throbber
Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 1/19
`
`

`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 2/19
`
`

`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 3/19
`
`

`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 4/19
`
`

`
`Clinical Neuropharmacology
`Vol. 26, No. 1, pp. 38–52
`© 2003 Lippincott Williams & Wilkins, Inc., Philadelphia
`
`Clinical Pharmacology of Antiepileptic Drugs
`
`Carl W. Bazil and Timothy A. Pedley
`
`The Comprehensive Epilepsy Center, The Neurological Institute of New York, Columbia University, New York, New York, USA
`
`Antiepileptic drugs (AEDs) are the mainstay of treat-
`ment for the majority of patients with epilepsy. How-
`ever, not all patients with seizures require treatment
`with AEDs, and many patients with some forms of epi-
`lepsy, especially children, need only 1 or 2 years of
`drug therapy (1). Thus, important questions for the
`treating physician to have in mind are “When to start
`AEDs?” and “When can AEDs be safely withdrawn?”
`Nonetheless, it is safe to say that, as a group, patients
`with epilepsy typically receive one or more drugs, usu-
`ally for many years. Therefore, it is essential that phy-
`sicians have a good understanding of the clinical phar-
`macology of these agents, including their mechanisms
`of action, utilization patterns, and potential for interac-
`tions with other therapeutic agents.
`
`SELECTION OF ANTIEPILEPTIC DRUGS
`
`Table 1 lists the drugs currently used to manage dif-
`ferent types of seizures. Table 2 gives the usual dosage,
`effective plasma concentration, half-life, and common
`side effects for the most commonly used of these agents
`(2).
`Carbamazepine, phenytoin, primidone, and pheno-
`barbital are equally effective for partial and secondarily
`generalized seizures, although one may be effective
`when another is not (3). Valproate is also as effective
`for secondarily generalized seizures but is somewhat
`less effective than carbamazepine (and, presumably,
`phenytoin) in managing simple or complex partial sei-
`zures (4). Despite relatively equal antiepileptic po-
`tency, however, these drugs differ substantially in
`terms of side effects, pharmacokinetic properties, and
`cost. Similar comparative data are generally lacking for
`
`AEDs introduced after 1994. Phenytoin, with its rela-
`tively long half-life, which usually allows the drug to
`be given once or twice daily after midadolescence, has
`traditionally been preferred to drugs with shorter half-
`lives (5). However, concern about phenytoin’s occa-
`sional undesirable cosmetic effects (gingival hypertro-
`phy, hirsutism, and coarsening of facial features) and
`other adverse consequences associated with long-term
`therapy have led to the preferential use of carbamaze-
`pine or one of the newer drugs (e.g., lamotrigine, topi-
`ramate, zonisamide) as initial treatment in many pa-
`tients today. Carbamazepine, despite its short half-life,
`can be given twice daily if an extended-release formu-
`lation (e.g., Tegretol-XR, Carbatrol) is used. Phenobar-
`bital and primidone are rarely used now, except in spe-
`cial circumstances, because of the high incidence of se-
`dation and cognitive side effects at therapeutic doses
`and plasma concentrations (3).
`As a group, generalized-onset seizures respond best
`to valproate (6). Valproate can be used effectively as
`monotherapy in about 80% of patients, even when sev-
`eral types of generalized-onset seizure coexist. La-
`motrigine and topiramate are appropriate alternatives
`when valproate fails (7). A growing number of neurolo-
`gists prefer lamotrigine to valproate because of a more
`favorable adverse effect profile. Phenytoin and carba-
`mazepine are also effective against generalized tonic–
`clonic seizures, but the response is less reliable than
`with valproate (2). However, they are ineffective
`against absence or myoclonic seizures, which com-
`monly coexist with generalized tonic–clonic seizures,
`which means another drug (e.g., ethosuximide, clonaz-
`epam) must be used. Furthermore, phenytoin, gabapen-
`tin, and especially carbamazepine can actually aggra-
`vate nonconvulsive generalized-onset seizures (7).
`
`Most of the material contained in this review was presented at the 54th
`Annual Meeting of the American Academy of Neurology and was in-
`cluded in a syllabus prepared for the Therapy in Neurology course given
`on April 15, 2002. It is reproduced here with the permission of the
`American Academy of Neurology.
`Address correspondence and reprint requests to Dr. Timothy A.
`Pedley, 710 W. 168th St., The Neurological Institute of New York,
`Columbia University, New York, NY 10032, USA; E-mail: tap2@
`columbia.edu.
`
`MONOTHERAPY OR POLYTHERAPY?
`
`Medical treatment of epilepsy in adults is generally
`less satisfactory than in children, especially when con-
`fronted with partial and secondarily generalized sei-
`zures. Fewer than 50% of such patients with recurrent
`seizures remain seizure free for more than 12 consecu-
`
`38
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 5/19
`
`

`
`CLINICAL PHARMACOLOGY OF ANTIEPILEPTIC DRUGS
`
`39
`
`TABLE 1. AEDs used in treating different types of seizures
`
`Seizure type
`
`Effective drugs
`
`BASIC PRINCIPLES OF CLINICAL
`PHARMACOLOGY
`
`Partial seizures (including
`secondarily generalized)
`
`Broad spectrum (all seizure
`types, including partial,
`absence, myoclonic, tonic, and
`both primary and secondarily
`generalized tonic-clonic)
`Absence only
`Infantile spasms
`
`* not available in the U.S.
`
`carbamazepine, phenytoin,
`gabapentin, tiagabine,
`oxcarbazepine, phenobarbital,
`primidone, levetiracetam,
`zonisamide, topiramate,
`lamotrigine, valproic acid,
`felbamate
`valproic acid, lamotrigine,
`topiramate, felbamate,
`zonisamide, levetiracetam (?)
`
`ethosuximide
`valproic acid, vigabatrin*,
`zonisamide
`
`tive months (8). In the nationwide collaborative VA
`Cooperative Study, monotherapy resulted in satisfac-
`tory control of seizures in about 60% of patients. Of
`those in whom the first drug was ineffective, about 55%
`responded to an alternative drug used alone. Of the re-
`maining patients, only half had better control of sei-
`zures with two drugs. Moreover, improved control of
`seizures with two or more drugs was usually accompa-
`nied by increased side effects. These were frequently
`the result of drug interactions, which were not neces-
`sarily reflected in “toxic” blood concentrations of ei-
`ther drug. A more recent study by Kwan and Brodie
`included newer antiseizure drugs (7,8). Forty-seven
`percent of patients treated with a first drug became sei-
`zure free without adverse effects. About one third of
`patients who failed to respond to treatment subse-
`quently became seizure free on a second drug if the first
`had been discontinued because of side effects or idio-
`syncratic reactions. Seizure-free rates were not differ-
`ent between add-on and substitution therapies. These
`results, like the earlier studies, also support trying a sec-
`ond drug in monotherapy even when the first drug fails.
`
`Pharmacodynamics
`
`Following absorption, a drug distributes between the
`plasma and various tissue compartments (Fig. 1). Be-
`cause the majority of AEDs are at least partially bound
`to serum proteins, equilibrium exists between the con-
`centration of protein-bound drug and the unbound
`(“free”) drug concentration in plasma. The concentra-
`tion of free drug in plasma is, in turn, in equilibrium
`with the drug concentration in extracellular fluid. Only
`free drug is capable of crossing the various lipoprotein
`membranes that surround receptor sites and, therefore,
`only that portion of the total drug concentration that is
`free is available to produce the desired effect.
`It is not possible to monitor drug concentrations at
`receptor sites directly in humans, but total plasma drug
`concentrations (the usual clinical measurement) reflect
`the equilibrium among concentrations in tissue, extra-
`cellular fluid, and plasma (including that portion which
`is protein bound). Plasma free levels reflect concentra-
`tions in the extracellular space (9).
`The mechanism of action of a drug refers to the ac-
`tual biochemical or physical process that is modified or
`elicited to produce a biologic response at a particular
`site (pharmacodynamic effect). In the case of AEDs,
`this refers to the action produced at a cellular level lead-
`ing to decreased epileptic excitability. However, the ac-
`tual clinical utility of any AED is affected not only by
`its mechanism of action but also by its utilization pat-
`tern, any active metabolites that are formed, its perme-
`ability across the blood–brain barrier, and its therapeu-
`tic index.
`
`Pharmacokinetics
`
`The plasma concentration that is achieved and main-
`tained following the administration of a fixed drug dose
`
`Drug
`
`Phenytoin (Dilantin)
`Carbamazepine (Tegretol, Carbatrol)
`Phenobarbital
`Valproate (Depakote, Depakene)
`Ethosuxamide (Zarontin)
`Felbamate (Felbatol)
`Gabapentin (Neurontin)
`Lamotrigine (Lamictal)
`Topiramate (Topamax)
`Tiagabine (Gabitril)
`Oxcarbazepine (Trileptal)
`Levetiracetam (Keppra)
`Zonisamide (Zonegran)
`
`TABLE 2. Pharmacokinetics of AEDs
`
`Usual
`adult dose
`(mg/day)
`
`300–400
`800–1600
`90–180
`1000–3000
`750–1500
`2400–3600
`1800–3600
`100–500
`200–400
`32–56
`600–1800
`1000–3000
`100–400
`
`Half-life
`(h)
`
`22
`8–22
`100
`15–20
`60
`14–23
`5–7
`12–60b
`19–25
`5–13
`8–10c
`6–8
`63
`
`Metabolism
`
`>90% hepatic with induction
`>90% hepatic with induction
`>90% hepatic with induction
`>95% hepatic with inhibition
`65% hepatic, no induction
`60% hepatic
`>95% renal
`>90% hepatic, no induction
`30% hepatic, no induction
`>90% hepatic, no induction
`>90% hepatic, mild induction
`>65% renal excretion
`70% hepatic, no induction
`
`Usual effective plasma
`concentration
`(µg/mL)
`
`Time to peak
`concentration
`(h)
`
`Bound
`fraction
`(%)
`
`10–20
`8–12
`15–40
`50–120
`40–100
`20–140a
`4–16a
`2–16a
`4–10
`NE
`10–35c
`5–45a
`10–40a
`
`3–8
`4–8
`2–8
`3–8
`3–7
`2–6
`2–3
`2–5
`2–4
`1
`3–13
`1
`2–6
`
`90–95
`75
`45
`80–90
`<5
`25
`<5
`55
`9–17
`95
`40c
`<10
`40
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 6/19
`
`

`
`40
`
`C. W. BAZIL AND T. A. PEDLEY
`
`FIG. 1. Principles of antiepileptic
`drug monitoring. From C.E. Pippenger
`and Ronald P. Lesser. Current Trends
`in Epilepsy–Unit II, copyright Epilepsy
`Foundation of America. Reproduced
`with permission of the authors and the
`Epilepsy Foundation.
`
`mechanisms become saturated. It is important to note
`than even when the initial portion of the dose–response
`curve seems to be linear (as in Fig. 2B), drug clearance
`is altered throughout the dosage range and at all plasma
`concentrations. Kinetics in this portion of the range
`does not truly parallel the kinetics observed in a first-
`order relationship.
`Fortunately, in clinical practice, only a few drugs ex-
`hibit zero-order kinetics. Phenytoin is the most com-
`mon and important example of an AED exhibiting
`zero-order kinetics.
`
`(A) Dose-response curve for drug observing first-order
`FIG. 2.
`kinetics (linear). (B) Dose-response curve for drug observing
`zero-order kinetics (non-linear or saturation).
`
`is a direct consequence of the interactions of a wide
`variety of interrelated processes (Fig. 1), which, in turn,
`can be affected by a variety of factors, including other
`drugs, many illnesses (especially those affecting gas-
`trointestinal absorption, the liver, and kidneys), and
`physiologic changes (e.g., age, pregnancy).
`The study of these interrelationships forms the basis
`of pharmacokinetics. Pharmacokinetics is the study of
`the time-course of drug and metabolite concentrations
`in different fluids, tissues, and body wastes, and of the
`mathematical relations that can be utilized to develop
`models for interpretation of the blood concentrations
`measured in a given patient.
`The introduction and subsequent widespread avail-
`ability of reliable therapeutic drug monitoring has
`made application of pharmacokinetic principles an ex-
`pected and realistic part of contemporary medical
`therapeutics. An awareness of some of the fundamental
`principles is essential to rational prescribing practice
`and use of AEDs (10).
`
`Drug Kinetics
`
`First-order kinetics (line A in Fig. 2) describes a lin-
`ear relation between plasma drug concentration and to-
`tal daily dose (mg/kg). In other words, a predictable
`increase or decrease in plasma drug concentration oc-
`curs in response to a change in daily dosage.
`Zero-order kinetics (curve B in Fig. 2) describes a
`drug utilization process that is nonlinear, so that at
`some point plasma concentration (and clearance)
`seems to be independent of drug dose. Zero-order ki-
`netics becomes apparent when enzyme or transport
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 7/19
`
`

`
`CLINICAL PHARMACOLOGY OF ANTIEPILEPTIC DRUGS
`
`41
`
`Drug Half-Life
`
`Drug half-life (or elimination half-time) is the time
`required for elimination of half the concentration of a
`drug present in the system, provided no additional drug
`is administered after a specified point in time. The half-
`life of a drug may differ when it is administered to a
`naive patient compared with a patient taking other
`drugs. Drug half-life reflects the net interaction of sev-
`eral different processes that regulate drug clearance, of
`which rates of drug metabolism and excretion are the
`most important. Drug half-life determines the dosing
`interval for AEDs.
`
`Steady State
`
`When long-term oral therapy is initiated with a drug,
`it will continue to accumulate within the body until
`such time as rate of clearance, which comprises all tis-
`sue distribution, metabolic, and excretion processes in-
`volved in drug disposition, equals the rate of adminis-
`tration. When equilibrium is achieved between drug
`clearance and intake, the system is said to be at steady
`state. That is, the amount of drug ingested in a 24-hour
`period is equal to the amount of drug eliminated in the
`same 24-hour period. It requires seven half-lives of
`drug administration before a true steady-state concen-
`tration is achieved. For practical purposes, however,
`steady state is practically complete (97%) within five
`half-lives. Time to steady state determines how fast
`dose increases may be made for any AED.
`
`Absorption and Clearance
`
`Following oral administration, the type of drug
`preparation, drug solubility, concomitant administra-
`tion of other agents, whether the drug is taken with
`meals, or any gastrointestinal illness can all alter the
`amount of drug that will be absorbed from the stomach
`or small intestine. Time of maximum plasma drug con-
`centration (Tmax) is dependent on the rate of drug ab-
`sorption and elimination. Maximum concentration
`(Cmax) is the concentration at Tmax. The trough level is
`the concentration immediately preceding onset of ab-
`sorption of the next dose. Transient symptoms of neu-
`rotoxicity occurring at Tmax or seizures associated with
`trough levels of an AED may be verified by measuring
`the drug’s concentration at those times, and, if neces-
`sary, adjustments can be made in the timing or amount
`of a drug dose.
`Parenteral administration permits rapid entrance of a
`drug into the circulation and avoids problems associ-
`ated with drug absorption following oral administra-
`tion. Thus, parenteral administration is used when
`
`therapeutic blood levels need to be achieved rapidly
`and reliably, as, for example, in status epileptics.
`Clearance is a measurement of the amount of a drug
`eliminated during a period of time at steady state. If
`clearance (and ingestion) does not change, AED levels
`are relatively stable over time. This means that compli-
`ance can be estimated by obtaining serial measure-
`ments of plasma drug concentrations. Variation in se-
`quential measurements by more than 25% indicates the
`probability of noncompliance, if one has eliminated the
`possibility of intercurrent illness and use of other drugs.
`Some patients who have persistently low levels of an
`AED may have a genetic variation in hepatic metabo-
`lism (i.e., fast metabolizers). Low levels can also result
`from noncompliance (see preceding paragraph) or mal-
`absorption. Fast drug metabolism can be distinguished
`from malabsorption as a cause of persistently low
`plasma levels by measuring serial plasma drug concen-
`trations at given time intervals after parenteral admin-
`istration of the prescribed dose. If malabsorption is
`present, Cmax and observed drug half-life will be sig-
`nificantly higher following intravenous dosing than af-
`ter oral administration. On the other hand, if the patient
`problem is fast drug metabolism, there will be rela-
`tively little difference in plasma concentration or ob-
`served half-life regardless of the route of administra-
`tion. Fast drug metabolizers will require a larger daily
`dose (mg/kg) and often more frequent dosing to
`achieve the same serum concentration as a “normal”
`metabolizer. Conversely, someone who metabolizes
`drugs more slowly than usual will invariably exhibit
`drug toxicity at usual dosing schedules.
`
`Bioavailability
`
`Bioavailability is a measure of the amount of a dose
`absorbed by the body. Because of differences in manu-
`facturing processes, bioavailability of an active drug
`may vary from product to product. The majority of cur-
`rently available AEDs have narrow therapeutic ranges,
`and even small differences in bioavailability may be
`sufficient to produce toxicity or breakthrough seizures.
`Switching between dosage forms, especially in persons
`with poorly controlled seizures who have been titrated
`to their maximal tolerated dose, is usually not cost-
`effective because of the extra tests or clinic visits that
`are usually necessary.
`These and other factors that influence drug disposi-
`tion patterns in individual patients are listed in Table 3.
`Most of the factors influencing drug disposition and
`utilization in individual patients can be accounted for
`by appropriate therapeutic monitoring of AEDs. The
`indications for obtaining drug levels are summarized in
`Table 4.
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 8/19
`
`

`
`42
`
`C. W. BAZIL AND T. A. PEDLEY
`
`TABLE 3. Factors influencing drug disposition
`
`Patient compliance, including dosing error and wrong medication
`Absorption via route of administration
`Drug distribution
`Biotransformation
`Excretion
`Genetic variability
`Pathophysiologic factors (acute or chronic disease)
`Drug interactions
`Drug tolerance
`Inappropriate drug effects
`
`CHARACTERISTICS OF
`REPRESENTATIVE ANTIEPILEPTIC DRUGS
`
`Phenytoin
`
`Phenytoin (formerly diphenylhydantoin; Dilantin)
`was introduced clinically in 1938 following pioneering
`laboratory studies by Merritt and Putnam. For more
`than 60 years, phenytoin has been a drug of choice for
`management of partial and secondarily generalized
`tonic–clonic seizures.
`
`Administration and Bioavailability
`
`It is administered orally, preferably as the sodium
`salt in an extended release formulation. Approximately
`90% is absorbed after an oral dose, mainly from the
`duodenum, and peak concentrations are reached 6–12
`hours after administration. Maintenance dose is about
`4–6 mg/kg/day in adults; for children, doses of 8–10
`mg/kg/day are necessary. The common therapeutic
`range is 10–20 µg/mL.
`Parenteral phenytoin is available in a solution con-
`taining 50 mg/mL in 40% propylene glycol and 10%
`ethanol. The solution is highly alkaline (pH 12) and
`precipitates readily. It should be given by slow intrave-
`nous infusion, directly by the physician, at a rate not
`exceeding 50 mg/min. It should not be hung as an in-
`travenous solution, and Soluset or Volutrol dispensers
`should not be used. Phenytoin should never be given
`intramuscularly.
`
`TABLE 4.
`
`Indications for monitoring plasma drug levels
`
`To establish levels associated with optimal seizure control and/or
`development of toxicity for a drug with a narrow but well-defined
`therapeutic range (e.g., phenytoin, carbamazepine)
`When there is a discrepancy between the expected effect and the
`observed effect
`When non-compliance is suspected
`When interactions with other drugs is likely or suspected
`To compensate for changes in drug utilization caused by a secondary
`disease (e.g., uremia) or changing physiological state (e.g.
`pregnancy)
`When there is a need for medico-legal verification of treatment
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Fosphenytoin (Cerebyx) was introduced in 1996 as
`an alternative to parenteral phenytoin. Chemically, fos-
`phenytoin is a phosphate ester prodrug of phenytoin.
`Because it is highly water soluble, fosphenytoin may be
`administered in aqueous solutions; therefore, the side
`effect profile is improved compared with parenteral
`phenytoin. Administration is also more rapid; fosphe-
`nytoin may be administered at 150 mg phenytoin
`equivalents (PE) as opposed to a maximum rate of 50
`mg/min intravenous phenytoin. The conversion half-
`life is 8–15 minutes, which is independent of age. At
`these rates of administration, the two preparations give
`bioequivalent plasma free phenytoin concentrations.
`Unlike phenytoin, fosphenytoin may be administered
`intramuscularly. Plasma phenytoin generally reaches
`maximum about 30 minutes following intramuscular
`administration of fosphenytoin (12) (13).
`Dilantin brand phenytoin is available in 30-, 60-, and
`100-mg capsules, and in a 50-mg Infatab. The latter is
`not bioequivalent to the capsules.
`
`Metabolism and Pharmacokinetics
`
`Phenytoin is metabolized in the liver and undergoes
`parahydroxylation by the cytochrome P-450 system. Its
`metabolite, p-5-(hydroxyphenyl)-5-phenylhydantoin
`(HPPH), is inactive and nontoxic; it is excreted in the
`urine and bile conjugated with glucuronic acid. Less
`than 5% of phenytoin is excreted unchanged.
`Phenytoin is highly protein bound (>90%). Sub-
`stances that compete with it for binding sites or condi-
`tions that reduce its binding will increase the concen-
`tration of free drug.
`Fosphenytoin is more tightly bound to plasma pro-
`teins than is phenytoin. Therefore, patients who take
`phenytoin and are given a bolus of fosphenytoin may
`have a transient increase in free phenytoin as a result of
`displacement by fosphenytoin. This is unlikely to be
`clinically significant, however, as these patients will
`typically have low phenytoin levels, and any change
`will resolve as the fosphenytoin is converted into phe-
`nytoin.
`Use of phenytoin is complicated by its zero-order ki-
`netics. It is one of the few commonly used drugs that
`have substantially nonlinear elimination characteristics
`at therapeutic doses. There are several practical conse-
`quences. First, time to steady state concentration may
`be considerably longer than rough estimates based on a
`dose-independent half-life would suggest. For ex-
`ample, at concentrations greater than 20 µg/mL, half-
`life may be 36 hours or longer. Second, phenytoin’s
`steady state concentration at one dose does not directly
`predict the steady state concentration at another dose.
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 9/19
`
`

`
`CLINICAL PHARMACOLOGY OF ANTIEPILEPTIC DRUGS
`
`43
`
`This means that if a physician attempts to increase or
`decrease the steady state concentration by simple linear
`extrapolation from a known plasma level, the result is
`often an unexpectedly high (and possibly toxic) or low
`(and subtherapeutic) concentration. Furthermore, in
`any patient with toxic serum levels, one cannot reliably
`predict the time that will be required for the phenytoin
`concentration to decrease into the therapeutic range. Fi-
`nally, phenytoin’s nonlinear pharmacokinetics exag-
`gerates differences in bioavailability between the brand
`and generic preparations.
`
`Toxicity
`
`Neurotoxicity is dose related. Nystagmus appears
`first and is usually clinically insignificant. Higher
`plasma levels are associated with ataxia, dysarthria,
`diplopia, nausea, and a feeling of dysequilibrium.
`Higher levels still (usually greater than 35 µg/mL) are
`accompanied by mental dulling, drowsiness, and,
`rarely, a paradoxical increase in seizure frequency. Re-
`versible movement disorders, including chorea, dys-
`tonic, myoclonus, and asterixis, can occur, most often
`in patients with underlying brain damage. Cerebellar
`atrophy occurs with long-term phenytoin use, but the
`existence of clinically important chronic cerebellar
`damage remains controversial. Long-term phenytoin
`use can cause a peripheral neuropathy, but this is usu-
`ally clinically evident only as minor findings on exami-
`nation, such as depressed stretch reflexes and de-
`creased vibratory sensation in the feet.
`Limiting side effects, apart from rare idiosyncratic
`reactions, including Stevens-Johnson syndrome, are
`cosmetic: gingival hyperplasia, hirsutism, and coarsen-
`ing of facial features. These seem to occur most often in
`children, adolescents, and young adults. Hypocalcemia
`and osteomalacia may be seen as a result of phenytoin-
`induced alterations in vitamin D metabolism. Megalo-
`blastic anemia occurs from folate deficiency.
`
`Mechanism of Action
`
`At clinically useful concentrations, phenytoin inhib-
`its high-frequency repetitive firing by a use-dependent
`blockade of the voltage-dependent sodium channel. At
`somewhat higher concentrations (but still partially
`within the clinical range), phenytoin also blocks volt-
`age-dependent calcium channels, but the relevance of
`this to phenytoin’s anticonvulsant action is controver-
`sial. Phenomenologically, phenytoin is especially ef-
`fective in preventing seizure spread from a focus.
`
`Drug Interactions
`
`These are summarized later in a separate section.
`
`Carbamazepine
`
`Carbamazepine (Tegretol) is an iminostilbene, a
`dibenzepine derivative that is chemically and pharma-
`cologically related to tricyclic antidepressant agents.
`Its spectrum of action, clinically and in animal seizure
`models, is similar to that of phenytoin. Carbamazepine
`was approved for management of seizures in 1974, al-
`though it had been introduced a decade earlier for man-
`agement of trigeminal neuralgia (13).
`
`Administration and Bioavailability
`
`Carbamazepine is even less soluble in water than
`phenytoin, a factor that has inhibited development of a
`parenteral solution for clinical use. Therefore, at pres-
`ent, carbamazepine is available only for oral adminis-
`tration. It is available in 200- and 100-mg tablets and as
`an oral suspension (100 mg/5 mL).
`An extended release formulation, Tegretol XR, be-
`came available in the United States in 1996. It appears
`as a standard coated tablet but contains an osmotically
`active core with an opening on one side for release of
`drug. It is available in 100-, 200-, and 400-mg tablets.
`Since the tablet is specifically formulated for slow re-
`lease, these may not be crushed or cut. Although the
`drug is released, the outer core may not dissolve and
`can sometimes be seen in the stool.
`Carbamazepine is absorbed slowly and somewhat
`erratically, probably because of the slow dissolution
`rates of the compound. Tegretol brand is somewhat
`more predictable than the generic forms. Tmax for a
`single dose ranges from 4 to 16 hours, with peaks as late
`as 24 hours being reported occasionally. With long-
`term administration, peak plasma levels occur at 1.5
`hours with the oral suspension, at 4–5 hours with con-
`ventional tablets, and at 3–12 hours with the extended
`release formulation. Because of the lack of a parenteral
`formulation, precise bioavailability data are lacking;
`however, the XR tablets demonstrate an 89% bioavail-
`ability compared with the oral suspension and are
`equivalent to the conventional tablets.
`Maintenance dosage is usually about 10 mg/kg/day
`in adults and 15–20 mg/kg/day in young children. The
`usual therapeutic concentration is 8–12 µg/mL.
`
`Metabolism and Pharmacokinetics
`
`Once absorbed, carbamazepine is distributed rapidly
`and fairly uniformly to all body tissues. On average,
`about 75% of absorbed carbamazepine is protein
`bound.
`Carbamazepine is eliminated by biotransformation.
`Its major metabolite is carbamazepine 10,11-epoxide,
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Argentum Pharm. v. Research Corp. Techs., IPR2016-00204
`RCT EX. 2082 - 10/19
`
`

`
`44
`
`C. W. BAZIL AND T. A. PEDLEY
`
`which itself undergoes further metabolism. The epox-
`ide metabolite has weak anticonvulsant properties and
`is likely responsible for some of carbamazepine’s neu-
`rotoxicity. Less than 2% of carbamazepine is excreted
`unchanged.
`Carbamazepine’s clinical pharmacokinetic proper-
`ties are complicated by the phenomenon of autoinduc-
`tion of its own metabolism by hepatic microsomal en-
`zymes. In naive patients, introduction of the drug is as-
`sociated with a half-life of about 30 hours. Within days
`to a few weeks, the half-life decreases to 10–20 hours.
`This process stabilizes at about 30 days. The clinical
`effect of autoinduction means that introducing the drug
`can be tricky. Therapy should be initiated with low
`doses (100 mg twice daily) and slowly increased every
`3–5 days. Another regimen that works in most patients
`is to give an initial dose of 6–8 mg/kg followed by 25%
`of that dose on day 2, 50% on days 3 and 4, and 75% on
`days 5–7. The initial dosing regimen recommended in
`the PDR is almost guaranteed to produce toxicity in the
`majority of patients. Once autoinduction is complete,
`the relatively short and variable half-life requires fre-
`quent daily dosing (at least three times daily, although
`many patients require or do better on four-times-daily
`dosing schedules). Tegretol XR and Carbatrol can be
`administered twice daily.
`
`Toxicity
`
`An acute dysequilibrium syndrome, fatigue, and
`drowsiness occur when treatment is started too rapidly.
`Dose-related, reversible neurotoxicity includes blurred
`or double vision, nystagmus, dizziness, headache, and
`incoordination. Mild symptoms may appear early in the
`course of treatment during dose adjustment, but toler-
`ance may develop. Similar symptoms can occur tran-
`siently with peak plasma concentrations following a
`dose (Cmax). Dystonia and chorea occur rarely, usually
`in the setting of polypharmacy and brain damage, but
`less often than with phenytoin.
`Skin rashes are more common with carbamazepine
`(15%) than with phenytoin (8%). Life-threatening id-
`iosyncratic reactions (e.g., Stevens-Johnson syndrome)
`are very rare.
`Mild elevations of hepatic enzymes (SGOT, SGPT)
`occur in 5–10% of treated patients and are of no clinical
`significance. Such mild abnormalities neither predict
`nor predispose to hepatitis, which is a hypersensitivity
`reaction presumably mediated by immune factors.
`Similarly, about 25–30% of patients treated with car-
`bamazepine develop transient leukopenia, which per-
`sists in about 10%. This benign leukopenia has no clini-
`cal significance and does not correlate with the rare
`cases (only 30 reported) of aplastic anemia.
`
`Clin. Neuropharmacol., Vol. 26, No. 1, 2003
`
`Carbamazepine has antidiuretic effects that result in
`hyponatremia and water retention. This may reflect re-
`nal and hypothalamic effects (anti-ADH). Mild to mod-
`erate hyponatremia is generally unimportant, although
`it sometimes relates to patient complaints of dizziness,
`headache, and nausea. Severe hyponatremia (<122
`mEq/L) and water retention can exacerbate seizures or
`contribute to congestive heart failure in patients with
`compromised cardiac function.
`
`Mechanism of Action
`
`Like phenytoin, carbamazepine inhibits voltage-
`dependent sodium channels at clinically relevant con-
`centrations. Carbamazepine also has high affinity for
`adenosine receptors, but it is unlikely that adenosine-
`receptor binding accounts for its anticonvulsant activ-
`ity.
`
`Drug Interactions
`
`These are summarized later in a separate section.
`
`Valproate
`
`Anticonvulsant properties of valproate (available as
`valproic acid [Depakene] or divalproex sodium [Depa-
`kote]) were discovered accidentally when it was being
`used as a solvent in tests of other compounds being
`screened for anticonvulsant activity. The FDA ap-
`proved it for treatment of absence seizures in 1978, al-
`though valproate had been used widely in Europe for a
`decade before that. It has a broad spectrum of anticon-
`vulsant activity, being the drug of choice for all forms
`of primary generalized epilepsy and is also useful for
`partial and secondarily generalized seizures (14).
`
`Administration and Bioavailability
`
`Absorption is rapid with valproic acid, slower and
`more sustained with the enteric-coated preparation
`(Depakote) that is commonly prescribed today. Peak
`times range from 3 to 8 hours. Food delays but does not
`decrease the extent of absorption. Divalproex sodium is
`available in 250-mg and 500-mg tablets. The therapeu-
`tic range for plasma concentration is usually given as
`50–100 µg/mL, but, in fact, this varies considerably de-
`pending on many factors. Many patients, especially
`those on pol

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