throbber
Bhatti et of. BMC Veterinary Research (2015) 11:176
`DOI 10.1186/s12917-015-0464-z
`
`CORRESPONDENCE
`
`(3:c
`Veterinary Research
`
`Open Access
`
`International Veterinary Epilepsy Task Force gcms'
`consensus proposal: medical treatment of
`canine epilepsy in Europe
`
`Sofie F.M. Bhatti", Luisa De Risio2, Karen Munana3, Jacques Penderis4, Veronika M. Steins, Andrea TipoIds,
`Mette Berendt6, Robyn G. Farquhar', Andrea Fischer8, Sam Long9, Wolfgang Loscherm, Paul JJ. Mandigers",
`Kaspar Matiasek12, Akos Pakozdy13, Edward E. Patterson14, Simon Platt's, Michael Podell16, Heidrun Potschka17,
`Clare Rusbridge18''9 and Holger A. Volk2°
`
`Abstract
`
`In Europe, the number of antiepileptic drugs (AEDs) licensed for dogs has grown considerably over the last years.
`Nevertheless, the same questions remain, which include, 1) when to start treatment, 2) which drug is best used
`initially, 3) which adjunctive AED can be advised if treatment with the initial drug is unsatisfactory, and 4) when
`treatment changes should be considered. In this consensus proposal, an overview is given on the aim of AED
`treatment, when to start long-term treatment in canine epilepsy and which veterinary AEDs are currently in use for
`dogs. The consensus proposal for drug treatment protocols, 1) is based on current published evidence-based literature,
`2) considers the current legal framework of the cascade regulation for the prescription of veterinary drugs in Europe,
`and 3) reflects the authors' experience. With this paper it is aimed to provide a consensus for the management of
`canine idiopathic epilepsy. Furthermore, for the management of structural epilepsy AEDs are inevitable in addition
`to treating the underlying cause, if possible.
`
`Keywords: Dog, Epileptic seizure, Epilepsy, Treatment
`
`Background
`In Europe, the number of antiepileptic drugs (AEDs) li-
`censed for dogs has grown considerably over the last
`years. Nevertheless, the same questions remain, which
`include, 1) when to start treatment, 2) which drug is best
`used initially, 3) which adjunctive AED can be advised if
`treatment with the initial drug is unsatisfactory, and 4)
`when treatment changes should be considered. In this
`consensus proposal, an overview is given on the aim of
`AED treatment, when to start long-term treatment in
`canine epilepsy and which veterinary AEDs are currently
`in use for dogs. The consensus proposal for drug treatment
`protocols, 1) is based on current published evidence-based
`literature [17], 2) considers the current legal framework of
`the cascade regulation for the prescription of veterinary
`
`• Correspondence: sofie.bhatti@ugentbe
`'Department of Small Animal Medicine and Clinical Biology, Faculty of
`Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke 9820,
`Belgium
`Full list of author information is available at the end of the article
`
`drugs in Europe, and 3) reflects the authors' experience.
`With this paper it is aimed to provide a consensus for the
`management of canine idiopathic epilepsy. Furthermore,
`for the management of structural epilepsy AEDs are in-
`evitable in addition to treating the underlying cause, if
`possible.
`At present, there is no doubt that the administration
`of AEDs is the mainstay of therapy. In fact, the term
`AED is rather inappropriate as the mode of action of
`most AEDs is to suppress epileptic seizures, not epilep-
`togenesis or the pathophysiological mechanisms of epi-
`lepsy. Perhaps, in the future, the term anti-seizure drugs
`might be more applicable in veterinary neurology, a term
`that is increasingly used in human epilepsy. Additionally,
`it is known that epileptic seizure frequency appears to
`increase over time in a subpopulation of dogs with un-
`treated idiopathic epilepsy, reflecting the need of AED
`treatment in these patients [63].
`In our consensus proposal on classification and termin-
`ology we have defined idiopathic epilepsy as a disease in
`
`BiolVled Central
`
`O 2015 Bhatti et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution License
`(httcr.acreativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium,
`provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (hap://
`creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
`ARGENTUM Exhibit 1056
` Argentum Pharmaceuticals LLC v. Research Corporation Technologies, Inc.
`IPR2016-00204
`
`Page 00001
`
`(cid:9)
`

`
`Bhatti et al. WC Veterinary Research (2015) 11:176
`
`Page 2 of 16
`
`its own right, per se. A genetic origin of idiopathic epilepsy
`is supported by genetic testing (when available) and a gen-
`etic influence is supported by a high breed prevalence
`(>2 %), genealogical analysis and /or familial accumulation
`of epileptic individuals. However in the clinical setting
`idiopathic epilepsy remains most commonly a diagnosis of
`exclusion following diagnostic investigations for causes of
`reactive seizures and structural epilepsy.
`
`Aims of AED treatment
`The ideal goal of AED therapy is to balance the ability
`to eliminate epileptic seizures with the quality of life of
`the patient. Seizure eradication is often not likely in
`dogs. More realistic goals are to decrease seizure fre-
`quency, duration, severity and the total number of epi-
`leptic seizures that occur over a short time span, with
`no or limited and acceptable AED adverse effects to
`maximize the dog's and owner's quality of life. Clinicians
`should approach treatment using the following paradigm
`[23, 76, 91, 92,120]:
`
`- Decide when to start AED treatment
`- Choose the most appropriate AED and dosage
`- Know if and when to monitor serum AED
`concentrations and adjust treatment accordingly
`- Know when to add or change to a different AED
`- Promote pet owner compliance
`
`When to recommend maintenance AED treatment?
`Definitive, evidence-based data on when to start AED
`therapy in dogs based on seizure frequency and type is
`lacking. As such, extrapolation from human medicine may
`be possible to provide treatment guidelines. Clinicians
`should consider the general health of the patient, as well
`as the owner's lifestyle, financial limitations, and comfort
`with the proposed therapeutic regimen. Individualized
`therapy is paramount for choosing a treatment plan. As a
`general rule, the authors recommend initiation of long-
`term treatment in dogs with idiopathic epilepsy when any
`one of the following criteria is present:
`
`- Interictal period of < 6 months (i.e. 2 or more
`epileptic seizures within a 6 month period)
`- Status epilepticus or cluster seizures
`- The postictal signs are considered especially
`severe (e.g. aggression, blindness) or last longer
`than 24 hours
`- The epileptic seizure frequency and/or duration
`is increasing and/or seizure severity is
`deteriorating over 3 interictal periods
`
`In humans, the decision regarding when to recommend
`AED treatment is based on a number of risk factors
`(e.g. risk of recurrence, seizure type, tolerability, adverse
`
`effects) [42, 115]. In people, clear proof exists that there is
`no benefit initiating AED treatment after a single unpro-
`voked seizure [42], but there is evidence to support start-
`ing treatment after the second seizure [43, 108]. In dogs,
`long-term seizure management is thought to be most suc-
`cessful when appropriate AED therapy is started early in
`the course of the disease, especially in dogs with a high
`seizure density and in dog breeds known to suffer from a
`severe form of epilepsy [12-14]. A total number of z 10
`seizures during the first 6 months of the disease appeared
`to be correlated with a poor outcome in Australian Shep-
`herds with idiopathic epilepsy [132]. Furthermore, recent
`evidence exists that seizure density is a crucial risk factor,
`experiencing cluster seizures, and being male is associated
`with poor AED response [84].
`A strong correlation exists in epileptic people between
`a high seizure frequency prior to AED treatment and
`poor AED response [16, 34, 59]. Historically, this has
`been attributed to kindling, in which seizure activity
`leads to intensification of subsequent seizures [117].
`However, there is little clinical evidence that kindling
`plays a role in either dogs [54] or humans [111] with re-
`current seizures. In humans, a multifactorial pathogen-
`esis is suggested [14, 52]. Recent epidemiologic data
`suggest that there are differences in the intrinsic severity
`of epilepsy among individuals, and these differences in-
`fluence a patient's response to medication and long-term
`outcome. Additionally, evidence for seizure-associated
`alterations that affect the pharmacodynamics and
`pharmacokinetics of AEDs have been suggested [99].
`Breed-related differences in epilepsy severity have been
`described in dogs, with a moderate to severe clinical
`course reported in Australian Shepherds [132], Border
`Collies [49, 84], Italian Spinoni [24], German Shepherds
`and Staffordshire Bull Terriers [84], whereas a less se-
`vere form of the disease has been described in a different
`cohort of Collies (mainly rough coated) [77], Labrador
`Retrievers [7] and Belgian Shepherds [45]. Consequently,
`genetics may affect the success of treatment and may ex-
`plain why some breeds are more predisposed to drug re-
`sistant epilepsy [3, 77].
`
`Choice of AED therapy
`There are no evidence-based guidelines regarding the
`choice of AEDs in dogs. When choosing an AED for the
`management of epilepsy in dogs several factors need to
`be taken into account (AED-specific factors (e.g. regula-
`tory aspects, safety, tolerability, adverse effects, drug inter-
`actions, frequency of administration), dog-related factors
`(e.g. seizure type, frequency and aetiology, underlying
`pathologies such as kidney/hepatic/gastrointestinal prob-
`lems) and owner-related factors (e.g. lifestyle, financial cir-
`cumstances)) [23]. In the end, however, AED choice is
`often determined on a case-by-case basis.
`
`Page 00002
`
`(cid:9)
`

`
`Bhatti et al. 8MC Veterinary Research (2015) 11:176
`
`Page 3 of 16
`
`Until recently, primary treatment options for dogs
`with epilepsy have focused mainly on phenobarbital (PB)
`and potassium bromide (KBr) due to their long standing
`history, widespread availability, and low cost. While both
`AEDs are still widely used in veterinary practice, several
`newer AEDs approved for use in people are also being
`used for the management of canine idiopathic epilepsy
`mainly as add-on treatment. Moreover, since early 2013,
`imepitoin has been introduced in most European coun-
`tries for the management of recurrent single generalized
`epileptic seizures in dogs with idiopathic epilepsy.
`Several AEDs of the older generation approved for
`humans have been shown to be unsuitable for use in
`dogs as most have an elimination half-life that is too
`short to allow convenient dosing by owners, these in-
`clude phenytoin, carbamazepine, valproic acid, and etho-
`suximide [119]. Some are even toxic in dogs such as
`lamotrigine (the metabolite is cardiotoxic) [26, 136] and
`vigabatrin (associated with neurotoxicity and haemolytic
`anemia) [113, 131, 138].
`Since the 1990s, new AEDs with improved tolerability,
`fewer side effects and reduced drug interaction potential
`have been approved for the management of epilepsy in
`humans. Many of these novel drugs appear to be rela-
`tively safe in dogs, these include levetiracetam, zonisa-
`mide, felbamate, topiramate, gabapentin, and pregabalin.
`Pharmacokinetic studies on lacosamide [68] and rufina-
`mide [137] support the potential use of these drugs in
`dogs, but they have not been evaluated in the clinical
`setting. Although these newer drugs have gained consid-
`erable popularity in the management of canine epilepsy,
`scientific data on their safety and efficacy are very lim-
`ited and cost is often prohibitive.
`
`Phenobarbital
`Efficacy
`PB has the longest history of chronic use of all AEDs in
`veterinary medicine. After decades of use, it has been
`approved in 2009 for the prevention of seizures caused
`by generalized epilepsy in dogs. PB has a favourable
`pharmacokinetic profile and is relatively safe [2, 87, 97].
`PB seems to be effective in decreasing seizure frequency
`in approximately 60-93 % of dogs with idiopathic epi-
`lepsy when plasma concentrations are maintained within
`the therapeutic range of 25-35 mg/I [10, 31, 74, 105].
`According to Charalambous et al. (2014) [17], there is
`overall good evidence for recommending the use of PB
`as a monotherapy AED in dogs with idiopathic epilepsy.
`Moreover, the superior efficacy of PB was demonstrated
`in a randomized clinical trial comparing PB to bromide
`(Br) as first-line AED in dogs, in which 85 % of dogs ad-
`ministered PB became seizure-free for 6 months com-
`pared with 52 % of dogs administered Br [10]. This
`study demonstrated a higher efficacy of PB compared to
`
`Br as a monotherapy, providing better seizure control
`and showing fewer side effects.
`
`Pharmacokinetics
`PB is rapidly (within 2h) absorbed after oral administra-
`tion in dogs, with a reported bioavailability of approxi-
`mately 90 % [2, 87]. Peak serum concentrations are
`achieved approximately 4-8h after oral administration in
`dogs [2, 97]. The initial elimination half-life in normal
`dogs has been reported to range from 37-73h after mul-
`tiple oral dosing [96]. Plasma protein binding is approxi-
`mately 45 % in dogs [36]. PB crosses the placenta and
`can be teratogenic.
`PB is metabolized primarily by hepatic microsomal en-
`zymes and approximately 25 % is excreted unchanged in
`the urine. There is individual variability in PB absorption,
`excretion and elimination half-life [2, 87, 97]. In dogs, PB
`is a potent inducer of cytochrome P450 enzyme activity in
`the liver [48), and this significantly increases hepatic pro-
`duction of reactive oxygen species, thus increasing the risk
`of hepatic injury [107]. Therefore PB is contraindicated in
`dogs with hepatic dysfunction. The induction of cyto-
`chrome P450 activity in the liver can lead to autoinduction
`or accelerated clearance of itself over time, also known as
`metabolic tolerance, as well as endogenous compounds
`(such as thyroid hormones) [40, 48]. As a result, with
`chronic PB administration in dogs, its total body clearance
`increases and elimination half-life decreases progressively
`which stabilizes between 30-45 days after starting therapy
`[97]. This can result in reduction of PB serum concen-
`trations and therapeutic failure and therefore, monitor-
`ing of serum PB concentrations is very important for
`dose modulation over time.
`A parenteral form of PB is available for intramuscular
`(1M) or intravenous (IV) administration. Different PB
`formulations are available in different countries, it should
`be emphasized, however, that IM formulations cannot be
`used IV and vice versa. Parenteral administration of PB is
`useful for administering maintenance therapy in hospital-
`ized patients that are unable to take oral medication. The
`pharmacokinetics of IM PB have not been explored in
`dogs, however, studies in humans have shown a similar
`absorption after IM administration compared to oral ad-
`ministration [135]. The elimination half-life in dogs after a
`single IV dose is approximately 93h [87].
`
`Pharmacokinetic interactions
`In dogs, chronic PB administration can affect the dispos-
`ition of other co-administered medications which are me-
`tabolized by cytochrome P450 subfamilies and/or bound
`to plasma proteins [48]. PB can alter the pharmacokinetics
`and as a consequence may decrease the therapeutic ef-
`fect of other AEDs (levetiracetam, zonisamide, and ben-
`zodiazepines) as well as corticosteroids, cyclosporine,
`
`Page 00003
`
`(cid:9)
`

`
`Bhatti et al. BMC Veterinary Research (2015) 11:176
`
`Page 4 of 16
`
`metronidazole, voriconazole, digoxin, digitoxin, phenyl-
`butazone and some anaesthetics (e.g. thiopental) [23, 33,
`72, 82, 130]. As diazepam is used as first-line medicine for
`emergency use (e.g. status epilepticus) in practice it should
`be emphasized to double the IV or rectal dose of diazepam
`in dogs treated chronically with PB [130]. Concurrent ad-
`ministration of PB and medications that inhibit hepatic
`microsomal cytochrome P450 enzymes such as cimetidine,
`omeprazole, lansoprazole, chloramphenicol, trimethoprim,
`fluoroquinolones, tetracydines, ketoconazole, fluconazole,
`itraconazole, fluoxetine, felbamate and topiramate may
`inhibit PB metabolism, increase serum concentration and
`can result in toxicity [10].
`
`Common adverse effects
`Most of the adverse effects due to PB are dose dependent,
`occur early after treatment initiation or dose increase and
`generally disappear or decrease in the subsequent weeks due
`to development of pharmacokinetic and pharmacodynamic
`tolerance [35, 121] (Table 1). The adverse effects include
`sedation, ataxia, polyphagia, polydipsia and polyuria. For an
`in-depth review on the adverse effects of PB, the reader is
`referred to comprehensive book chapters [23, 32, 91].
`
`Idiosyncratic adverse effects
`These effects occur uncommonly in dogs and include
`hepatotoxicity [13, 22, 39, 75], haematologic abnormalities
`(anaemia, and/or thrombocytopenia, and/or neutropenia)
`[51, 56]), superficial necrolytic dermatitis [66], potential
`risk for pancreatitis [38, 46], dyskinesia [58], anxiety [58],
`and hypoalbuminaemia [41] (Table 1). Most of these
`idiosyncratic reactions are potentially reversible with dis-
`continuation of PB. For an in-depth review on the idiosyn-
`cratic adverse effects of PB the reader is referred to
`comprehensive book chapters [23, 32, 91].
`
`Laboratory changes
`Laboratory changes related to chronic PB administration in
`dogs include elevation in serum liver enzyme activities [39,
`41, 75], cholesterol and triglyceride concentrations [41]. Alter-
`ations in some endocrine function testing may occur (thyroid
`and adrenal function, pituitary-adrenal axis) [21, 41, 128]. For
`an in-depth review on these laboratory changes the reader is
`referred to comprehensive book chapters [23, 32, 91].
`
`Dose and monitoring (Fig. 1)
`The recommended oral starting dose of PB in dogs is
`25-3 mg/kg BID. Subsequently, the oral dosage is tailored
`
`Table 1 Most common reported adverse effects seen in dogs treated with PB, imepitoin and KBr (rarely reported and/or
`idiosyncratic adverse effects are indicated in grey
`AED
`
`Adverse effects in dogs
`
`PB
`
`Imepitoin
`
`KBr
`
`Sedation
`
`Ataxia
`
`Polyphagia
`
`Polydipsia/polyuria
`
`Hepatotoxicity
`
`Haematologic abnormalities
`
`Superficial necrolytic dermatitis
`
`Potential risk of pancreatitis
`
`Dyskinesia and anxiousness
`
`I I ypoalhuminaemie
`
`Polyphagia (often transient)
`Ilyperactivity. apathy, polyuria, polydipsia. hypersalivation, somnolence.
`
`vomiting, ataxia, apathy, diarrhoea, prolapsed nictitating membrane.
`
`decreased sight and sensitivity to sound
`Sedation
`
`Ataxia and pelvic limb weakness
`
`Polydipsia/polyuria
`
`Polyphagia
`
`Nausea, vomiting and/or diarrhea
`
`Personality changes (aggression, irritability, hyperactivity)
`
`Megancsophagus
`
`Persistent cough
`
`Increased risk of pancreatitis
`
`Page 00004
`
`(cid:9)
`(cid:9)
`

`
`Bhatti et al. 8MC Veterinary Research (2015)11:176
`
`Page 5 of 16
`
`Start PB 2.5 - 3 mg/kg BID
`
`Test serum drug concentrations 14 days after
`starting treatment or after a change in dose
`
`Adepsteselallit casiror
`
`rid seam rB < 33 sail
`
`Monitor complete blood cell
`count, biochemical profile, bile
`acid stimulation test, serum PB
`level at 3 months then every 6
`months. lithe dog is in
`remission or has no seizures:
`every 12 months.
`
`*Serum drug concentration < 30 mg/I increase PB dose using formula A
`*Serum drug concentration 30 - 35 mg/I start KBr therapy (see Figure 3)
`*Serum drug concentration > 35 mg/I reduce PB dose aiming for serum
`drug concentration < 35 mg/I and start KBr therapy (see Figure 3)
`
`Fig. 1 PB treatment flow diagram for decision making during seizure management in an otherwise healthy dog. The authors advise to start with
`PB (and add KBr if inadequate seizure control after optimal use of PB (Fig. 3)): in dogs with idiopathic epilepsy experiencing recurrent single
`generalised epileptic seizures; in dogs with idiopathic epilepsy experiencing cluster seizures or status epilepticus; in dogs with other epilepsy
`types. *Criteria for (in)adequate seizure control with regard to efficacy and tolerability (see Consensus proposal: Outcome of therapeutic
`interventions in canine and feline epilepsy (94)). 1. Treatment efficacious: a: Achievement of complete treatment success (i.e. seizure freedom or
`extension of the interseizure interval to three times the longest pretreatment interseizure interval and for a minimum of three months (ideally > 1
`year); b: Achievement of partial treatment success (i.e. a reduction in seizure frequency including information on seizure incidence (usually at least
`50 % or more reduction defines a drug responder), a reduction in seizure severity, or a reduction in frequency of seizure clusters and/or status
`epilepticus). 2. Treatment not tolerated i.e. appearance of severe adverse effects necessitating discontinuation of the AED
`
`to the individual patient based on seizure control, adverse
`effects and serum concentration monitoring.
`Because of considerable variability in the pharmaco-
`kinetics of PB among individuals, the serum concentra-
`tion should be measured 14 days after starting therapy
`(baseline concentration for future adjustments) or after
`a change in dose. To evaluate the effect of metabolic
`tolerance, a second PB serum concentration can be
`measured 6 weeks after initiation of therapy. Recom-
`mendations on optimal timing of blood collection for
`serum PB concentration monitoring in dogs vary among
`studies [23]. Generally, serum concentrations can be
`checked at any time in the dosing cycle as the change
`in PB concentrations through a daily dosing interval is
`not therapeutically relevant once steady-state has been
`achieved [62, 70]. However, in dogs receiving a dose of
`5 mg/kg BID or higher, trough concentrations were sig-
`nificantly lower than non-trough concentrations and
`serum PB concentration monitoring at the same time
`post-drug dosing was recommended, in order to allow
`accurate comparison of results in these dogs [70]. Another
`study recommended performing serum PB concentration
`monitoring on a trough sample as a significant difference
`between peak and trough PB concentration was identified
`in individual dogs [10]. The therapeutic range of PB in
`serum is 15 mg/1 to 40 mg/1 in dogs. However, it is the au-
`thors' opinion that in the majority of dogs a serum PB
`concentration between 25-30 mg/1 is required for optimal
`seizure control. Serum concentrations of more than 35
`mg/1 are associated with an increased risk of hepatotoxicity
`
`and should be avoided [22, 75]. In case of inadequate seiz-
`ure control, serum PB concentrations must be used to
`guide increases in drug dose. Dose adjustments can be cal-
`culated according to the following formula (Formula A):
`
`New PB total daily dosage in mg
`
`= (desired serum PB concentration/actual serum PB concentration)
`
`x actual PB total daily dosage in mg
`
`A dog with adequate seizure control, but serum drug
`concentrations below the reported therapeutic range,
`does not require alteration of the drug dose, as this
`serum concentration may be sufficient for that individ-
`ual. Generally, the desired serum AED concentration for
`individual patients should be the lowest possible concen-
`tration associated with >50 % reduction in seizure fre-
`quency or seizure-freedom and absence of intolerable
`adverse effects [23].
`In animals with cluster seizures, status epilepticus or
`high seizure frequency, PB can be administered at a
`loading dose of 15-20 mg/kg IV, IM or PO divided in
`multiple doses of 3-5 mg/kg over 24-48h to obtain a
`therapeutic brain concentration quickly and then sustain
`it [10]. Serum PB concentrations can be measured 1-3
`days after loading. Some authors load as soon as possible
`(over 40 to 60 min) and start with a loading dose of 10
`to 12 mg/kg IV followed by two further boluses of 4 to 6
`mg/kg 20 min apart.
`Complete blood cell count, biochemical profile (includ-
`ing cholesterol and triglycerides), and bile acid stimulation
`
`Page 00005
`
`(cid:9)
`

`
`Bhatti et al. BMC Veterinary Research (2015) 11:176
`
`Page 6 of 16
`
`test should be performed before starting PB treatment and
`periodically at 3 months and then every 6 months during
`treatment. In case of adequate seizure control, serum PB
`concentrations should be monitored every 6 months. If
`the dog is in remission or has no seizures, a periodical
`control every 12 months is advised.
`
`Imepitoin
`Efficacy
`Imepitoin was initially developed as a new AED for
`humans, but, the more favourable pharmacokinetic profile
`of imepitoin in dogs versus humans led to the decision to
`develop imepitoin for the treatment of canine idiopathic
`epilepsy [102]. Based on randomized controlled trials that
`demonstrated antiepileptic efficacy, high tolerability and
`safety in epileptic dogs, the drug was approved in 2013 for
`this indication in Europe [64, 98, 122]. It has been recom-
`mended to use imepitoin in dogs with idiopathic epilepsy
`experiencing recurrent single generalized epileptic sei-
`zures, however, its efficacy has not yet been demonstrated
`in dogs with cluster seizures or status epilepticus [30]. In a
`recent randomized controlled study [122], the efficacy of
`imepitoin was compared with PB in 226 client-owned
`dogs. The administration of imepitoin twice daily in incre-
`mental doses of 10, 20 or 30 mg/kg demonstrated that the
`majority of dogs with idiopathic epilepsy were managed
`successfully with imepitoin without significant difference
`to the efficacy of PB. The frequency of adverse events (e.g.
`sedation, polydipsia, polyphagia) was significantly higher
`in the PB group [122]. In a study by Rieck et al. (2006)
`[98], dogs with chronic epilepsy not responding to PB or
`primidone received imepitoin (in its initial formulation) or
`KBr as adjunct AED and the seizure frequency improved
`to a similar degree in both groups. According to Chara-
`lambous et al. (2014) [17], there is good evidence for
`recommending the use of imepitoin as monotherapy in
`dogs with recurrent single generalized epileptic seizures,
`but insufficient evidence for use as adjunct AED. At
`present, scientific data and evidence-based guidelines on
`which AED can best be combined with imepitoin are lack-
`ing, and further research is needed. Nevertheless, at this
`moment, the authors recommend the use of PB as adjunct
`AED in dogs receiving the maximum dose of imepitoin
`and experiencing poor seizure control. According to the
`authors, in case of combined therapy with imepitoin and
`PB, it is advised to slowly wean off imepitoin over several
`months if seizure control appears successful on PB and/or
`to reduce the dose of imepitoin if adverse effects (e.g. sed-
`ation) occur (Fig. 2).
`
`Pharmacokinetics
`Following oral administration of imepitoin at a dose of
`30 mg/kg in healthy Beagle dogs, high plasma levels
`were observed within 30 min, but maximal plasma levels
`
`were only reached after 2-3h following a prolonged ab-
`sorption time [101]. The elimination half-life was found
`to be short; approximately 1.5 to 2h. However, in an-
`other study in Beagle dogs, a longer half-life (-6 h) was
`found after higher doses of imepitoin, and accumulation
`of plasma levels was seen during chronic BID treatment
`[64]. Also, it has to be considered that Beagle dogs elim-
`inate AEDs more rapidly than other dog strains [122].
`Despite the short half-life in healthy Beagle dogs, this
`pharmacokinetic profile is reported as adequate to main-
`tain therapeutically active concentrations with twice
`daily dosing in dogs [64, 122]. Imepitoin is extensively
`metabolized in the liver prior to elimination. In dogs,
`imepitoin is mainly excreted via the faecal route rather
`than the urinary route. Neither reduced kidney function
`nor impaired liver function is likely to greatly influence
`the pharmacokinetics of imepitoin [122].
`
`Pharmacokinetic interactions and adverse reactions
`There is no information on pharmacokinetic interactions
`between imepitoin and other medications. Although,
`imepitoin is a low affinity partial agonist for the benzodi-
`azepine binding site of the GABAA receptor it has not
`prevented the pharmacological activity of full benzodi-
`azepine agonists such as diazepam in the clinical setting
`(e.g. in dogs with status epilepticus) [122). Consequently,
`because the affinity of diazepam for the GABAA receptor
`is much higher than imepitoin, care should be taken in
`the emergency setting [122]. Therefore, dogs with idio-
`pathic epilepsy treated with imepitoin and presented in
`status epilepticus might require, in addition to diazepam,
`an additional AED parenterally (e.g. PB, levetiracetam).
`Mild and most commonly transient adverse reactions
`(Table 1) have been reported in dogs administered 10-30
`mg/kg BID of imepitoin in its initial formulation; polypha-
`gia at the beginning of the treatment, hyperactivity,
`polyuria, polydipsia, somnolence, hypersalivation, em-
`esis, ataxia, lethargy, diarrhoea, prolapsed nictitating mem-
`branes, decreased vision and sensitivity to sound [64, 98].
`As part of the development of imepitoin for the treat-
`ment of canine epilepsy, a target animal safety study in
`dogs was conducted [96]. Under laboratory conditions,
`healthy Beagle dogs were exposed to high doses (up to
`150 mg/kg ql2h) of imepitoin for 6 months. Clinical
`signs of toxicity were mild and infrequent and they were
`mostly CNS (depression, transient ataxia) or gastrointes-
`tinal system (vomiting, body weight loss, salivation) re-
`lated. These clinical signs were not life-threatening and
`generally resolved within 24h if symptomatic treatment
`was given. These data indicate that imepitoin is a safe
`AED and is well tolerated up to high doses in dogs
`treated twice daily [96]. However, the safety of imepitoin
`has not been evaluated in dogs weighing less than 5 kg
`or in dogs with safety concerns such as renal, liver,
`
`Page 00006
`
`(cid:9)
`

`
`Bhatti et al. BMC Veterinary Research (2015) 11:176
`
`Page 7 of 16
`
`Start imepitoin 10 -20 mg/kg BID
`
`,111111011,
`
`
`
`Staarelaskliair
`
`Increase dose 30 mg/kg BID
`
`Adaqrshe mime ccarce
`
`Maintain dose and monitor dog
`clinically and neurologically +
`monitor complete blood cell
`count and biochemical profile
`every 6 months. If the dog is in
`remission or has no seizures:
`every 12 months.
`
`Add adjunct AED (Bather research
`necessary to decide which AED can best be
`used as adjunct ABD)N
`
`in case of combined therapy with imepitoin
`and PB, it is advised to slowly wean off
`imepitoin if seizure control appears successful
`on PB and /or to reduce the dose of imepitoin
`if adverse effects (e.g. sedation) occur.
`
`Fig. 2 Imepitoin treatment flow diagram for decision making during seizure management in an otherwise healthy dog. The authors advise to
`start with imepitoin in dogs with idiopathic epilepsy experiencing recurrent single generalised epileptic seizures. *Criteria for (in)adequate seizure
`control with regard to efficacy and tolerability (see Consensus proposal: Outcome of therapeutic interventions in canine and feline epilepsy (94)).
`1. Treatment efficacious: a: Achievement of complete treatment success (i.e. seizure freedom or extension of the interseizure interval to three
`times the longest pretreatment interseizure interval and for a minimum of three months (ideally > 1 year), b: Achievement of partial treatment
`success (i.e. a reduction in seizure frequency including information on seizure incidence (usually at least 50 % or more reduction defines a drug
`responder), a reduction in seizure severity, or a reduction in frequency of seizure clusters and/or status epilepticus). 2. Treatment not tolerated i.e.
`appearance of severe adverse effects necessitating discontinuation of the AED. 'Currently there are no data available on which AED should be
`added to imepitoin in case of inadequate seizure control. At this moment, the authors recommend the use of PB as adjunct AED in dogs
`receiving the maximum dose of imepitoin and experiencing poor seizure control
`
`cardiac, gastrointestinal or other disease. No idiosyncratic
`reactions have been demonstrated so far. The routinely
`measured liver enzymes' activity do not appear to be in-
`duced by imepitoin [96]. Compared with the traditional
`b

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