throbber
(cid:9) CURRENT REVIEW IN CLINICAL SCIENCE
`
`RUFINAMIDE: A NOVEL
`BROAD-SPECTRUM ANTIEPILEPTIC
`DRUG
`
`James W. Wheless, MD, FAAP1 and Blanca Vazquez, MD2
`
`'Professor and Chief of Pediatric Neurology & LeBonheur
`Chair in Pediatric Neurology, University of Tennessee Health
`Science Center, Tennessee; Director, Neuroscience Institute
`& LeBonheur Comprehensive Epilepsy Program, LeBonheur
`Children's Medical Center, Tennessee; and Clinical Chief &
`Director of Pediatric Neurology, St Jude Children's Research
`Hospital, Tennessee
`'Assistant Professor of Neurology, New York University, New
`York, New York
`
`The last 20 years have witnessed a tremendous explosion in the
`number of antiepileptic drugs (AEDs) as well as the introduction
`ofAEDS developed for specific epilepsy syndromes. The study of the
`efficacy and side effect profile ofAEDs for unique epilepsy syndromes
`has allowed neurologists to utilize evidence-based medicine when
`treating patients. In late 2008, the Food and DrugAdministration
`approved rufinamide for adjunctive use in the treatment of seizures
`associated with Lennox—Gastaut syndrome. This unique chemical
`compound is also the first new AED to reach the market in the
`United States having a pediatric indication prior to approval for
`adults. Rufinamide appears to have a broad spectrum ofefficacy, is
`well tolerated and may be rapidly initiated—properties that will
`likely extend its use outside of Lennox—Gastaut syndrome.
`
`Rufinamide's chemical name is: 1-[(2,6-difluorophenyl)
`methyl]-1H-1,2,3-triazole-4 carboxamide (see Figure 1); it is
`a triazole derivative structurally unrelated to any currently mar-
`keted antiepileptic drug (AED) (1). Rufinamide was granted
`orphan drug status for adjunctive treatment of patients with
`
`Address correspondence to James W. Wheless, MD, FAAP, Professor
`and Chief of Pediatric Neurology & LeBonheur Chair in Pediatric Neu-
`rology, University of Tennessee Health Science Center, Tennessee; Di-
`rector, Neuroscience Institute & LeBonheur Comprehensive Epilepsy
`Program, LeBonheur Children's Medical Center, Tennessee; and Clin-
`ical Chief & Director of Pediatric Neurology, St Jude Children's Re-
`search Hospital, 777 Washington Avenue, Suite 335, Memphis, TN
`38105. E-mail: jwheless@utmem.edu
`Epilepsy Currents, Vol. 10, No. 1 ( January/February) 2010 pp. 1-6
`Wiley Periodicals, Inc.
`American Epilepsy Society
`
`Lennox—Gastaut syndrome in October 2004, received its mar-
`keting authorization in Europe in January 2007, and was ap-
`proved by the FDA in December in 2008 for adjunctive treat-
`ment of seizures associated with Lennox—Gastaut syndrome for
`children 4 years or older and for adults. The purposes of this
`paper are to present the significant parameters for the use of
`rufinamide in clinical practice and to summarize the results of
`phases II and III clinical trials.
`
`Pharmacology
`
`The precise mechanisms by which rufinamide exerts its
`antiepileptic effect are unknown. In vitro studies suggest that
`a principal mechanism of action is the modulation of activity
`in sodium channels, particularly prolongation of the inactive
`state. In cultured cortical neurons from immature rats, rufi-
`namide significantly slowed sodium channel recovery from in-
`activation after a prolonged prepulse and limited the sustained
`repetitive firing of sodium-dependant action potentials (1,2).
`Rufinamide has no effect on benzodiazepine or GABA recep-
`tors or on adenosine uptake; it also has no significant interac-
`tions with glutamate, adrenergic, tryptophan, histamine, and
`muscarinic cholinergic receptors.
`The antiepileptic effect of rufinamide has been assessed
`in several animal models of generalized and partial seizures.
`For instance, oral rufinamide exhibited acute anticonvul-
`sive activity in mice and rat models, suppressing maximal
`electroshock-induced tonic—clonic seizures in both species and
`pentylenetetrazol-induced clonic seizures in mice (2). In the
`maximal electroshock test conducted in mice, the effective dose
`required for a 50% response against induced seizures (i.e., ED50)
`was 23.9 mg/kg for rufinamide compared to values of 9.0, 20.1,
`664.8, and >2,000 mg/kg for the established AEDs pheny-
`toin, phenobarbital, valproate, and ethosuximide, respectively.
`In mouse pentylenetetrazol tests, the ED50 values were lower for
`rufinamide (45.8 mg/kg) than for ethosuximide (192.7 mg/kg),
`phenytoin (>300 mg/kg), and valproate (388.3 mg/kg). Sim-
`ilarly, the behavioral toxicity of rufinamide was equivalent or
`much better than the four AEDs tested in this study. Intraperi-
`toneal rufinamide suppressed pentylenetetrazol-, bicuculline-
`and picrotoxin-induced clonus in mice. Efficacy in all seizure
`models suggests that rufinamide is likely to be of value in a broad
`spectrum of seizure types, although results in animal models
`may not translate to humans.
`
`Pharmacokinetics
`
`Rufinamide is well absorbed after oral administration. The ex-
`tent of absorption decreases slightly as the dose is increased,
`4$ (cid:9)
`EXHIBIT
`ARGENTUM Exhibit 1064
` Argentum Pharmaceuticals LLC v. Research Corporation Technologies, Inc.
`IPR2016-00204
`
`196/m/id
`
`• (cid:9)
`
`Page 00001
`
`

`
`2
`
`Current Review in Clinical Science
`
`removed during dialysis. There is no autoinduction of rufi-
`namide metabolism. The effect of hepatic impairment has not
`been studied.
`Clinical trials have shown no significant differences in the
`pharmacokinetic parameters as a function of age within the
`range tested (i.e., age 4 years to elderly subjects). However, ap-
`plying the parameters derived from the pooled population phar-
`macokinetic analysis, one would predict rufinamide clearance
`at a full dose (45 mg/kg/day) to be 50% higher in a 4-year-old
`child than in an adult. Serum rufinamide levels can help guide
`the clinical decision making for a given patient, as variability
`in the rate and extent of absorption, comedications, and indi-
`vidual differences in drug clearance may impact the serum level
`and clinical efficacy. In addition, the significant relationship
`between therapeutic and adverse effects and plasma rufinamide
`concentrations suggests that measurement of rufinamide levels
`will be of value in clinical practice. Identifying the concen-
`tration at which a patient shows a good response provides a
`reference when evaluating the cause of a subsequent change in
`clinical status (4,5). Population pharmacokinetic studies reveal
`a positive correlation between reduction in seizure numbers and
`plasma rufinamide concentrations. Rufinamide reduced partial
`seizures and seizures associated with Lennox—Gastaut syndrome
`in a concentration-dependant manner. The mean plasma rufi-
`namide concentration to reduce seizure frequency by 25% or
`50% was predicted to be 15 and 30 mcg/mL, respectively (3).
`
`Drug Interactions
`
`Rufinamide does not have significant pharmacokinetic in-
`teractions with benzodiazepines, carbamazepine, lamotrigine,
`phenytoin, phenobarbital, valproate, topiramate, vigabatrin,
`oxcarbazepine, or primidone (3). However, cytochrome P450
`enzyme inducers, such as phenobarbital, primidone, phenytoin,
`and carbamazepine, increase the clearance of rufinamide, which
`likely is secondary to induction of carboxylesterases activity. The
`coadministration of these enzyme-inducing AEDs with rufi-
`namide leads to dramatically decreased rufinamide levels and
`potentially decreased efficacy (6). These patients may require
`a higher rufinamide dose. In contrast, valproate administra-
`tion may lead to elevated levels of rufinamide; the effect was
`most dramatic in children, for whom rufinamide concentra-
`tions can increase by 60 to 70 percent (1,3). The highest serum
`levels of rufinamide are noted in patients with high serum val-
`proate levels and who are concurrently taking high doses of rufi-
`namide. The exact mechanism for this interaction is unclear, but
`valproate is known to inhibit a number of drug-metabolizing
`enzymes.
`Clinical studies have shown that rufinamide can increase
`the clearance of oral contraceptives, specifically ethinyl estra-
`diol and norethindrone. The clinical significance of this mild
`
`N H 2
`
`FIGURE I . Chemical structure of rufinamide.
`
`however the effect is negligible at most clinical doses (3). Rufi-
`namide absorption is enhanced by food, probably by improved
`solubility. This enhancement results in over a 50% increase in
`the peak exposure (Cmax) and approximately a one-third in-
`crease in overall absorption. Patients will need to be advised
`to take rufinamide each time in the same temporal relation to
`their meals to maintain steady concentrations from one dose
`to the next. Rufinamide has low protein binding (about 34%),
`suggesting that competition for protein binding would not be
`a source of drug interaction, and its volume of distribution af-
`ter an oral dose approximates total body water (i.e., 50-80 L)
`(Table 1).
`The elimination of rufinamide occurs via hepatic
`metabolism with the primary metabolite, resulting from
`carboxylesterase-mediated enzymatic hydrolysis of the carboxy-
`lamide moiety, to form an inactive carboxylic acid derivative
`(CGP 47,292) (1,3). The metabolite has no known pharmaco-
`logic activity, is excreted in the urine, and the metabolic route is
`not cytochrome P450 dependant. Rufinamide is a weak inducer
`of CYP3A4 enzymes and is susceptible to induction by other
`AEDs, with the resulting effect of a decrease in rufinamide
`serum levels in their presence. Rufinamide pharmacokinetics
`are not affected by impaired renal function. The renal excre-
`tion of unchanged rufinamide is less than 2% of the total dose.
`The half-life of rufinamide is approximately 6 to 10 hours and
`does not change with renal impairment. Dose adjustment is
`likely necessary for patients undergoing hemodialysis, as the
`drug's low protein binding would result in the free drug being
`
`TABLE 1. Pharmacokinetics of Rufinamide
`Bioavailability (cid:9)
`Tmax (cid:9)
`Tye
`Protein binding (cid:9)
`Volume of distribution ( VdIF)
`Serum levels (cid:9)
`
`Fed-85%
`4 to 6 hours
`6 to 10 hours
`26 to 34%
`50 to 80 L (0.8-1.2 L/kg)
`5 to 55 mcg/mL
`
`Page 00002
`
`(cid:9)
`(cid:9)
`

`
`Current Review in Clinical Science
`
`3
`
`interaction is not known. The extent of the decreased plasma
`concentrations caused by rufinamide is much less than that
`caused by phenytoin, carbamazepine, and phenobarbital. The
`finding is consistent with the weak induction of the P450 3A4
`enzyme by rufinamide.
`
`Efficacy Demonstrated in Clinical Studies
`
`Placebo-controlled studies for rufinamide that have efficacy data
`include studies involving: 1) patients with Lennox—Gastaut syn-
`drome (see Table 2), 2) adult partial onset seizures (for both
`monotherapy and adjunctive therapy), 3) pediatric partial on-
`set seizures as adjunctive therapy, and 4) patients with refractory
`generalized tonic—clonic seizures (7).
`
`Seizures Associated with Lennox—Gastaut Syndrome
`
`An international, multicentered, double-blind, placebo-
`controlled, randomized, parallel-group study, performed be-
`tween early 1998 and fall of2000, enrolled 138 patients (ages 4-
`30 years) with a diagnosis of inadequately controlled seizures as-
`sociated with Lennox—Gastaut syndrome (including both drop
`attacks and atypical absence seizures) and who were being
`treated with one to three AEDs (felbamate therapy was not
`allowed in this study) (8). Each patient was required to have
`had at least 90 seizures in the month prior to study entry. After
`a 4-week baseline phase, patients were randomized to receive
`either rufinamide or placebo during a 12-week double-blind
`phase. The double-blind phase consisted of a titration period
`(over 1-2 weeks) and a maintenance period (10 weeks). During
`the titration period, the dose was increased to approximately
`45 mg/kg/day (maximum dose 3,200 mg/day); 77% of pa-
`tients achieved their final dose level by the end of the first week,
`which was kept stable during the maintenance period. Doses
`were given on a twice-daily schedule.
`
`The primary end points evaluated were the percent of
`change in drop attacks (tonic—atonic seizures), total seizure fre-
`quency, and the seizure severity rating taken from a global eval-
`uation of the patient's condition. Rufinamide-treated patients
`had a 42.5% median reduction in drop attacks per 28 days rel-
`ative to the baseline compared to placebo-treated patients, who
`had a 1.4% median increase (p < 0.0001). The rufinamide-
`treated patients also had a significant decrease in the total seizure
`frequency per 28 days relative to the baseline (p = 0.0015: me-
`dian reduction for rufinamide was 32.7% vs 11.7% for placebo).
`These results are comparable to the findings in other clini-
`cal trials involving topiramate, lamotrigine, and felbamate (see
`Figure 2). In addition, there was significant improvement on
`the seizure severity global evaluation for the rufinamide group
`compared with the placebo group (p < 0.005). Population
`pharmacokinetic modeling revealed that the reduction in atonic
`seizures, total seizures, and seizure severity was correlated with
`rufinamide serum concentrations. Patients who received rufi-
`namide were approximately four times more likely to experience
`at least a 50% reduction in drop attacks, compared with those
`receiving placebo. The response to rufinamide could be seen
`as early as week 2. In the open label extension phase, patients
`who switched from double-blind rufinamide to open-label rufi-
`namide continued responding to treatment (9). Figures 2 and 3
`compare the clinical response to other trials involving patients
`with Lennox—Gastaut syndrome (10-14).
`
`Partial Onset Seizures
`
`Two double-blind, placebo-controlled, randomized, parallel-
`group studies (n = 313 and 647) have been performed using
`rufinamide as adjunctive therapy for partial onset seizures. One
`was a fixed-dose study of adolescents and adults, 16 years or
`older, and the other was a dose-ranging study of adolescents
`
`TABLE 2. Summary of Clinical Studies with Rufinamide
`
`STUDY
`TYPE
`
`Adjunct
`
`Adjunct
`
`SEIZURE TYPE
`
`DAILY DOSE
`
`Lennox—Gastaut
`syndrome
`Partial onset
`
`45 mg/kg (maximum
`3,200 mg) or placebo
`200, 400, 800, 1,600
`or placebo
`3,200 mg or placebo
`3,200 mg or placebo
`
`Partial onset
`Adjunct
`Monotherapy Partial onset
`
`AGE
`(YEARS)
`
`4 to 30
`
`>15
`
`>16
`>12
`
`OUTCOME*
`
`REFERENCE
`
`,,Drop attacks 4, Total seizures
`4.Seizure severity
`4.Total seizures (+) Responder rate
`
`4. Total seizures (+) Responder rate
`Fewer seizures and longer time
`to first, second, and third seizure
`for rufinamide
`No difference vs. placebo
`
`8
`
`1
`
`1
`
`7
`
`Adjunctt
`
`Primary GTC
`
`800 mg or placebo
`
`>4
`
`Abbreviations: GTC, generalized tonic—clonic.
`*All were significant ( p < 0.05) except study Ref. 7.
`The doses used did not provide patients with plasma rufinamide concentrations that are therapeutic for other seizure types, which could explain the lack of
`efficacy seen in this study.
`
`Page 00003
`
`

`
`4
`
`Current Review in Clinical Science
`
`FIGURE 2. Short-term, double-blind stud-
`ies on Lennox-Gastaut syndrome. Abbrevi-
`ations: FLB, felbamate; TPM, topiramate;
`LTG, lamotrigine; RFM, rufinamide; CLB,
`clobazam. References: 1, #10; 2, #11;
`3, #12; 4, #8; 5, #15. Felbamate: ap-
`proved for all ages. Lamotrigine and topi-
`ramate: approved for ages 2 years and older
`in Lennox-Gastaut syndrome. Clobazam:
`high dose is 1 mg/kg/day, max 40 mg/day,
`given BID; low dose is 0.25 mg/kg/day, max
`10 mg/day. There was a significant 14% de-
`crease over 4 weeks.
`
`85%
`
`44%
`
`34%
`
`42.5%
`
`14.8%
`
`Placebo
`n = 50
`
`Placebo
`(n = 60)
`
`FLB' Placebo
`(n = 28) (n = 22)
`
`TPM2
`(n = 48)
`
`LTG3 Placebo
`(n .75)(n= 89)
`
`RFM4
`(n = 73) 1.4%
`
`CLEP
`(n = 36)
`
`5.1%
`
`-85
`
`—45
`
`—25
`
`0
`
`+10
`
`% Decrease in Drop Attacks
`
`and adults, ages 16 to 65 years (1). In both studies, patients had
`inadequately treated partial seizures and were on AED therapy.
`In the first study, the patients were required to have had at least
`one partial seizure in each 4-week period of a baseline phase
`and were then randomized to rufinamide or placebo during a
`13-week double-blind phase (1). Titration of rufinamide oc-
`curred over 1 to 2 weeks. The initial dose of 800 mg/day was
`increased to a target dose of 3,200 mg/day, given as a twice-daily
`dose for an 11-week maintenance period. Rufinamide-treated
`patients experienced a significant, although modest, reduction
`(p = 0.0158) in partial seizure frequency per 28 days com-
`pared with placebo-treated patients (a 20.4% median decrease
`vs a 1.6% median increase). In addition, the responder rate (at
`least a 50% reduction in partial seizure frequency per 28 days)
`during the double-blind phase relative to the baseline phase was
`28.2% for rufinamide compared with 18.6% for placebo (p =
`0.0381).
`
`In the second adjunctive trial for partial onset seizures, pa-
`tients were required to have experienced nine or more seizures
`during the 12-week baseline phase (1). They were then random-
`ized to one of five treatment groups (placebo or rufinamide at
`200, 400, 800, or 1,600 mg/day); treatments were administered
`on a twice-daily schedule for the 3-month double-blind phase.
`Significant dose response was observed and pairwise compar-
`isons between placebo and each rufinamide treatment group
`showed that the seizure frequency ratio was statistically signifi-
`cantly lower for the 400-, 800-, and 1,600-mg groups. In addi-
`tion, a significant dose response was observed for the responder
`rate (p < 0.04).
`A single monotherapy study has been performed—a
`double-blind, placebo controlled, randomized, parallel-group
`study (n = 104) involving inpatients, ages 12 years and older,
`with uncontrolled partial seizures, who had just completed an
`inpatient presurgical evaluation. The patients had a 48-hour
`
`70%
`
`a)
`al 60%
`fi2
`0 so%
`V CI
`
`Cl,
`
`C. (cid:9)
`to N
`w (cid:9)
`CC
`
`40%
`
`30%
`
`° e,) 20%
`0
`A 10%
`
`0
`
`Drop Attacks
`
`Total Seizure Count
`
`66%
`
`55%
`
`51%
`
`47.9%
`
`45%
`
`41%
`
`FLB1
`(n = 50;
`12 mo)
`
`TPM2
`(n = 82;
`6 mo)
`
`RFM3
`(n = 56;
`12 mo)
`
`FLB
`(n = 71;
`12 mo)
`
`TPM
`(n = 84;
`6 mo)
`
`RFM
`(n = 50;
`12 mo)
`
`FIGURE 3. Long-term, open-label stud-
`ies of AED efficacy for Lennox-Gastaut
`syndrome. Abbreviations: FLB, felbamate;
`TPM, topiramate; RFM, rufinamide. Ref-
`erences: 1, #13; 2, #I4; 3, #9. Lamotrig-
`ine: no long-term data reported.
`
`Page 00004
`
`

`
`Current Review in Clinical Science (cid:9)
`
`5
`
`baseline prospective phase and then were randomized to either
`to rufinamide, 2,400 mg/day on day 1 and 3,200 mg/day on
`days 2 to 10 (given three times per day), or to placebo. The pri-
`mary efficacy variable was the mean time to meet the exit crite-
`ria. Outcome data favored rufinamide (p < 0.05) over placebo,
`with a median time to exit of 4.8 days compared with 2.4 days.
`Statistically significantly differences between treatments were
`observed for the time to first, second, and third partial seizures
`(p < 0.04), however the time to the fourth partial seizure failed
`to reach significance (p = 0.0509).
`
`Long-Term Follow-Up
`Both the Lennox—Gastaut study and the studies on par-
`tial seizures were followed by long-term, open-label extension
`studies. The patients who switched from double-blind placebo
`to open-label rufinamide quickly responded to treatment, with
`a marked decrease in seizure frequency. There was no evidence
`of tolerance to the anticonvulsant effect of rufinamide, during
`up to 3 years of follow-up (1).
`
`Dosing, Tolerability, and Safety
`
`Table 3 provides the authors suggestions for dosing in children
`and adults. The clinical trials were performed with administra-
`tion of the drug with food (resulting in enhanced absorption),
`which is the recommended protocol.
`Based on the clinical trials, rufinamide appears to be well
`tolerated. A small number of rufinamide-treated patients (9%
`vs 4% for placebo) discontinued treatment because of adverse
`effects (15). The adverse experiences most commonly associ-
`ated with discontinuation of rufinamide (>1%) were similar
`in adults and children: dizziness (1.8%), fatigue (1.6%), and
`headache (1.1%). The majority of adverse events in the clinical
`trials were judged to be mild to moderate and often transient in
`nature, largely occurring during the titration phase. The most
`commonly observed adverse events (i.e., occurring in >10%
`and at a higher frequency than placebo-treated patients), pooled
`from all of the studies of patients with epilepsy, were headache,
`dizziness, fatigue, somnolence, and nausea. Adverse events were
`
`reported more often in adults than in children and with plasma
`rufinamide concentrations in the higher ranges. Only somno-
`lence and vomiting were significantly more common in the ru-
`finamide group of the Lennox—Gastaut syndrome trial. At the
`fixed titration dose of 45 mg/kg/day in all pediatric trials, only
`somnolence, vomiting, and headache were significantly more
`common with rufinamide than placebo (i.e., observed >5%
`more often). In doses up to 3,200 mg/day in all adult clini-
`cal trials, only dizziness, fatigue, and diplopia were significantly
`more common with rufinamide than placebo. Neuropsychiatric
`side effects were rare (all <5%) and were no more common in
`rufinamide than in placebo groups. The rufinamide side effect
`profile is similar to other drugs that have an effect on the sodium
`channel.
`The overall tolerability of rufinamide is good. During the
`clinical trials, there were no cases of Stevens—Johnson syndrome,
`hepatic failure, agranulocytosis, or pancytopenia. The incidence
`of cognitive disorders in rufinamide-treated patients was higher
`than placebo-treated patients only because of the increased oc-
`currence of somnolence. Psychiatric adverse events were similar
`between rufinamide and placebo patients.
`AED hypersensitivity syndrome has occurred in association
`with rufinamide therapy. While the clinical symptoms varied,
`patients generally presented with fever and rash associated with
`other organ system involvement. In the clinical trials, this syn-
`drome occurred in close temporal association (within the first
`4 weeks) to the initiation of rufinamide therapy and was more
`likely in the pediatric population. If a serious rash related to ru-
`finamide is suspected, rufinamide should be discontinued and
`alternative treatment started.
`In the randomized trial, cognitive assessments were per-
`formed at baseline (before rufinamide treatment) and after
`3 months of adjunctive therapy at doses of 200, 400, 800, and
`1,600 mg/day for adolescents and adults (ages 15-64 years) with
`partial seizures (16). None of the cognitive tests for psychomo-
`tor speed and attention or for working memory demonstrated
`a significant worsening at any of the doses of rufinamide. In a
`placebo-controlled study of the QT interval, a higher percentage
`
`(cid:9) (cid:9)
`
`TABLE 3. Rufinamide Dosing
`
`LABEL (FDA)
`
`AUTHORS' RECOMMENDATIONS
`
`Children Given BID: Begin 10 mg/kg/day, Increase by 10 mg/kg,
`every other day to 45 mg/kg/day or 3,200 mg/day
`(whichever is less)
`Given BID: Begin with 400 to 800 mg/day Increase by
`400 to 800 mg every 2 days, up to a maximum of
`3,200 mg/day
`
`Adults
`
`Given BID or TID: Begin 15 mg/kg/day Increase by
`15 mg/kg/day, every week to 45 mg/kg/day or
`3,600 mg/day (whichever is less)
`Given BID or TID: Begin with 1,200 mg/day Increase
`by 1,200 mg/day every week up to 3,600 mg/day
`
`Abbreviations: BID, twice daily dosing; TID, three times daily dosing.
`Take with food. Supplied in 200- and 400-mg scored tablets (and 100 mg in Europe), which can be administered whole, in half tablets, or crushed.
`
`Page 00005
`
`(cid:9)
`

`
`6 (cid:9)
`
`Current Review in Clinical Science
`
`of subjects taking 2,400-4,800 mg of rufinamide per day had a
`QT shortening of greater than 20 milliseconds compared with
`placebo, but none had a reduction below 300 milliseconds. Pa-
`tients with potassium channelopathy associated with familial
`short QT syndrome cannot be treated with rufinamide. Cau-
`tion is advised when administering rufinamide with other drugs
`or disease states that shorten the QT interval (e.g., digoxin tox-
`icity, hypercalcemia, hyperkalemia, and acidosis). There is no
`known clinical risk associated with the degree of QT shortening
`induced by rufinamide. No meaningful changes in laboratory
`data were observed, and rufinamide is designated pregnancy
`Category C. When assessing the risk of rufinamide or any new
`drug, it is important to remember that not all potential risks
`may have been identified, which is because only a relatively
`small number of patients have been exposed to the drug for a
`long period of time.
`
`Conclusions
`
`Rufinamide is a new broad-spectrum AED that is structurally
`unique. It offers various advantages: 1) the ability to rapidly
`escalate dosing and obtain a clinical response, 2) few drug in-
`teractions, and 3) a good cognitive and psychiatric adverse event
`profile. The CNS-related adverse events (primarily somnolence)
`largely occurred during the first 2 weeks of therapy, which may
`be related to the rapid fixed titration schedule used in the clinical
`trials. Slower titration helps minimize side effects. No labora-
`tory monitoring is required, and plasma levels correlate with
`clinical efficacy. All of these characteristics will make it a com-
`monly used drug in Lennox-Gastaut syndrome. Further tri-
`als are ongoing. Continuing clinical experience may elucidate
`whether rufinamide eventually will prove beneficial for a wider
`spectrum of patients.
`
`References
`
`1. Arroyo S. Rufinamide. Neurotherapeutics 2007;4:155-162.
`2. White HS, Franklin MR, Kupferberg HJ, Schmutz M, Stables JP,
`Wolf HH. The anticonvulsant profile of rufinamide (CGP33101)
`in rodent seizure models. Epilepsia 2008;49:1213-1220.
`3. Perucca E, Cloyd J, Critchley D, Fuseau E. Rufinamide: Clini-
`cal Pharmacokinetics and concentration-response relationships in
`patients with epilepsy. Epilepsia 2008;49:1123-1141.
`
`4. Patsalos PN, Berry DJ, Bourgeois BF, Cloyd JC, Glauser TA,
`Johannessen SI, Leppick IE, Tomson T, Perucca E. Antiepileptic
`drugs-best practice guidelines for therapeutic drug monitoring: A
`position paper by the subcommission on therapeutic drug mon-
`itoring, ILAE Commission on Therapeutic Strategies. Epilepsia
`2008;49:1239-1276.
`5. Perucca E. Is there a role for therapeutic drug monitoring of new
`anticonvulsants? Clin Pharmacokinet 2000;38:191-204.
`6. Brodie M, Li H, Wang W, Narurkar M, Richardson S. Differential
`effects of rufinamide in adults with partial onset seizures as a
`function of concomitant anti-epileptic drug therapy: A post hoc
`analysis. Neural 2009;72(11, suppl 3):A228.
`7. Biton V, Rosenfeld W, Schachter S, Perdomo C, Arroyo S. Mul-
`ticenter, randomized, double-blind, placebo-controlled, parallel
`trial comparing the safety and efficacy of rufinamide. Ann Neural
`2005;58(suppl 9):S114.
`8. Glauser T, Kluger G, Sachdeo R, Krauss G, Perdomo C, Arroyo
`S. Rufinamide for generalized seizures associated with Lennox-
`Gastaut Syndrome. Neurology 2008;70:1950-1958.
`9. Glauser T, Kluger G, Krauss G, Perdomo C, Arroyo S. Short
`term and long term efficacy and safety of rufinamide as adjunctive
`therapy in patients with inadequately controlled Lennox-Gastaut
`Syndrome. Neurol 2006;66(5, suppl 2):A36.
`10. Felbamate Study Group in Lennox-Gastaut Syndrome. Efficacy
`of Felbamate in Childhood Epileptic Encephalopathy (Lennox-
`Gastaut Syndrome). NEnglJMed 1993:328:29-33.
`11. Sachdeo RC, Glauser TA, Ritter F, Reife R, Lim P, Pledger G. A
`double-blind, randomized trial of topiramate in Lennox-Gastaut
`Syndrome. Topiramate YL Study Group. Neural 1999;52:1882-
`1887.
`12. Motte J, Trevathan E, Arvidsson JF, Barrera MN, Mullens EL,
`Manasco P. Lamotrigine for generalized seizures associated with
`the Lennox-Gastaut Syndrome. Lamictal Lennox-Gastaut Study
`Group. NEnglJMed 1997;337:1807-1812.
`13. Dodson WE Felbamate in the treatment of Lennox-Gastaut Syn-
`drome: Results of a 12-month open-label study following a ran-
`domized clinical trial. Epilepsia 1993;34(suppl 7):S18-S24.
`14. Glauser TA, Levisohn PM, Ritter F, Sachdeo RC, and the Topira-
`mate YL Study. Topiramate in Lennox-Gastaut Syndrome: Open-
`label treatment of patients completing a randomized controlled
`trial. Epilepsia 2000;41(suppl 1):S86-S90.
`15. Wheless J, Conry J, Krauss G, Mann A, LoPresti A, Narurkar
`M. Safety and tolerability of rufinamide in children with
`epilepsy: A pooled analysis of seven clinical trials. J Child Neu-
`rol 2009;50:1158-1166.
`16. Aldenkamp AP, Apherts WCJ. The effect of the new antiepileptic
`drug rufinamide on cognitive functions. Epilepsia 2006;47:1153-
`1159.
`
`Page 00006

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket