throbber
REVIEWS
`
`New avenues for anti-epileptic
`drug discovery and development
`
`Wolfgang Löscher1,2, Henrik Klitgaard3, Roy E. Twyman4 and Dieter Schmidt5
`
`Abstract | Despite the introduction of over 15 third-generation anti-epileptic drugs, current
`medications fail to control seizures in 20–30% of patients. However, our understanding of
`the mechanisms mediating the development of epilepsy and the causes of drug resistance
`has grown substantially over the past decade, providing opportunities for the discovery and
`development of more efficacious anti-epileptic and anti-epileptogenic drugs. In this Review
`we discuss how previous preclinical models and clinical trial designs may have hampered
`the discovery of better treatments. We propose that future anti-epileptic drug development
`may be improved through a new joint endeavour between academia and the industry,
`through the identification and application of tools for new target-driven approaches,
`and through comparative preclinical proof-of-concept studies and innovative clinical
`trials designs.
`
`Epilepsy
`A chronic brain disorder that
`is characterized by partial
`or generalized spontaneous
`(unprovoked) recurrent
`epileptic seizures and,
`often, comorbidities such
`as anxiety and depression.
`
`1Department of
`Pharmacology, Toxicology
`and Pharmacy, University
`of Veterinary Medicine,
`Hannover 30559, Germany.
`2Center for Systems
`Neuroscience, Hannover
`30559, Germany.
`3UCB Pharma, Neurosciences
`Therapeutic Area,
`Braine‑l’Alleud 1420,
`Belgium.
`4Janssen Research &
`Development, Raritan,
`New Jersey 08869, USA.
`5Epilepsy Research Group,
`Berlin 14163, Germany.
`Correspondence to W.L. 
`e‑mail: wolfgang.loescher@
`tiho‑hannover.de
`doi:10.1038/nrd4126
`Published online
`20 September 2013
`
`Epilepsy is a life-shortening brain disorder affecting
`approximately 1% of the worldwide population1. Although
`repeated epileptic seizures are the clinical hallmark
`of epilepsy, the disease process (epileptogenesis) begins
`before the first seizure and may also lead to the progres-
`sion of epilepsy after the onset of seizures. Epilepsy is
`diverse, with over 15 different seizure types and over
`30 epilepsy syndromes2, and is associated with sub-
`stantial comorbidity, including depression, anxiety and
`increased mortality3.
`During the past three decades, the introduction of
`over 15 third-generation anti-epileptic drugs (AEDs) has
`provided physicians and patients with more options
`for the treatment of many types of seizures4. However,
`although approximately 70–80% of patients with new-
`onset epilepsy eventually enter remission with current
`AEDs, these medications fail to control seizures in
`20–30% of patients5,6. Furthermore, no AED has been
`shown to prevent the development of epilepsy in
`patients prior to the first seizure; these drugs seem to
`purely act to symptomatically suppress seizures once
`they occur7,8. For some AEDs, an anti-epileptogenic
`effect has actually been suggested in certain preclinical
`epilepsy models9,10, but this has not been proven in
`humans. Indeed, with the exception of traumatic brain
`injury7, none of the therapies found to be effective in
`preclinical studies has been adequately tested using an
`appropriately designed clinical trial in humans.
`Unfortunately, there are few aetiologically relevant
`animal models used in epilepsy research today that have
`
`been validated at the clinical level — a fact that obviously
`hampers clinical trial design using the appropriate
`patient population.In addition, there is no compelling
`evidence that third-generation AEDs are generally much
`better tolerated11–13. However, individual modern AEDs
`such as gabapentin (Neurontin; Pfizer) or levetiracetam
`(Keppra; UCB Pharma) cause fewer or no dermatological
`hypersensitivity reactions. Also, non-enzyme-inducing
`modern AEDs such as gabapentin or levetiracetam do
`not induce the drug interactions seen with older AEDs
`that have been reported to substantially lower the effi-
`cacy of other medications, including other AEDs given
`in combination14.
`AEDs are also unable to prevent or reverse the devel-
`opment of drug-resistant epilepsy, to treat comorbidities
`or to reduce the burden of disease in a holistic sense4.
`A particularly disquieting aspect of current epilepsy
`treatments is that we have not made substantial pro-
`gress in seizure control over the past 40–50 years since
`the introduction of carbamazepine and valproate to the
`market4,15.
`The consequences of the standstill in the development
`of more efficacious drugs for the treatment of epilepsy are
`several-fold. Patients and physicians are increasingly dis-
`appointed and have thus become less interested in using
`recently developed, pricier AEDs. Payers are hesitant to
`pay premium prices for drugs that do not differentiate
`from established low-cost generic medications, and the
`pharmaceutical industry is losing interest in developing
`novel compounds for epilepsy (BOX 1).
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 12 | OCTOBER 2013 | 757
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`MYLAN - EXHIBIT 1034
`
`

`
`R E V I E W S
`
`Box 1 | Business challenges and opportunities for anti-epileptic drug development
`
`In the 1990s, epilepsy presented an opportunity to enter a therapeutic space in which there was a good chance for return
`on investment. Drivers for this included a significant unmet need with few treatment options (especially for patients with
`refractory epilepsy), good potential for reimbursement at competitive pricing with few competitors in the field, as well as
`manageable technical and regulatory hurdles.
`The adjunctive or add‑on treatment paradigm in the clinical management of refractory epilepsy was well suited for
`bringing forward new agents to the market. The placebo‑controlled adjunctive model for evaluating the efficacy of a test
`compound in refractory patients established efficacy and tolerability at an early stage and could be performed using
`cost‑efficient short‑term clinical studies. Furthermore, following the introduction of felbamate (Felbatol; MedPointe)
`to the market, a new regulatory path existed for the clinical development and labelling of anti‑epileptic drugs (AEDs).
`Together, these commercial, scientific, technical and regulatory factors drove confidence and reduced the risk
`associated with developing and obtaining a value‑returning marketable product for epilepsy. This template provided
`an incentive for several companies to confidently invest in bringing new AEDs to the market.
`Loss of industry interest in AEDs
`Prior incentives for investment in AED development are now negatively balanced by the drug development challenges
`facing industry overall144–146. Payer reimbursement requires that future AEDs bring additional value or differentiation
`(principally an improvement in efficacy) to an already crowded, highly generic AED field. No AED to date has convincingly
`been demonstrated to be superior in efficacy to any other AED in adjunctive therapy for partial seizures, and
`differentiation by safety profile for new AEDs is not a principal component for optimizing pricing and reimbursement.
`New regulatory hurdles have also evolved over the past 15–20 years. A generally lower risk tolerance for new drugs and
`recent class labelling regarding safety signals (that is, suicide) have affected opportunities in non‑epilepsy indications
`and had an impact on the overall value proposition for AEDs. New AEDs can require commitments for long‑term safety
`data in a variety of age populations, and paediatric investigational plans necessitate the development and testing of
`new formulations in very young patients (babies who are ≥1 month old). Commercialization models indicate that the
`adjunctive indication alone for a marginally differentiated product is not adequate. Product promotion for additional
`uses requires those specific indications to be established in the label. A monotherapy indication can move an AED earlier
`into the epilepsy treatment paradigm. However, the approval of a monotherapy has so far required the prior approval
`of an adjunctive therapy and this causes a considerable time delay.
`Future business opportunities for AED development
`Interesting business cases seem to exist for the very disabling epilepsy syndromes — which are associated with an
`increased risk of premature death — such as infantile spasms and Lennox–Gastaut syndrome. These may present viable
`business opportunities for orphan indications, for which tax incentives are provided, investments are smaller and there
`is a potentially less demanding path for approval.
`Another more immediate business opportunity may involve the repurposing of drugs from other therapeutic areas that
`possess either relevant disease‑modifying properties for epilepsy or a novel mechanism of action that provides substantial
`synergistic efficacy against drug‑resistant epilepsy when combined with an existing AED therapy. This would markedly
`reduce the level of investment necessary for discovery and development, and also potentially lower the technical hurdles
`and regulatory data requirements, thereby improving the premises for a very positive business case.
`A substantial level of investment, beyond that required for traditional AED development, will be necessary for the future
`development of new AEDs that have evidence of superior efficacy against a relevant standard of care for the treatment
`of drug‑resistant epilepsy, or that have the ability to markedly alter the course or the prognosis of epilepsy. However, as these
`types of new epilepsy therapies address a major unmet medical need, they also offer a promising business case to drive
`incentive for future AED development.
`The figure illustrates a hypothetical investment example for the development of an AED: a new molecular entity
`(NME) transitions from discovery into clinical development to be ultimately approved for marketing authorization.
`From discovery, the lead molecule passes through late‑stage preclinical toxicology testing and chemistry scale‑up into
`clinical testing at a cost of US$10 million and a success rate of 70%. The NME passes through each stage with an overall
`success rate of about 5% at a total cost of $350 million. A key inflection point is at the Phase II stage prior to the most
`significant spending investment in Phase III. A reduction of risk at this stage can greatly influence the overall success
`rate and total expenditure for the development of an AED. Note that a cost‑effective proof‑of‑differentiation step
`early in Phase II can further reduce the investment risk, cost and time. Sales and marketing costs add to the investment
`and can be of a similar
`magnitude to the development
`costs. Following marketing
`approval, there are costs
`for sales and marketing,
`launch, sales force, Phase IV
`medical affairs studies and
`post‑marketing regulatory
`commitments. Investments
`in the initial monotherapy
`indication and an alternative
`non‑epilepsy indication could
`add up to approximately
`$50–250 million.
`
`Epileptogenesis
`The gradual process (also
`termed latent period) by
`which epilepsy develops in the
`normal brain following brain
`insults or gene mutations.
`
`Anti-epileptic drugs
`(AEDs). Also termed
`anti convulsant or anti-seizure
`drugs. Compounds that, when
`administered systemically in
`animal models or to patients,
`inhibit or control seizures that
`are associated with epilepsy
`or other conditions.
`
`Stage
`Preclinical
`Phase I
`NME
`success
`Chance of progressing
`
`70%
`success
`

`
`50%
`success
`
`Cost (millions of US$)
`
`$10
`
`+
`
`$15
`
`Time (years)
`
`2
`
`+
`
`1
`
`Phase II
`success
`
`Phase III
`success
`
`Regulatory
`success
`
`=
`
`Marketing
`approval
`
`35%
`success
`
`$93
`
`2
`

`
`+
`
`+
`
`50%
`success
`
`$225
`
`4
`

`
`+
`
`+
`

`
`+
`
`+
`
`80%
`success =
`
`Overall
`success
`rate: 5%
`
`$10
`
`1
`
`=
`
`=
`
`Total cost:
`$350 million
`
`Total time:
`10 years
`
`758 | OCTOBER 2013 | VOLUME 12
`
`Nature Reviews | Drug Discovery
` www.nature.com/reviews/drugdisc
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`

`
`R E V I E W S
`
`MES seizure test
`(Maximal electroshock seizure
`test). A model in which a short
`(0.2-second) transcorneal or
`transauricular application of a
`50 or 60 Hz electrical stimulus
`in rodents induces generalized
`tonic–clonic seizures that are
`mediated by brainstem
`structures.
`
`Pentylenetetrazole
`(PTZ). A chemical convulsant
`that, when administered
`systemically to rodents, induces
`characteristic myoclonic and
`clonic convulsions that
`are mediated by forebrain
`structures.
`
`Amygdala kindling
`Repeated administration of an
`initially subconvulsive electrical
`stimuli via a depth electrode in
`the amygdala, which induces
`seizures that progressively
`increase in severity and
`duration; once established, the
`increased susceptibility to the
`induction of kindled seizures is
`a permanent phenomenon.
`
`GAERS rat
`(Genetic absence epileptic rat
`from Strasbourg). A genetic
`rat model that displays
`characteristic 6–7 Hz
`spike-wave electrographic
`seizures and a pharmacological
`profile that is consistent with
`generalized absence epilepsy.
`
`6-Hz psychomotor
`seizure model
`A seizure model in which
`a prolonged (4-second)
`transcorneal application of a
`6-Hz electrical stimulus in mice
`induces limbic seizures that
`are characterized by a stun,
`vibrissae chomping, forelimb
`clonus and a Straub tail;
`these seizures are resistant
`to phenytoin and some
`other anti-epileptic drugs.
`
`Non-inferiority trial design
`A clinical trial design that
`determines whether a test
`compound is inferior to
`another compound; the lower
`limit (95% confidence
`interval) of a test compound’s
`treatment efficacy or
`effectiveness is to be
`compared to a preset lower
`boundary of efficacy or
`effectiveness relative to the
`adequate comparator’s
`point estimate of efficacy
`or effectiveness.
`
`In this Review we briefly examine the experimental
`and clinical strategies for AED discovery and develop-
`ment over the past few decades and discuss why these
`approaches may have failed to address unmet medical
`needs. We also outline the challenges for the pharma-
`ceutical industry that have had an impact on its attitude
`towards the discovery and development of AEDs. Given
`the serious unmet clinical needs in epilepsy treatment,
`we present new ideas on how to revitalize the pharma-
`cological and clinical development of better AEDs that
`could provide the foundation for a new, joint endeavour
`between academia and the industry.
`
`Previous AED discovery and development
`Until recently, the discovery and development of a new
`AED almost exclusively relied on preclinical testing in
`animal seizure models to establish anti-seizure efficacy
`prior to conducting clinical trials in humans16. This
`approach has been successful and crucially contributed
`to the development of numerous clinically effective
`AEDs4,17. Indeed, animal models with a similarly high
`predictive value do not exist for other central nervous
`system (CNS) disorders, such as bipolar disorders or
`migraine18.
`Since Merritt and Putnam19 first described the use
`of an electroshock seizure model to assess drugs for
`anti-seizure properties in 1937 (FIG. 1 (TIMELINE)), simple
`models of acute seizures — such as the MES seizure test
`and the subcutaneous pentylenetetrazole (PTZ) seizure
`test in mice and rats — have been widely used in AED
`discovery. These models were considered to be ideal
`for AED discovery, which necessitates the screening
`of large numbers of compounds; acute seizure models
`should therefore be easy to perform, time- and cost-
`efficient, and predictive of clinical activity. The rodent
`MES test created by Toman, Swinyard and Goodman20
`in 1946 is still the most commonly used first screen in
`the search for new AEDs and is quite effective in identi-
`fying drugs that block generalized tonic–clonic seizures
`in patients17. The MES test has also repeatedly been
`proposed to identify drugs that are active against partial
`seizures in patients, but this test failed to detect several
`AEDs (for example, levetiracetam and vigabatrin (Sabril;
`Lundbeck)) that are effective against partial seizures in
`patients; therefore, other models such as amygdala kindling
`are better for identifying anticonvulsant effects against
`partial seizures21.
`Following the report of Everett and Richards22 in
`1944 that the PTZ test can identify the anti-absence
`efficacy of AEDs, two simple animal models — the
`MES and PTZ tests — were thought to be sufficient
`for differentiating among AEDs with different clinical
`effects. This subsequently formed the basis for the pro-
`posal made by Swinyard and colleagues23,24 that the
`MES and subcutaneous PTZ tests in mice and rats be
`used as standard procedures for predicting the clinical
`anticonvulsant activity of investigational drugs (FIG. 1
`(TIMELINE)). However, because of false-positive and false-
`negative findings in these models, more complex chronic
`epilepsy models that were developed in the 1980s and
`1990s (FIG. 1 (TIMELINE)) have subsequently been included in
`
`later-stage screening to further characterize anti-epileptic
`efficacy — the most notable of these models being the
`kindling model and genetic models of epilepsy, such as
`the absence-epilepsy-prone GAERS rat. More recently, the
`6-Hz psychomotor seizure model in mice has been intro-
`duced for differentiating an investigational AED from
`existing AEDs. This model is resistant to some of the
`old AEDs and enables the screening of a large number of
`compounds17,25, which is not possible with more elaborate
`models such as the kindling model.
`
`Preclinical strategies. Three strategies have been used
`in AED discovery: first, the random, phenotypic
`screening of newly synthesized compounds of diverse
`structural categories with as yet unknown mechanisms;
`second, the structural variation of known AEDs; and
`third, hypothesis-driven, target-based drug design4,17,18.
`All three strategies have generated clinically useful
`AEDs but only very few AEDs have been identified
`by rational, target-based strategies. These have been
`based on previously presumed mechanisms of seizure
`generation: that is, impaired GABA (γ-aminobutyric
`acid)-ergic inhibition and increased glutamatergic
`excitation, resulting in AEDs that either potentiate
`GABA transmission (such as vigabatrin and tiagabine)
`or inhibit glutamate receptors (such as perampanel
`(Fycompa; Eisai))17. However, the old reductionistic
`view that seizures or epilepsy are due to an imbalance
`between GABAergic inhibition and glutamatergic exci-
`tation ignores the complexity of the alterations within
`these neurotransmitter systems in the brain of a patient
`suffer ing from epileptic seizures26.
`
`Clinical strategies. Marketing approval of new AEDs for
`the treatment of epilepsy has been routinely obtained by
`adjunctive therapy placebo-controlled Phase III trials
`in adult patients with refractory seizures27. In the 1960s
`and 1970s, when few AEDs were available4, the enrol-
`ment of patients into these trials was straightforward
`and the use of placebo treatments was deemed acceptable
`given the lack of alternative treatment options28. This
`clinical strategy was very successful and has resulted in
`over 15 new AEDs entering the market since the 1980s
`(TABLE 1). Many AEDs that are marketed for adjunc-
`tive treatment are subsequently tested in monotherapy
`trials in patients with either refractory or previously
`untreated epilepsy. Because regulatory guidelines for
`monotherapy approval differ between Europe and the
`United States, sponsors need to pursue two separate and
`costly development programmes. The mono therapy
`development paradigm currently used in Europe for
`new-onset epilepsy is the non-inferiority trial design,
`which establishes a preset limit for the allowed differ-
`ence in outcome between the test drug and a standard
`AED27. In the United States, the preferred develop-
`ment path is conversion to monotherapy in refractory
`patients using historical controls. These designs have
`demonstrated that several AEDs are efficacious as
`monotherapies, including levetiracetam and zonis-
`amide (Zonegran; Eisai) in Europe and lamotrigine
`(Lamictal-XR; GlaxoSmithKline) in the United States28.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 12 | OCTOBER 2013 | 759
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`

`
`R E V I E W S
`
`Timeline | Milestones in the development of animal models for AED discovery and development*
`
`Putnam and
`Merritt19; EST
`test (phenytoin)
`
`Swinyard23; MES plus
`PTZ tests (standard
`AED assay)
`
`Anticonvulsant Screening Project of
`the National Institute of Neurological
`Disorders and Stroke (NINDS) of the
`US National Institutes of Health
`(NIH)29,30; MES, PTZ and rotarod tests
`
`Vergnes et al.157;
`GAERS rat
`(spontaneous
`absences)
`
`Barton et al.25; 6-Hz
`model (first described
`by Toman in 1951)159
`
`1937
`
`1944
`
`1949
`
`1969
`
`1975
`
`1979
`
`1982
`
`1991
`
`2001
`
`Everett and
`Richards22; PTZ test
`(trimethadione)
`
`Goddard et al.155;
`kindling model
`(focal seizures)
`
`Ben-Ari et al.156;
`kainate-induced status
`epilepticus (SRS)
`
`Cavalheiro et al.158; pilocarpine-
`induced status epilepticus (SRS)
`
`Löscher and Rundfeldt148;
`phenytoin non-responders and
`responders in kindled rats
`
`AED, anti-epileptic drug; EST, electroshock threshold; GAERS, genetic absence epilepsy rat from Strasbourg; MES, maximal electroshock; PTZ, pentylenetetrazole;
`SRS, spontaneous recurrent seizures. *All animal models shown (except for the SRS models described by Ben-Ari et al.156,Vergnes et al.157 and Cavalheiro et al.158)
`are those in which seizures are electrically or chemically induced. All models, except for the EST method in cats described by Putnam and Merritt19, are still
`used in the development of new epilepsy therapies21. Various models are important for different purposes in epilepsy research21 and can be assigned to four
`major categories: first, acute seizure models in which single seizures are electrically or chemically induced in healthy, neurologically intact rodents, such as the
`MES, subcutaneous PTZ or 6-Hz tests; second, chronic seizure (or epilepsy) models in which single or multiple seizures are electrically or chemically induced in
`rodents with chronic brain alterations, such as amygdala kindling; third, genetic animal models with inborn chronic epilepsy, such as the GAERS rat (which is
`better suited than the PTZ test to identify drugs that are active against absence seizures); and fourth, chronic epilepsy models in which epilepsy with SRS is
`induced by brain insults, such as status epilepticus (for example, induced by pilocarpine or kainate) or traumatic brain injury21. The MES and subcutaneous PTZ
`tests, which were developed more than 60 years ago, have been widely used in the search for new AEDs but they obviously do not predict efficacy against
`difficult-to-treat (or pharmacoresistant) seizures4. Löscher and Rundfeldt148 were the first to describe a chronic model of pharmacoresistant seizures in which
`AED-resistant rats were selected from large groups of amygdala-kindled rats by repeated testing with phenytoin. Later, Löscher et al. also described the
`selection of AED-resistant subgroups of rats for post-status epilepticus models of temporal lobe epilepsy with SRS21,160.
`
`Limitations of previous strategies
`Despite the development of various new AEDs since the
`early 1990s, the available evidence indicates that there has
`been a failure to deliver drugs with improved efficacy4.
`What are the reasons for this apparent failure to dis-
`cover drugs that can effectively control drug-refractory
`seizures and comorbidities as well as prevent or modify
`the disease?
`
`Problems with preclinical models. Simple seizure models
`such as the MES and PTZ tests in rodents have been
`instrumental in the identification of most AED candi-
`dates. The advantages of such acute seizure models are
`their technical simplicity and the ability to screen large
`numbers of compounds. A disadvantage is that the sei-
`zures do not mirror epilepsy (that is, spontaneous seizure
`occurrence) and occur in ‘normal’, non-epileptic brains.
`Furthermore, older AEDs provide complete seizure sup-
`pression in these tests, hampering the identification of
`new AED candidates with greater efficacy, including
`those that might be effective in patients who are resistant
`to the older drugs.
`More recently, large AED screening programmes
`such as the Anticonvulsant Screening Project (ASP) of
`the National Institute of Neurological Disorders and
`Stroke (NINDS) of the US National Institutes of Health
`(NIH), which was initiated in 1975 to stimulate the dis-
`covery and development of new chemical entities for
`the symptomatic treatment of human epilepsy29,30, have
`included models for pharmacoresistant partial seizures
`in drug screening. One particular model is the 6-Hz
`mouse test, which was also introduced to avoid missing
`
`the identification of compounds like levetiracetam;
`levetiracetam is ineffective in the MES and PTZ models
`but is among the most effective AEDs in the clinic16,25,31.
`However, although several novel AEDs — including bri-
`varacetam, retigabine (Potiga; Valeant Pharmaceuticals/
`GlaxoSmithKline) and carisbamate — are highly effective
`in the 6-Hz mouse model, they are not more effective in
`patients with pharmaco resistant partial seizures21.
`Thus, it seems that the simple acute seizure screening
`models used in the ASP and other programmes fail
`to differentiate between compounds with promising
`potential for efficacy against drug-resistant seizures and
`compounds that work through mechanisms that are not
`detected by these models. Importantly, chronic seizure
`models, such as the lamotrigine-resistant kindled rat32,
`in which seizures are induced in animals with chronic
`brain alterations, were therefore recently included in the
`ASP. However, none of the emerging models of therapy-
`resistant epilepsy (FIG. 1 (TIMELINE)) has actually been
`validated at predicting clinical success in the therapy-
`resistant patient population. Thus, it remains to be estab-
`lished whether the use of chronic models such as kindling
`or models with spontaneous recurrent seizures will lead
`to the identification of more effective anti-epileptic treat-
`ments, but we consider this approach to be much more
`viable than the exclusive use of simple acute seizure
`models, particularly when testing hypothesis-driven,
`target-based strategies of drug development21.
`
`Problems with broad-spectrum approaches. An impor-
`tant aim of previous research and development (R&D)
`efforts was to discover novel AEDs that exert a broad
`
`760 | OCTOBER 2013 | VOLUME 12
`
` www.nature.com/reviews/drugdisc
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`

`
`R E V I E W S
`
`Table 1 | Characteristics of clinically approved AEDs*
`AED
`Companies
`Year of
`Presumed main
`approval
`mechanisms of
`action
`
`Approved indications Main utility
`
`Main limitations
`
`First-generation drugs
`Potassium
`Dow
`bromide
`
`1857‡
`
`GABA potentiation?
`
`GTCS, myoclonic
`seizures
`
`Broad use for focal
`and generalized
`seizures
`
`Phenobarbital
`
`Bayer
`
`1912‡
`
`GABA potentiation
`
`Phenytoin
`
`Parke-Davis/
`Pfizer
`
`1938
`
`Sodium channel
`blocker
`
`PGCS, sedation,
`anxiety disorders, sleep
`disorders
`PGCS
`
`Broad use for focal
`and generalized
`seizures
`First-line AED, i.v. use
`
`Trimethadione
`
`Abbott
`
`1946
`
`T-type calcium
`channel blocker
`
`Absence seizures
`
`Rare use for absence
`seizures
`
`Primidone
`
`Imperial
`Chemical
`Industries
`
`1954
`
`GABA potentiation
`
`PGCS
`
`Broad use for focal
`and generalized
`seizures
`
`Ethosuximide
`
`Parke-Davis/
`Pfizer
`
`1958
`
`T-type calcium
`channel blocker
`
`Absence seizures
`
`First-line AED, no skin
`hypersensitivity
`
`Second-generation drugs
`Diazepam
`Roche
`
`1963
`
`GABA potentiation
`
`Convulsive disorders,
`status epilepticus,
`anxiety, alcohol
`withdrawal
`
`Broad use for focal and
`generalized seizures,
`i.v. use, no clinical
`hepatotoxicity, no skin
`hypersensitivity
`
`Carbamazepine Novartis
`
`1964
`
`Sodium channel
`blockade
`
`PGCS, trigeminal pain,
`bipolar disorder
`
`First-line AED
`
`Valproate
`
`Sanofi/Abbott
`
`1967
`
`Clonazepam
`
`Roche
`
`1968
`
`Multiple (for example,
`GABA potentiation,
`glutamate (NMDA)
`inhibition, sodium
`channel and T-type
`calcium channel
`blockade)
`GABA potentiation
`
`Clobazam
`
`Hoechst Roussel/
`Lundbeck/Sanofi
`
`1975
`
`GABA potentiation
`
`PGCS, absence
`seizures, migraine
`prophylaxis, bipolar
`disorder
`
`Broad use for focal
`and generalized
`seizures, first-line
`AED, i.v. use, no skin
`hypersensitivity
`
`Lennox–Gastaut
`syndrome, myoclonic
`seizures, panic
`disorders
`
`Broad use for focal
`and generalized
`seizures, no clinical
`hepatotoxicity
`
`Lennox–Gastaut
`syndrome, anxiety
`disorders
`
`Broad use for focal
`and generalized
`seizures, no clinical
`hepatotoxicity
`
`Currently for
`adjunctive use only,
`not in wide use
`anymore; acts as a
`sedative
`Enzyme inducer;
`skin hypersensitivity;
`no absence seizures
`Enzyme
`inducer; skin
`hypersensitivity;
`NLPK; not useful
`for absence or
`myoclonic seizures
`Not in wide
`use anymore;
`teratogenic
`Enzyme
`inducer; skin
`hypersensitivity; no
`absence seizures;
`acts as a sedative
`Somnolence, loss
`of appetite, nausea,
`vomiting, singultus,
`depression,
`psychotic episodes,
`insomnia, rare
`aplastic anaemia
`
`Currently for
`adjunctive use
`only; emergency
`use only; acts as a
`sedative; leads to
`tolerance (loss of
`efficacy)
`Enzyme
`inducer; skin
`hypersensitivity;
`not useful for
`absence or
`myoclonic seizures
`Enzyme inhibitor;
`substantial
`teratogenicity;
`weight gain
`
`Currently for
`adjunctive use only;
`acts as a sedative;
`leads to tolerance
`(loss of efficacy)
`Currently for
`adjunctive use only;
`acts as a sedative;
`leads to tolerance
`(loss of efficacy)
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 12 | OCTOBER 2013 | 761
`
`© 2013 Macmillan Publishers Limited. All rights reserved
`
`

`
`R E V I E W S
`
`Table 1 (cont.) | Characteristics of clinically approved AEDs*
`AED
`Companies
`Year of
`Presumed main
`approval
`mechanisms of
`action
`
`Approved
`indications
`
`Main utility
`
`Main limitations
`
`Third-generation drugs
`Progabide
`Synthelabo
`
`1985
`
`GABA potentiation
`
`Vigabatrin
`
`Sanofi/Lundbeck
`
`1989
`
`GABA potentiation
`
`Lamotrigine
`
`GlaxoSmithKline
`
`1990
`
`Oxcarbazepine Novartis
`
`1990
`
`Sodium channel
`blocker
`
`Sodium channel
`blocker
`
`PGCS, Lennox–Gastaut
`syndrome, myoclonic
`seizures, muscle
`hypertonia
`Infantile spasms,
`complex partial
`seizures (currently for
`adjunctive use only)
`
`Rarely used for focal
`seizures
`
`No clinical
`hepatotoxicity
`
`PGCS, Lennox–Gastaut
`syndrome, bipolar
`disorder
`Partial seizures
`
`Broad use for focal
`and generalized
`seizures, first-line AED
`First-line AED
`
`Felbamate
`
`Carter-Wallace/
`MedPointe
`Pharmaceuticals
`
`1993
`
`Multiple (GABA
`potentiation,
`glutamate (NMDA)
`inhibition, sodium
`and calcium channel
`blockade)
`
`PGCS, Lennox–Gastaut
`syndrome
`
`Broad use for focal
`and generalized
`seizures
`
`Gabapentin
`
`Parke-Davis/
`Pfizer
`
`1993
`
`Calcium channel
`blocker (α2δ subunit)
`
`PGCS, postherpetic
`and diabetic neuralgia,
`restless legs syndrome
`
`No clinical
`hepatotoxicity
`
`Topiramate
`
`Janssen/Johnson
`& Johnson
`
`1995
`
`Tiagabine
`
`Novo Nordisk
`
`1996
`
`Multiple (GABA
`potentiation,
`glutamate (AMPA)
`inhibition, sodium
`and calcium
`channel blockade)
`GABA potentiation
`
`PGCS, Lennox–Gastaut
`syndrome, migraine
`prophylaxis
`
`Partial seizures
`
`Levetiracetam
`
`UCB Pharma
`
`2000
`
`SV2A modulation
`
`PGCS, partial seizures,
`GTCS, JME
`
`Zonisamide
`
`Elan/Eisai
`
`2000
`
`Sodium channel
`blocker
`
`Partial seizures
`
`Stiripentol
`
`Biocodex
`
`2002
`
`Pregabalin
`
`Pfizer
`
`2004
`
`GABA potentiation,
`sodium channel
`blocker
`Calcium channel
`blocker (α2δ subunit)
`
`Dravet syndrome
`
`Partial seizures,
`neuropathic pain,
`generalized
`anxiety disorder,
`fibromyalgia
`
`Broad use for focal
`and generalized
`seizures, first-line
`AED, no clinical
`hepatotoxicity
`
`No clinical
`hepatotoxicity
`
`First-line AED, i.v.
`use, no clinical
`hepatotoxicity
`Broad use for focal
`and generalized
`seizures, no clinical
`hepatotoxicity
`No clinical
`hepatotoxicity
`
`No clinical
`hepatotoxicity
`
`Clinical
`hepatotoxicity,
`not in wide use
`anymore
`Not useful for
`absence or
`myoclonic seizures;
`vision loss;
`weight gain
`Enzyme
`inducer; skin
`hypersensitivity
`Enzyme
`inducer; skin
`hypersensitivity;
`not useful for
`absence or
`myoclonic seizures
`Currently for
`adjunctive use
`only; aplastic
`anaemia; clinical
`hepatotoxicity; skin
`hypersensitivity;
`clinical
`hepatotoxicity;
`not in wide use
`anymore
`Currently for
`adjunctive use only;
`weight gain; not
`useful for absence
`or myoclonic
`seizures
`Somno

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