throbber
Epilepsy Research 51 (2002) 31 /71
`
`www.elsevier.com/locate/epilepsyres
`
`Progress report on new antiepileptic drugs: a summary of the
`Sixth Eilat Conference (EILAT VI)
`
`M. Bialer a,*, S.I. Johannessen b, H.J. Kupferberg c, R.H. Levy d, P. Loiseau e,
`E. Perucca f
`
`a School of Pharmacy and David R. Bloom Centre for Pharmacy, Faculty of Medicine, Ein Karem, The Hebrew University of Jerusalem,
`Jerusalem 91120, Israel
`b The National Centre for Epilepsy, Sandvika, Norway
`c Kupferberg Consultants, LLC, Bethesda, MD, USA
`d Department of Pharmaceutics and Neurological Surgery, University of Washington, Seattle, WA, USA
`e Department of Neurology, Bordeaux University Hospital Pellegrin, Bordeaux, France
`f Clinical Pharmacology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
`
`Received 20 May 2002; accepted 23 May 2002
`
`Abstract
`
`The Sixth Eilat Conference on New Antiepileptic Drugs (AEDs) took place in Taormina, Sicily, Italy from 7th to
`11th April, 2002. Basic scientists, clinical pharmacologists and neurologists from 27 countries attended the conference,
`whose main themes included dose /response relationships with conventional and recent AEDs, teratogenic effects of
`conventional and recent AEDs, update on clinical implications of AED metabolism, prevention of epileptogesis, and
`seizure aggravation by AEDs. According to tradition, the central part of the conference was devoted to a review of
`AEDs in development, as well to updates on AEDs, which have been marketed in recent years. This article summarizes
`the information presented on drugs in preclinical and clinical development, including carabersat (SB-204269), CGX-
`1007 (Conantokin-G), pregabalin, retigabine (D-23129), safinamide, SPD421 (DP-VPA), SPM 927, talampanel and
`valrocemide (TV 1901). Updates on fosphenytoin, gabapentin, lamotrigine, levetiracetam, oxcarbazepine, tiagabine,
`topiramate, vigabatrin, zonisamide, new formulations of valproic acid, and the antiepileptic vagal stimulator device are
`also presented. # 2002 Elsevier Science B.V. All rights reserved.
`
`Keywords: Antiepileptic drugs; Drug development; Epilepsy; Pharmacology; Clinical trials; Conference
`
`* Corresponding author. Tel.: /972-2-6758610; fax: /972-
`2-675746
`E-mail address: bialer@md.huji.ac.il (M. Bialer).
`
`1. Introduction
`
`The Sixth Eilat Conference on New Antiepilep-
`tic Drugs (AEDs) took place in Taormina, Sicily,
`Italy from 7th to 11th April, 2002. The conference
`saw active participation of basic scientists, clinical
`pharmacologists and neurologists from 27 coun-
`tries, with representatives from academia,
`the
`
`0920-1211/02/$ - see front matter # 2002 Elsevier Science B.V. All rights reserved.
`PII: S 0 9 2 0 - 1 2 1 1 ( 0 2 ) 0 0 1 0 6 - 7
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 1
`
`

`
`32
`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`pharmaceutical industry, and regulatory agencies
`from both sides of the Atlantic. Main themes
`included dose /response relationships with conven-
`tional and recent AEDs, teratogenic effects of
`conventional and recent AEDs, update on clinical
`implications of AED metabolism, prevention of
`epileptogenesis, and seizure aggravation by AEDs.
`The central part of the conference was dedicated to
`discussion of drugs in development, as well as to
`updates on AEDs marketed in recent years, new
`formulations of valproic acid (VPA) and vagal
`nerve stimulator device. The following is a sum-
`mary on the contributions related to these topics.
`
`2. Drugs in development
`
`2.1. Carabersat (SB-204269)
`
`J.A. Messenheimer
`GlaxoSmithKline (GSK), Research Triangle Park,
`NC, USA.
`
`2.1.1. Introduction
`Carabersat (CRB; SB-204269) is a novel fluor-
`obenzoylamino benzopyran compound which ex-
`hibits
`excellent
`anticonvulsant potency
`and
`efficacy relative to phenytoin (PHT), carbamaze-
`pine (CBZ), DZP, lamotrigine (LTG) and GBP in
`a wide range of in vivo (pentylenetetrazol (PTZ)
`
`infusion, maximal electroshock (MES) seizure
`threshold,
`supramaximal electroshock) and in
`vitro (elevated potassium and zero magnesium
`brain slice assays) rodent seizure models (Upton
`et al., 1997). Overall, the profile of activity in these
`models strongly suggests that the compound acts
`by preventing seizure spread. This particular mode
`of action is also shared by AEDs such as CBZ and
`LTG and may translate into clinical utility for
`symptomatic control of partial (with and without
`generalization) and generalized tonic-clonic sei-
`zures. In addition, CRB appears to slow the
`development of amygdala-kindled seizures
`in
`rats,
`indicating that it may be able to inhibit
`epileptogenesis. Importantly, the anticonvulsant
`properties of CRB are of long duration and there
`is no evidence of a decline in efficacy following
`repeated administration.
`
`2.1.2. Mechanism of action
`Although the precise molecular mechanism(s) of
`CRB remain(s) to be determined, a specific bind-
`ing site for the compound (first discovered using
`[3H]-CRB) has been identified in the brain tissue of
`several species including mouse, rat, cat, dog,
`marmoset and, most importantly, man. The high-
`est levels of binding are found in the superficial
`layers of the cerebral cortex and granule cell layer
`of the cerebellum, with moderate levels in CA
`fields and dentate gyrus of
`the hippocampus
`(Herdon et al., 1997). Several lines of evidence
`now strongly support the relevance of this binding
`site to the anticonvulsant properties of CRB. For
`example,
`there is a good correlation between
`binding affinity and anticonvulsant activity in the
`mouse MES seizure threshold test for a wide range
`of analogues of CRB. To date, no activity of CRB
`(pKi5/5) has been identified in over 50 radioli-
`gand-binding assays. These include binding to
`amino acid receptors and ion channels, sodium
`and potassium channels, purinergic and aminergic
`receptors, and opioid and other peptidergic recep-
`tors. In addition, CRB did not demonstrate
`significant activity in a range of in vitro functional
`assays. These findings indicate that CRB interacts
`selectively with its own binding site and, unlike
`other AEDs, has no known effect on sodium
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 2
`
`

`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`33
`
`channels or GABAergic or glutamatergic path-
`ways.
`
`2.1.3. Pharmacokinetics
`A total of 136 healthy volunteers have com-
`pleted dosing with CRB in eight separate studies.
`CRB was well tolerated. Pharmacokinetic data
`from these studies have shown that the current
`formulation exhibits dose proportional changes in
`maximum concentration (Cmax) and area-under-
`the-curve (AUC), a variable time to maximum
`concentration (tmax) of several hours, and an
`apparent terminal half-life of 24 h. Oral bioavail-
`ability is enhanced by food. Radiolabeled studies
`with [14C]-carabersat have shown that the com-
`pound is predominantly cleared by hepatic meta-
`bolism in man.
`
`2.1.4. Efficacy
`A parallel-design, multicenter, double-blind,
`placebo-controlled evaluation of the safety and
`efficacy of CRB given as add-on therapy in 305
`patients (228 randomized to CRB, 77 to placebo)
`with refractory simple or complex partial seizures
`(with or without secondary generalization) has
`been completed. The parallel groups received 400,
`800 or 1200 mg/day CRB or placebo. This study,
`which used the initial formulation of CRB, showed
`a mean reduction in seizure frequency of 20 /30%
`in the 1200 mg/day dose group compared with
`placebo (P B/0.05). In general, CRB was well
`tolerated at all dose groups. These findings suggest
`that CRB has biological activity in epilepsy and
`displays a favorable safety profile. A novel for-
`mulation of CRB has been developed to provide a
`more advantageous pharmacokinetic profile (de-
`creased Cmax, delayed tmax, plasma levels sustained
`above a Cmin level). Future clinical trials will be
`conducted using this new formulation.
`
`2.2. CGX-1007 (Conantokin-G)
`
`T. McCabea, S.C. Schachterb
`aCognetix, Salt Lake City, UT, USA, and bBeth
`Israel Deaconess Medical Center and Harvard
`Medical School, Boston, MA, USA.
`
`2.2.1. Introduction
`CGX-1007 (Conantokin-G) is a synthetic ver-
`sion of a conopeptide, derived from Conus geo-
`graphus cone snail venom,
`that has exhibited
`anticonvulsant activity in preclinical
`studies.
`CGX-1007 is 17 amino acids in length. Among
`the approaches for epilepsy management is N -
`methyl-D-aspartate (NMDA) receptor modula-
`tion. CGX-1007 is a selective NMDA receptor
`antagonist (Donevan and McCabe, 2000). It is
`proposed that CGX-1007 be administered in-
`trathecally (i.t.) to patients with epilepsy using
`the SynchroMed† Infusion System. The Medtro-
`nic SynchroMed† Infusion System consists of a
`small pump implanted in the abdominal region
`and a catheter that delivers medication to a
`specific site within the body. The system bypasses
`the digestive system and the blood /brain barrier,
`two factors important for effective delivery of a
`central nervous system (CNS) active peptide. The
`SynchroMed† Infusion System is approved by the
`FDA for the chronic intraspinal infusion of sterile
`preservative-free morphine sulfate for intractable
`chronic pain,
`including cancer pain;
`for
`the
`intrathecal infusion of baclofen for severe spasti-
`city of spinal and cerebral origin; as well as
`intravascular infusion of floxuridine for the treat-
`ment of primary or secondary metastatic cancer.
`In animal seizure models, CGX-1007 has been
`shown to exhibit broad-spectrum anticonvulsant
`activity with very low behavioral toxicity. More-
`over, CGX-1007 has been demonstrated to be
`effective in animal models of complex partial
`seizures. The objective is to develop the compound
`for the treatment of patients with partial seizures
`with or without secondary generalization refrac-
`tory to available AEDs.
`In humans, CGX-1007 appears to be safe and
`well tolerated via the intravenous (i.v.) route.
`While CGX-1007 possesses a broad-spectrum
`anticonvulsant profile, it is also recognized that
`direct administration to the CNS will be required
`for treatment of patients with epilepsy. The i.t.
`route represents the most feasible delivery ap-
`proach. Given the novel molecular mechanism of
`action, broad spectrum of activity and low beha-
`vioral toxicity, CGX-1007 may represent a unique
`and novel anticonvulsant agent and may provide a
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 3
`
`

`
`34
`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`2.2.3. Toxicology
`In support of a completed phase I clinical i.v.
`study, 14 day i.v. toxicology studies were con-
`ducted in both the rat and dog. An additional 13
`week subcutaneous study was conducted in the rat
`to evaluate the effect of
`longer-term systemic
`exposure as well as any local effects at the site of
`repeated injections. A battery of genetic toxicity
`tests
`(Ames, mouse
`lymphoma
`and mouse
`micronucleus) and immunogenicity studies also
`were conducted. To support delivery of CGX-
`1007 to the brain by direct infusion into the
`cerebrospinal fluid (CSF), i.c.v. and i.t. toxicology
`studies of 28 days and 13 weeks duration
`in both the rat and dog were conducted. It was
`also shown that,
`in general, pharmacokinetic
`parameters were similar after i.c.v. or i.t. admin-
`istration. Based on these studies, CGX-1007
`should be safe for i.t. administration at doses
`many times greater than those required to reduce
`seizure activity.
`
`2.2.4. Drug interactions
`In animal models, based on the protection
`against MES-induced seizures, there was no po-
`tentiation, but a strictly additive interaction when
`CGX-1007 was combined with PHT, CBZ, or
`VPA.
`
`2.2.5. Planned studies
`Based on the data described above, it is pro-
`posed that CGX-1007 be administered to patients
`with
`epilepsy
`by
`i.t.
`delivery
`using
`the
`SynchroMed† Infusion System.
`
`2.3. Pregabalin
`
`A.R. Kuglera, C.M. de Meynardb, L.E. Knappa
`aPfizer Global Research and Development, Ann
`Arbor, MI, USA and bPfizer Global Research and
`Development, Fresnes, France.
`
`significant advance in the treatment of patients
`with uncontrolled seizures.
`
`2.2.2. Pharmacology
`CGX-1007 is a specific NMDA receptor an-
`tagonist that does not interact with any other
`receptors or binding sites
`examined in the
`NovaScreen † receptor profiling study (concentra-
`tion tested was 10 mM). CGX-1007 exhibits wide
`and uniform brain distribution when given i.t. in
`rats and dogs.
`CGX-1007 has been characterized as an antic-
`onvulsant compound in an in vivo reflex model of
`epilepsy. CGX-1007 dose-dependently blocked
`sound-induced tonic extension following i.t. and
`intracerebroventricular (i.c.v.) injection in Frings
`audiogenic seizure-susceptible mice. In addition,
`CGX-1007 did not
`induce behavioral
`toxicity
`(TD50, median behaviorally toxic dose or minimal
`motor impairment as measured by rotarod) in
`animals until
`significantly greater doses were
`administered. The separation between the ED50
`and TD50 doses was greater than that of many
`established AEDs, therefore yielding a greater
`protective index (PI/TD50/ED50). In addition,
`CGX-1007 displayed a better PI than either the
`non-competitive NMDA antagonist MK-801 or
`the polyamine antagonist ifenprodil in the audio-
`genic, chemically- and electrically-induced seizure
`models. In the Frings model, CGX-1007 displays a
`rapid onset (within 1 /3 min) and a prolonged
`duration (2 /4 h) of action following i.c.v. admin-
`istration of a single dose.
`CGX-1007 was also found to display a broad-
`spectrum anticonvulsant profile that was similar to
`that of VPA. Thus,
`it was effective at non-
`behaviorally toxic doses against tonic extension
`seizures induced by both threshold and MES as
`well as clonic seizures induced by PTZ, bicuculline
`(BIC) and picrotoxin (PIC). CGX-1007 is effective
`in rat kindling models of partial seizures when
`given by either i.t. or i.c.v. administration. Collec-
`tively, these results indicate that CGX-1007 has
`the potential to be effective in a variety of human
`seizure disorders including generalized tonic-clonic
`seizures (anti-MES activity), generalized absence/
`myoclonic seizures (anti-PTZ activity), and partial
`seizures (kindling models).
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 4
`
`

`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`35
`
`2.3.1. Introduction
`Pregabalin (PGB), a structural analogue of the
`mammalian neurotransmitter gamma-aminobuty-
`ric acid (GABA), is the pharmacologically active
`S -enantiomer of 3-aminomethyl-5-methylhexa-
`noic acid or 3-(S )-isobutyl GABA.
`
`2.3.2. Pharmacology
`
`2.3.2.1. Anticonvulsant profile. PGB is active in a
`number of animal models of epileptic seizures
`including MES seizures, chemical convulsant sei-
`zures (PTZ, BIC, PIC), kindled seizures in rats,
`and audiogenic seizures in genetically susceptible
`animals.
`PGB prevents electroshock tonic extensor sei-
`zures when given per os (p.o.) and i.v. in mice
`low-
`(maximal stimulus ED50, 20 mg/kg, p.o.;
`intensity stimulus ED50, 1.4 mg/kg, i.v.) and rats
`(ED50, 1.8 mg/kg, p.o.). The ED50 values obtained
`following i.v. and p.o. administration were quite
`similar. PGB did not cause ataxia except at high
`doses (ED50, 60 /260 mg/kg, p.o.). PGB prevented
`threshold clonic seizures induced by PTZ in mice,
`with an ED50 value of 100 mg/kg, p.o. or
`intraperitoneally (i.p.). In mice, threshold seizures
`from BIC or PIC were not completely blocked
`(maximal 70% protection, 250 mg/kg, i.p.), and
`seizures from strychnine were not blocked. Spon-
`taneous absence seizures (6 Hz spike/wave dis-
`charges in neocortical EEG) were not altered by
`10, 40, or 100 mg/kg PGB, but were slightly
`increased by 200 or 400 mg/kg i.p. PGB decreased
`seizures in DBA/2 audiogenic mice after dosages
`of 3 and 10 mg/kg p.o. PGB exhibits a high
`potency in animal seizure models compared to
`other anticonvulsant agents, based on mg/kg
`doses.
`
`2.3.2.2. Mechanisms of action. The mechanism of
`action of PGB is unknown, but it appears to be
`different from that of conventional AEDs. PGB
`does not appear to have any direct action at ion
`channels (Na, Ca2) or transmitter responses
`(glutamate, NMDA, GABA), does not change
`neurotransmitter uptake (glutamate, GABA), and
`does not displace radioligand binding at a variety
`of
`receptors
`(glutamate, GABA, monoamine,
`
`adenosine, cholinergic, opiate) or Na and Ca2
`channels. PGB increases GABA content in neuro-
`nal tissues and enhances glutamic acid decarbox-
`ylase activity. In vitro studies show that PGB
`interacts with an auxiliary subunit (a2-d protein)
`of voltage-gated calcium channels in brain, po-
`tently displacing [3H]-gabapentin or [3H]-L-leu-
`cine. Studies with the R-enantiomer of PGB and
`a number of structural derivatives of pregabalin
`indicate that binding at the a2-d site is required for
`analgesic and anticonvulsant activity in animal
`models.
`
`2.3.3. Pharmacokinetics
`Plasma and brain concentrations of PGB in rats
`were compared with the time course of antic-
`onvulsant effects (MES). In the rat, PGB pharma-
`cokinetics was dose proportional and PGB was
`not significantly metabolized. Maximal MES ac-
`tion was observed approximately 2 /4 h after i.v.
`dosing. Plasma concentrations decreased mono-
`exponentially.
`Phase I clinical studies indicate oral bioavail-
`ability to be approximately 90%, plasma half-life
`(t1/2) 6 h, time to maximum concentration (tmax) 1
`h, and Cmax and AUC to be dose proportional.
`PGB is not significantly metabolized in man, with
`approximately 98% of an absorbed dose being
`excreted unchanged in the urine. PGB is not
`bound to plasma proteins. AUC and t1/2 remain
`unchanged following administration with a stan-
`dardized meal, while Cmax is reduced by approxi-
`25 /30% and tmax
`is
`increased to
`mately
`approximately 3 h.
`
`2.3.4. Drug interactions and tolerability
`PGB is well tolerated, with dose-related CNS
`adverse events (headache, dizziness, somnolence)
`most commonly reported by normal volunteers
`with doses up to 900 mg/day. PGB may be used as
`adjunctive therapy without changes in the blood
`concentrations of other AEDs or PGB itself (no
`drug /drug interactions identified or expected).
`Therapeutic drug monitoring is not required for
`dosing PGB. It is recommended that patients with
`impaired renal
`function have their PGB dose
`reduced.
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 5
`
`

`
`36
`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`2.3.5. Efficacy data
`PGB has been investigated in three multicenter,
`12 week, double-blind, placebo-controlled studies
`as add-on therapy in patients with partial seizures
`uncontrolled by currently available AEDs. These
`studies investigated dose /response (50 /600 mg/
`day), dose regimen (two times a day (b.i.d.) or
`three times a day (t.i.d.) dosing), and titration
`rates. The mean age at epilepsy diagnosis was 13.7
`years (range 0 /73.5) and mean duration of epi-
`lepsy was 25.0 years. Mean and median baseline
`seizure rates were 22.5 and 10.0 seizures per 28
`days, respectively. Seventy-five percent of
`the
`patients were taking two or more concurrent
`AEDs at baseline. Patient demographics were
`well distributed among the treatment groups.
`PGB administered b.i.d. or
`t.i.d. was highly
`effective in reducing seizure frequency compared
`with placebo in all three add-on trials and a clear
`dose /response relationship was shown. Responder
`rates (]/50% reduction in seizure frequency from
`baseline) ranged from 14 /31% with 150 mg/day to
`44 /51% with 600 mg/day. In an additional analy-
`sis of pooled data from all effective dose groups
`from the three controlled studies, statistically
`significant efficacy was seen as early as day 2 of
`treatment. PGB was well tolerated overall by these
`patients. The most frequently reported adverse
`events were CNS-related (dizziness, somnolence,
`and ataxia), most of which were mild to moderate
`in intensity. CNS adverse events tended to increase
`with increasing PGB doses, particularly 300 and
`600 mg/day. Overall, PGB was efficacious and well
`tolerated as reflected by the fact that 83% of PGB-
`treated patients enrolled in open-label extensions
`to the double-blind treatment phases. (French et
`al., 1999; Robbins et al., 2001; Ramsay et al.,
`2001). A presurgical, inpatient monotherapy study
`provided initial evidence of PGB anticonvulsant
`efficacy.
`In conclusion, PGB is a novel CNS active
`compound that has shown anticonvulsant activity
`as adjunctive therapy in epilepsy patients with
`partial seizures. PGB has been well tolerated and
`safe in clinical studies to date, following exposure
`in a total of approximately 1600 patients with
`epilepsy.
`
`2.4. Retigabine
`
`R. Hermanna, E. Schneidera, M. Mentha, N.
`Knebelb, G.M Ferronc, J. Borlakd, K. Erbe, A.
`Partiotc, J-M. Germainc, M. Locherb, R. Porterc
`aClinical Development VIATRIS GmbH & Co.
`KG, Frankfurt am Main, Germany, bEarly Phase
`Development VIATRIS GmbH & Co. KG,
`Frankfurt am Main, Germany, cWyeth Research,
`Radnor, PA, USA and Paris, France, dFraunhofer
`Institute of Toxicology, Drug Research and Clin-
`ical Inhalation, Center for Drug Research and
`Medical Biotechnology, Hannover, Germany and
`eCliphase, Frankfurt, Germany.
`
`2.4.1. Introduction
`Retigabine (RGB; D-23129), N -(2-amino-4-(4-
`fluorobenzyl-amino)-phenyl) carbamic acid ethyl
`ester,
`is a compound structurally unrelated to
`other marketed AEDs. Its remarkable antiepileptic
`potential was identified in the course of a screening
`at the National Institute of Health (NIH).
`
`2.4.2. Pharmacology
`
`2.4.2.1. Anticonvulsant profile. RGB exerts potent
`anticonvulsant activity in a broad variety of
`animal models including models with electrical
`and chemical induction of convulsions and genetic
`models of epilepsy (Rostock et al., 1996). RGB has
`shown a potent effect in the kindling model of
`epileptogenesis (Tober et al., 1996) and is effective
`in a rat model of status epilepticus.
`The drug is active after oral administration, and
`anticonvulsant effects can be clearly separated
`from adverse neurological effects. Unlike standard
`anticonvulsants, RGB is more active in the amyg-
`dala kindling model, a model of complex partial
`seizures in humans, than in models of generalized
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 6
`
`

`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`37
`
`seizures like the MES. PI calculations based on
`effective doses in the amygdala kindling model are
`remarkable. The N -acetyl metabolite of retigabine
`was also found to have some anticonvulsant
`activity.
`
`2.4.2.2. Other pharmacological properties. RGB
`improved learning performance in a model of
`cerebral ischemia and during electroshock amne-
`sia, indicating possible neuroprotective activity in
`models of cerebral deficit. RGB showed dose-
`dependent suppression of chronic neuropathic
`pain in two animal models (formalin test, spinal
`nerve injury test).
`
`2.4.2.3. Mechanism of action. RGB is a first-in-
`class new antiepileptic compound. The primary
`mechanism of action was shown to be a selective
`and potent M-current potassium channel opening
`effect at the KCNQ2/3 and KCNQ3/5 potassium
`channels, which are involved in the control of
`neuronal
`excitability (Rundfeldt and Netzer,
`2000). Mutations in these channels have recently
`been linked to benign neonatal familial convul-
`sions (Hirsch et al., 1999). An ancillary mode of
`action of RGB is the potentiation of GABA-
`evoked currents, which, however, occurs at much
`higher concentrations.
`
`2.4.3. Toxicology
`Repeated-dose toxicity studies of up to 12
`months duration in different animal species in-
`dicate that RGB is well tolerated. Predominant
`central side effects are sedation accompanied by
`hyperexcitability and a decrease in body tempera-
`ture. No dependence liability was observed during
`chronic treatment.
`
`2.4.4. Pharmacokinetics and metabolism
`Single- and multiple-dose pharmacokinetics of
`RGB were assessed in male and female young and
`elderly white subjects and in young male black
`subjects.
`The absolute bioavailability (F ) of oral immedi-
`ate release RGB dosage forms is 50% (data on
`file). Pharmacokinetics are linear for daily doses
`up to 700 mg and are not modified on multiple
`
`administrations. In patients, dose linearity of RGB
`could be demonstrated up to 1200 mg/day. RGB is
`rapidly absorbed, with mean maximum concentra-
`tions of 387 ng/ml (normalized to a 100 mg dose)
`occurring within 1.5 h after intake of a single dose.
`In young white subjects the mean terminal half-life
`of RGB is 8 /9 h and the apparent oral clearance
`0.70 l/h/kg. In black subjects, RGB clearance and
`volume of distribution (Vss) are 25 and 30% lower,
`respectively,
`leading to higher exposure in this
`population (Ferron et al., 2002).
`Compared with young men, higher peak RGB
`concentrations and exposure, but similar clearance
`normalized by body weight, were observed in
`young women,
`suggesting that differences
`in
`body weight between sexes may explain these
`findings. In the elderly (66 /81 years) of both
`sexes, peak plasma concentrations were not altered
`as compared with young subjects, but RGB was
`eliminated more slowly (30% lower weight-nor-
`malized clearance), resulting in higher exposure
`(42%) and a longer terminal half-life (30%) (Her-
`mann et al., 2000).
`In humans, RGB undergoes extensive and
`virtually exclusively phase II biotransformation
`and renal elimination. There is no indication that
`oxidative metabolic pathways, e.g. cytochrome
`P450 isozymes, may play any role in RGB
`biotransformation (Hempel et al., 1999). RGB is
`predominantly metabolized by N -glucuronidation,
`resulting in the formation of two distinct N -
`glucuronides, and to a considerably lesser extent
`by N -acetylation to form a pharmacologically
`active N -acetyl derivative.
`Recently, the specific UGT isozymes 1A1, 1A9,
`and to a lesser extent 1A4 were identified as the
`main isozymes involved in RGB major metabolic
`pathway (Hiller et al., 1999). The kidney contri-
`butes to the formation of both RGB N -glucur-
`onides, indicating that the metabolism of the drug
`is not exclusively confined to the liver. UGT1A1
`and 1A4 are also expressed in all parts of the
`gastrointestinal tract and UGT1A9 is expressed in
`the colon, which suggests that the gut may also
`contribute to the metabolism of RGB. The pre-
`sence of RGB’s major metabolic enzymes in
`various organs further suggest that the pharmaco-
`kinetics of the drug may be not be greatly altered
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 7
`
`

`
`38
`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`in patients with impaired liver function. Specific
`studies in this regard, however, are yet to be
`performed.
`Systemic cleavage of N -glucuronides with sub-
`sequent recycling of the parent drug has been
`suggested. After drug intake, a considerable frac-
`tion of the RGB dose is initially converted to the
`inactive N -glucuronides, with a subsequent gra-
`dual release of free parent drug from the N -
`glucuronide pool. This suggests the modeling of
`a favorable slow-release mechanism from endo-
`genous N -glucuronide sources.
`
`2.4.5. Interactions
`RGB shows no interaction with food (data on
`file) and does not alter the phramcokinetics of the
`oral contraceptive steroids ethinylestradiol/norges-
`trel (Hiller et al., 1999). Lack of pharmacokinetic
`interaction with phenobarbital
`(PB) has been
`shown, suggesting that no dosage adjustment is
`necessary when RGB and PB are coadministered.
`While the pharmacokinetics of RGB is not
`altered by LTG, LTG clearance appears to be
`slightly increased and LTG terminal half-life
`slightly decreased after multiple dosing with
`RGB.
`RGB pharmacokinetics is not altered by VPA,
`topiramate (TPM), PHT or CBZ, and RGB does
`not alter the pharmacokinetics of these drugs in
`epileptic patients. PHT and CBZ, however, mod-
`erately increase the clearance of RGB. Thus,
`increased RGB doses may be required when co-
`administered with PHT and/or CBZ, and caution
`should be taken when reducing or removing co-
`administered CBZ and PHT since RGB concen-
`trations may increase.
`
`2.4.6. Efficacy and tolerability data
`More than 200 patients aged 16 /64 years were
`treated in five phase IIa trials, and in long-
`term extension studies with daily doses of 600 /
`2400 mg. This phase IIa studies were of explora-
`tory character and were designed to assess the
`safety, tolerability, and preliminary efficacy of
`RGB.
`
`2.4.6.1. Efficacy. A confirmatory randomized,
`double-blind, placebo controlled, 4-parallel-group,
`
`multinational multicenter, phase IIb dose ranging
`study involved a total of 399 patients according to
`an add-on study design. This trial confirmed
`statistically significant superiority of 900 and
`1200 mg daily doses in the primary and secondary
`efficacy variables as compared to placebo. The
`median% changes
`in monthly
`total partial
`seizure rates from baseline were 13, 23, 29 and
`35% for placebo and for daily RGB doses of 600,
`900, and 1200 mg, respectively. Corresponding
`responder rates (% of patients showing at least
`50% seizure reduction), assessed as a secondary
`efficacy measure, were 16, 23, 32, and 33%,
`respectively.
`
`2.4.6.2. Adverse events. In patients with epilepsy,
`RGB has been administered as add-on therapy
`and as monotherapy using a gradual dose-titra-
`tion. The maximum tolerated dose (MTD) in
`patients was usually in the range from 1200 to
`1600 mg/day although some patients have toler-
`ated higher doses. The minimum daily dose
`considered to be effective ranged from 400 to 600
`mg (administered b.i.d.). It also appears that a
`t.i.d. dose regimen is equally or better tolerated
`compared with b.i.d. The most frequent adverse
`events that were reported in patients include
`asthenia, dizziness, headache, somnolence, tremor,
`speech disorder, amnesia, vertigo, abnormal think-
`ing (a COSTART/WHO preferred term used to
`indicate symptoms related to mental slowing, not
`psychosis), lack of coordination, nervousness, and
`paresthesia. With regard to cardiac safety, RGB is
`devoid of adverse effects on cardiac repolarization,
`such as prolongation of the QTc interval of the
`ECG.
`
`2.5. Safinamide
`
`R.G. Farielloa, C. Cattaneoa, S. Wischerb, R.
`Majb, F. Tergaub, W. Paulusb
`aNewron Pharmaceuticals Gerenzano, Varese,
`Italy and bDepartment of Clinical Neurophysiol-
`ogy University of Goettingen Medical School,
`Goettingen, Germany.
`
`Actavis v. Research Corp. Techs.
`IPR2014-01126
`RCT EX. 2015 page 8
`
`

`
`M. Bialer et al. / Epilepsy Research 51 (2002) 31 /71
`
`39
`
`via an electrode stereotactically implanted in the
`basolateral amygdala, both the electrical after-
`discharges and the behavioral component of the
`seizures were reduced more prominently by SAF
`than by PHT (Fariello et al., 2000).
`
`2.5.2.2. Behavioral pharmacology. SAF does not
`affect locomotor activity in rats until toxic doses
`are reached (700 mg/kg p.o.
`in rats, which
`correspond to 55 times the MES ED50). At a
`dose 40-fold higher than the ED50, SAF did not
`affect passive avoidance in mice.
`
`2.5.2.3. Mechanisms of action. SAF shows an IC50
`of 8.2 mM at the batrachotoxin sensitive site of the
`Na channel. In electrophysiological tests, safina-
`mide shows use- and frequency-dependent inhibi-
`effects on Na currents, with more
`tory
`pronounced effects compared with PHT, CBZ
`and LTG (Salvati et al., 1999). SAF inhibits
`sustained repetitive firing at concentrations of 3
`mM, without affecting the first action potentials of
`the burst. Safinamide also modulates Ca2 cur-
`rents of the L- and N-type. Perhaps as a con-
`sequence of these actions, safinamide inhibits in
`hippocampal slices glutamate release induced by
`addition of veratridine as well as by high mM K,
`a situation that more closely mimics pathological
`events during seizure activity.
`SAF has no affinity for noradrenaline, dopa-
`mine, serotonin, glutamate or GABA receptors
`(Ki/100 mM).
`SAF is a selective and reversible MAO B
`inhibitor and through this mechanism it increases
`neostriatal dopamine levels in primates, while
`decreasing the levels of dopamine metabolites.
`Other MAO inhibitors have demonstrated antic-
`onvulsant activity in rodent models of epilepsy and
`seizures (Loscher and Honack, 1995, Loscher and
`Lehmann, 1996). In addition, MAO B inhibitors
`reduce production of free radicals and may con-
`tribute to inhibit epileptogenesis, particularly fol-
`lowing post- traumatic or post hemorrhagic injury.
`
`2.5.3. Toxicology
`tolerated. Rats and
`SAF is generally well
`monkeys were chosen for acute and chronic
`studies. Six- and nine-month studies are approach-
`
`2.5.1. Introduction
`Safinamide (SAF; formerly NW-1015) is the S -
`isomer of the fluorobenzyloxy-benzylamino pro-
`panamide methansulfonate salt, which has been
`selected out of a medicinal chemistry program on
`new sodium channel blockers (Pevarello et al.,
`1998). Safinamide uniquely combines use and
`frequency-dependent blockade of Na channels,
`antagonism of N- and L-type Ca2 channels,
`inhibition of glutamate

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