and Allison Coppola, both of UCB for editorial support. We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines. Disclosure of conflicts of interest: P. Halsz and M. MazurkiewiczBeldzin´ska have no conflict of interest to disclose. R. Klviinen has acted as a paid consultant or received speakers honoraria from Algol Pharma, Cephalon, Eisai, GlaxoSmithKline, Johnson & Johnson, Janssen Cilag, Orion, Pfizer, Schwarz Pharma GmbH and UCB, Inc. F. Rosenow has received research grants, educational grants, and honoraria from UCB, Inc. and Schwarz Pharma GmbH. P. Doty and D. Hebert are employees and T. Sullivan is a former employee of Schwarz Biosciences, Inc., a member of the UCB group.

References Andurkar SV, Stables JP, Kohn H. (1999) The anticonvulsant activities of N-benzyl 3-methoxypropionamides. Bioorg Med Chem 7:2381– 2389. 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. Beyreuther B, Callizot N, Stçhr T. (2006) Antinociceptive efficacy of lacosamide in a rat model for painful diabetic neuropathy. Eur J Pharmacol 539:64–70. Beyreuther BK, Freitag J, Heers C, Krebsfanger N, Scharfenecker U, Stohr T. (2007) Lacosamide: a review of preclinical properties. CNS Drug Reviews 13:21–42. Bialer M. (2002) New antiepileptic drugs currently in clinical trials: is there a strategy in their development? Ther Drug Monit 24:85–90. Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Loiseau P, Perucca E. (2002) Progress report on new antiepileptic drugs: a summary of the Sixth Eilat Conference (EILAT VI). Epilepsy Res 51:31–71. Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Perucca E, Tomson T. (2007) Progress report on new antiepileptic drugs: a summary of the Eighth Eilat Conference (EILAT VIII). Epilepsy Res 73:1–52. Biton V, Rosenfeld WE, Whitesides J, Fountain NB, Vaiciene N, Rudd G. (2008) Intravenous lacosamide as replacement for oral lacosamide in patients with partial-onset seizures. Epilepsia 49:418–424. Brandt C, Heile A, Potschka H, Stoehr T, Loscher W. (2006) Effects of the novel antiepileptic drug lacosamide on the development of amygdala kindling in rats. Epilepsia 47:1803–1809. Brodie MJ, Kwan P. (2002) Staged approach to epilepsy management. Neurology 58(Suppl 5):S2–S8. Cawello W, Kropeit D, Schiltmeyer B, Hammes W, Horstmann R. (2004) Food does not affect the pharmacokinetics of SPM 927 (Abstract 2.342). Epilepsia 45(Suppl 7):307. Cramer JA, Ben-Menachem E, French J. (2001) Review of treatment options for refractory epilepsy: new medications and vagal nerve stimulation. Epilepsy Res 47:17–25. Doty P, Rudd GD, Stoehr T, Thomas D. (2007) Lacosamide. Neurotherapeutics 4:145–148. Errington AC, Coyne L, Stçhr T, Selve N, Lee G. (2006) Seeking a mechanism of action for the novel anticonvulsant lacosamide. Neuropharmacology 50:1016–1029. Errington AC, Stçhr T, Heers C, Lees G. (2008) The investigational anticonvulsant lacosamide selectively enhances slow inactivation of voltage-gated sodium channels. Mol Pharmacol 73:157–169. Horstmann R, Bonn R, Cawello W, Doty P, Rudd D. (2002) Basic clinical pharmacological investigations of the new antiepileptic drug SPM 927 (Abstract 2.174). Epilepsia 43(Suppl 7):188. Horstmann R, Bonn R, Cawello W, Doty P, Rudd D. (2003) SPM 927 does not interact with valproic acid and carbamazepine (Abstract 1.271). Epilepsia 44(Suppl 9): 97. Hovinga CA. (2003) SPM-927 (Schwarz Pharma). IDrugs 6:479–485. ILAE. (1981) Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for revised clinical and electroencephalographic classification of epileptic seizures. Epilepsia 22:489–501.

453 Lacosamide for Partial-Onset Seizures Krauss G, Vaiciene N, Ben-Menachem E, Johnson M, Mameniskiene R, Brock M. (2007) Intravenous lacosamide as replacement for oral Lacosamide in subjects with partial seizures: a multicenter, openlabel, inpatient trial examining safety and tolerability of 10- and 15minute infusion durations. Presented at the 61st Annual American Epilepsy Society Meeting, Philadelphia, PA, November 30–December 4, 2007. Kropeit D, Scharfenecker U, Schiltmeyer B, Koch B, Rudd GD, Cawello W, Horstmann R. (2006) Lacosamide has low potential for drug-drug interaction (Abstract 851). J Pain 7:S63. Kwan P, Brodie MJ. (2000) Early identification of refractory epilepsy. N Engl J Med 342:314–319. Marson AG, Kadir ZA, Chadwick DW. (1996) New antiepileptic drugs: a systematic review of their efficacy and tolerability. BMJ 313:1169– 1174. Marson AG, Kadir ZA, Hutton JL, Chadwick DW. (1997) The new antiepileptic drugs: a systematic review of their efficacy and tolerability. Epilepsia 38:859–880.

Sander JW. (1998) New drugs for epilepsy. Curr Opin Neurol 11:141– 148. Sander JW, Bell GS. (2004) Reducing mortality: an important aim of epilepsy management. J Neurol Neurosurg Psychiatry 75:349–351. Sheets PL, Heers C, Stoehr T, Cummins TR. (2008) Differential block of sensory neuronal voltage-gated sodium channels by lacosamide, lidocaine and carbamazepine. J Pharmacol Exp Ther 326: 89–99. Shneker BF, Fountain NB. (2003) Epilepsy. Dis Mon 49:426–478. Stçhr T, Kupferberg HJ, Stables JP, Choi D, Harris RH, Kohn H, Walton N, White HS. (2007) Lacosamide, a novel anti-convulsant drug, shows efficacy with a wide safety margin in rodent models for epilepsy. Epilepsy Res 74:147–154. Thomas D, Horstmann R, Scharfenecker U, Yates S, Nickel B, Doty P. (2007) Lacosamide has low potential for drug-drug-interaction. Poster presented at: The American Society of Consultant Pharmacists 38th Annual Meeting, Philadelphia, PA, November 14–17, 2007.

Epilepsia, 50(3):443–453, 2009 doi: 10.1111/j.1528-1167.2008.01951.x