throbber
Acta Neurol Scand 2008: 118: 69–86 DOI: 10.1111/j.1600-0404.2008.01004.x
`
`Copyright Ó 2008 The Authors
`Journal compilation Ó 2008 Blackwell Munksgaard
`ACTA NEUROLOGICA
`SCANDINAVICA
`
`Review Article
`Benzodiazepines in epilepsy: pharmacology
`and pharmacokinetics
`
`Riss J, Cloyd J, Gates J, Collins S. Benzodiazepines in epilepsy:
`pharmacology and pharmacokinetics.
`Acta Neurol Scand 2008: 118: 69–86.
`Ó 2008 The Authors Journal compilation Ó 2008 Blackwell Munksgaard.
`
`Benzodiazepines (BZDs) remain important agents in the management
`of epilepsy. They are drugs of first choice for status epilepticus and
`seizures associated with post-anoxic insult and are also frequently used
`in the treatment of febrile, acute repetitive and alcohol withdrawal
`seizures. Clinical advantages of these drugs include rapid onset of
`action, high efficacy rates and minimal toxicity. Benzodiazepines are
`used in a variety of clinical situations because they have a broad
`spectrum of clinical activity and can be administered via several routes.
`Potential shortcomings of BZDs include tolerance, withdrawal
`symptoms, adverse events, such as cognitive impairment and sedation,
`and drug interactions. Benzodiazepines differ in their pharmacologic
`effects and pharmacokinetic profiles, which dictate how the drugs are
`used. Among the approximately 35 BZDs available, a select few are
`used for the management of seizures and epilepsy: clobazam,
`clonazepam, clorazepate, diazepam, lorazepam and midazolam.
`Among these BZDs, clorazepate has a unique profile that includes a
`long half-life of its active metabolite and slow onset of tolerance.
`Additionally, the pharmacokinetic characteristics of clorazepate
`(particularly the sustained-release formulation) could theoretically
`help minimize adverse events. However, larger, controlled studies of
`clorazepate are needed to further examine its role in the treatment of
`patients with epilepsy.
`
`J. Riss1, J. Cloyd1, J. Gates2,
`S. Collins3
`1Center for Orphan Drug Research, Department of
`Experimental and Clinical Pharmacology, College of
`Pharmacy, University of Minnesota, Minneapolis, MN,
`USA; 2Minnesota Epilepsy Groupâ, St Paul, MN, USA;
`3Ovation Pharmaceuticals, Inc., Deerfield, IL, USA
`
`Key words: benzodiazepines; epilepsy;
`pharmacokinetics; pharmacology; clorazepate
`
`James Cloyd, PharmD, McGuire Translational Research
`Facility, 2001 6th Street SE, Minneapolis, MN 55455,
`USA
`Tel.: +1 612 624 4609
`Fax: +1 612 626 9985
`e-mail: cloyd001@umn.edu
`
`Accepted for publication January 28, 2008
`
`Introduction
`
`Much attention has been focused on the introduc-
`tion of new antiepileptic drugs (AEDs) into the US
`market during the past 15 years. Nonetheless,
`benzodiazepines (BZDs), which have been used
`since the 1960s, remain important
`in epilepsy
`management and are the drugs of first choice for
`status epilepticus and seizures associated with post-
`anoxic insult. Benzodiazepines also continue to
`play major roles in treating other conditions such
`as febrile seizures, acute repetitive seizures and
`alcohol withdrawal seizures. The major clinical
`advantages of BZDs are high efficacy rates, rapid
`onset of action and minimal toxicity. Few other
`drugs possess comparable attributes.
`
`All BZDs share similar neuropharmacologic
`properties including anxiety reduction, sedation,
`sleep induction, anticonvulsant effects and muscle
`relaxation (1). There are, however, differences
`among BZDs in affinity for receptor subtypes,
`which may produce different pharmacologic
`effects. Thus, some BZDs are more effective than
`others as anticonvulsants; few of the approxi-
`mately 35 BZDs available worldwide (2) are used
`for managing epilepsy. Diazepam and lorazepam
`are primarily used for management of seizure
`emergencies, whereas clobazam, clonazepam and
`clorazepate are commonly used in chronic epilepsy
`management. Midazolam often is used as an
`alternative to diazepam and lorazepam in seizure
`emergencies and for treating refractory status
`
`69
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`Riss et al.
`
`epilepticus. The BZDs also have widely varying
`pharmacokinetic profiles, with differences
`in
`absorption, onset and duration of action and
`formation of active metabolites. Thus, pharmaco-
`kinetic differences often dictate the use of specific
`BZDs, route(s) of administration and formula-
`tion(s). Additionally, the nature and importance of
`side effects and drug interactions have been iden-
`tified and clarified in recent years.
`The purpose of
`this review was to provide
`clinicians with information for selecting BZDs
`and for managing BZD therapy in their patients.
`The article considers BZD pharmacology, phar-
`macokinetics, use in epilepsy management, toler-
`ance and withdrawal. Also included in this review
`is an analysis and discussion of the effects of missed
`daily doses of immediate- and extended-release
`plasma N-des-
`clorazepate
`formulations
`on
`methyldiazepam (DMD) concentrations.
`
`Benzodiazepine pharmacology
`
`There are three principal c-aminobutyric acid
`(GABA) receptor subtypes. Ligand-activated ion
`channels that are selectively blocked by bicuculline
`and modulated by steroids, BZDs, and barbitu-
`rates are known as GABAA receptors (3). The
`second receptor subtype, GABAB, consists of
`G-protein-coupled,
`seven-transmembrane recep-
`tors, which are selectively activated by (R)-())-
`baclofen and 3-aminopropylphosphinic acid and
`are blocked by phaclofen (3). Transmitter-gated
`chloride channels, GABAC receptors, are selec-
`tively
`activated by
`certain conformationally
`restricted GABA analogs and are not modulated
`by steroids, BZDs or barbiturates (3).
`Benzodiazepines bind to GABAA receptors,
`ionotropic transmembrane proteins located in the
`neuronal membranes of
`the
`central nervous
`system (CNS) (3). The GABAA receptor consists
`of a pentameric structure with multiple subunits
`that are necessary for normal physiologic func-
`tion. The receptor subunits are assembled from
`combinations of 19 polypeptides (i.e. a1–6, b1–3,
`c1–3, d, e, p, h and q1–3) (4); different subunit
`combinations determine the pharmacologic prop-
`erties of the receptor (5, 6). The number and
`types of subunits vary depending on the location
`of the receptor in the CNS (7). The inhibitory
`neurotransmitter GABA binds to the receptor to
`open the chloride ion gates and produce an
`inhibitory current (6, 8). Binding of BZDs to the
`c subunit of the receptor is important in the
`potentiation of GABAergic inhibition (9). Differ-
`entiation between BZDs and GABA is impor-
`tant. Benzodiazepines do not
`substitute
`for
`
`70
`
`GABA, but instead enhance the inhibitory effects
`of GABA. Benzodiazepines allosterically bind to
`the receptor at a different location than GABA
`does and enhance the chloride channelÕs conduc-
`tance by increasing the frequency of gated chan-
`nel opening (6, 7, 10–12).
`In the search for BZD site ligands with higher
`therapeutic selectivity and a more favorable safety
`profile, GABAA receptor subtypes have long been
`considered promising targets (13). The pharmaco-
`logic relevance of GABAA receptor subtypes has
`been identified using a gene knock-in strategy in
`rodents. Based on in vivo point mutations,
`a-1-GABAA receptors have been found to mediate
`sedation and anterograde amnesia and to par-
`tially mediate anticonvulsant activity, whereas
`a-2-GABAA receptors mediate anxiolysis (14, 15).
`The basic chemical structure of BZDs is formed
`from the fusion of a benzene ring and a seven-
`membered diazepine ring (16) (Fig. 1). Clobazam is
`an exception with its 1-5-BZD structure (17). The
`common chemical structure of the BZDs accounts
`for their similar mechanisms of action.
`In pharmacologic terms, BZD potency refers to
`the in vivo affinity of
`the drug (or its active
`metabolites) for its receptor (18). Benzodiazepines
`are classified as low, medium (e.g. clorazepate and
`diazepam) or high (e.g. clonazepam and loraze-
`pam) potency (18, 19).
`
`Benzodiazepine pharmacokinetics
`
`Benzodiazepines have differences in their physio-
`chemical properties, most notably lipid solubility,
`which influence their
`rate of absorption and
`diffusion into tissue compartments and their phar-
`macokinetics. Each BZD has a unique pharmaco-
`kinetic profile that must be considered when the
`optimal agent is selected for a particular patient
`and condition. Key factors to consider include
`
`R1
`
`N
`
`O
`
`R2
`
`N
`
`R3
`
`ortho
`
`R7
`
`Figure 1. General chemical structure of 1,4- benzodiazepines.
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`route of administration, rate and extent of absorp-
`tion, metabolism, formation of active metabolites,
`elimination and drug interactions (20).
`
`Absorption and distribution
`
`When orally administered, most BZDs are exten-
`sively and rapidly absorbed, with bioavailabilities
`varying from 80% to 100% and times to peak
`concentration ranging from minutes to several hours
`(Table 1). Midazolam is an exception, with low oral
`bioavailability due to metabolism by cytochrome
`P-450 (CYP) enzyme 3A5 in intestinal epithelial
`tissue, which can reduce by up to 50% the fraction of
`the dose reaching the bloodstream (24). Benzodiaze-
`pines
`cross
`the blood–brain barrier
`rapidly,
`although the diffusion rate into the brain varies by
`drug and is largely determined by lipophilicity (21).
`The faster the diffusion rate, the earlier is the onset of
`pharmacodynamic effects. Rapid entry of BZDs
`into the CNS and highly perfused tissues is consis-
`tent with their short distribution half-lives (21, 25).
`Following rapid uptake, BZDs redistribute into less
`well-perfused tissues; the rate of redistribution is the
`fastest for the most lipid-soluble drugs (25). After an
`intravenous (i.v.) BZD administration, BZD phar-
`macokinetics can be characterized by a multicom-
`partmental mathematical model, with the first phase
`being distribution, followed by a longer elimination
`phase. Benzodiazepines also have large volumes of
`distribution, are highly bound to plasma proteins
`(Table 1) and readily cross into the placenta and
`breast milk (25).
`
`Table 1 Summary of absorption and distribution pharmacokinetics of selected
`BZDs
`
`Drug
`
`F (%)
`
`Tmax
`
`Clobazam
`Clonazepam
`Clorazepateb
`
`Diazepam
`
`Lorazepam
`
`Midazolam
`
`87
`>80
`PO: 100
`IM: 91
`PO: 100
`R: 90 (23)
`
`PO: 99
`IM: 96
`SL: 94
`PO: 40
`IM: 100
`
`1.3–1.7 h
`1–4 h
`PO: 0.5–2 h
`IM: 2.7–11 h
`PO: 30–90 min
`IM: 30–60 min
`R: 10–45 min
`PO: 2.4 h
`IM: 1.2 h
`SL: 2.3 h
`PO: 0.5–0.97 h
`IM: 0.24–0.51 h
`
`Protein
`binding
`(% bound)
`
`Distribution
`half-lifea
`(min)
`
`Vd (l ⁄ kg)
`
`82–90
`86
`96–98
`
`NA
`30 min (21)
`6–29 min (22)
`
`0.87–1.83
`1.5–4.4
`0.7–2.2
`
`96–99
`
`2–13 min (21)
`
`0.95–2.0
`
`93.2
`
`<11 min (21)
`
`0.85–1.5
`
`96
`
`4–19 min (21)
`
`0.7–1.7
`
`Values refer to adults receiving monotherapy and are from Anderson and Miller
`(24) unless otherwise specified. BZD, benzodiazepine; F, bioavailability; Tmax, time
`to maximum concentration; Vd, volume of distribution; NA, not applicable; PO, oral;
`IM, intramuscular; R, rectal; SL, sublingual.
`aAfter intravenous administration.
`bPharmacokinetics for N-desmethyldiazepam after administration of clorazepate.
`
`Benzodiazepines in the treatment of epilepsy
`
`Metabolism and elimination
`
`Benzodiazepines differ in their rates of elimination
`and the formation of pharmacologically active
`metabolites (Table 2). The elimination half-life
`(t1 ⁄ 2) of a BZD or of its active metabolite is used
`to categorize BZD duration of effect: short acting
`(<10 h;
`lorazepam, midazolam),
`intermediate
`acting (10–24 h; clonazepam) or
`long acting
`(>24 h; clobazam, clorazepate and diazepam) (43).
`Benzodiazepine metabolism is primarily cata-
`lyzed by CYP-dependent hydroxylation, demeth-
`ylation and nitroreduction (26, 44, 45). The CYP
`isoenzymes catalyzing these reactions include 3A4,
`3A5, 2B6, 2C9 and 2C19 (Tables 2 and 3). Uridine
`diphosphate
`glucuronosyltransferase
`is
`also
`involved in the conjugation of some BZDs (Table 3).
`Several BZDs have active metabolites. Diazepam
`and clorazepate are metabolized into the long-acting
`metabolite DMD (56). With multiple doses, the
`pharmacologic and toxic effects of diazepam are
`attributable to the parent drug, DMD, and other
`minor active metabolites (i.e. temazepam and oxaz-
`epam) (24). By contrast, clorazepate undergoes rapid
`and complete chemical conversion to DMD in the
`gastrointestinal tract; its pharmacologic effects are
`largely due to DMD (24, 56). N-Desmethyldiazepam
`undergoes glucuronidation to form a glucuronide
`conjugate (25%) and is hydroxylated (50%) by CYP
`2C19 and CYP 3A4 to form oxazepam (24, 37).
`Approximately
`5–9% of DMD is
`excreted
`unchanged in the urine (24). The t1 ⁄ 2 of DMD
`ranges widely from 20 to 179 h (24, 33, 34, 38).
`Other BZDs also have pharmacologically active
`metabolites. Clobazam is demethylated into an
`(N-desmethylclobazam)
`active metabolite
`(27).
`Midazolam is rapidly converted by CYP 3A4 and
`CYP 3A5 to 1-hydroxymidazolam, which contrib-
`utes approximately 10% to the biologic activity of
`its parent drug (24, 41). Clonazepam and loraze-
`pam undergo extensive metabolism, but no active
`metabolites are formed (18, 24).
`
`Effects of pharmacokinetics and pharmacodynamics
`on BZD use – The differences in BZD pharmacoki-
`netics and pharmacodynamics must be considered
`in order to use these drugs safely and effectively.
`Equivalent doses of BZDs differ as much as 20-fold
`because of differences in potency (57). The intensity
`of single-dose effects may vary, even if equipotent
`doses are used, because of varying oral absorption
`rates (58). Duration of action should be considered
`when choosing a BZD. When maintenance therapy
`is required (e.g. epilepsy and anxiety), long-acting
`BZDs are preferred because of their prolonged t1 ⁄ 2,
`as effective drug concentrations can be maintained
`
`71
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`Riss et al.
`
`Table 2 Summary of metabolism and elimination pharmacokinetics of selected BZDs
`
`Drug
`
`Clobazam
`Clonazepam
`
`Clorazepate
`
`Diazepam
`
`Lorazepam
`Midazolam
`
`Primary metabolic pathway
`
`Active metabolites
`
`Elimination half-life of
`parent druga (h)
`
`Elimination half-life of
`active metabolites (h)
`
`Demethylation (26)
`Nitroreduction (CYP 3A4),
`acetylation (NAT), hydroxylation (29–31)
`Decarboxylation, glucuronidation,
`hydroxylation (CYP 2C19 and 3A4) (24)
`Demethylation (CYP 2C9, 2C19, 2B6, 3A4,
`and 3A5), hydroxylation (CYP 3A4 and 2C19),
`glucuronidation (24, 35, 36)
`Glucuronidation (24)
`Hydroxylation (CYP 3A4 and 3A5) (25, 41, 42)
`
`N-desmethylclobazam (27)
`NA
`
`10–30 (28)
`19–60 (32)
`
`36–46 (28)
`NA
`
`DMD (major), oxazepam (minor) (24)
`
`NA
`
`DMD (major), oxazepam (minor),
`temazepam (minor) (24, 35, 37)
`
`21–70 (23, 38)
`
`NA
`1-hydroxymidazolam (minor) (24)
`
`7–26 (39, 40)
`1–4 (24)
`
`20–160 (24, 33, 34)
`Oxazepam: 6–24 (18)
`DMD: 49–179 (33, 38)
`Oxazepam: 6–24 (18)
`Temazepam: 8–24 (18)
`NA
`1 (24)
`
`BZD, benzodiazepine; CYP, cytochrome P-450; NAT, N-acetyltransferase; NA, not applicable; DMD, N- desmethyldiazepam.
`aHealthy subjects.
`
`Table 3 Enzyme-mediated BZD metabolism and drug interactions
`
`Enzyme
`associated
`with
`metabolism
`
`BZD
`substrates
`
`CYP 2C19
`
`Diazepam (46)
`
`CYP 3A4
`
`Clonazepam (29)
`
`Diazepam (46)
`Midazolam (46)
`
`UGT
`
`Lorazepam (53)
`Oxazepam (54)
`
`Inhibitors
`
`Inducers
`
`Fluvoxamine (46)
`MHD (weak) (47)
`Omeprazole (46)
`Oxcarbazepine (46)
`Ticlopidine (46)
`Topiramate (46)
`Azole antifungals
`(e.g. ketoconazole) (46)
`Cimetidine (46)
`Clarithromycin (46)
`Diltiazem (46)
`Erythromycin (46)
`Fluoxetine (51)
`Grapefruit juice (46)
`HIV protease
`inhibitor (46)
`Nefazodone (46)
`Sertraline (51)
`Valproate (55)
`
`Dexamethasone (48)
`Phenobarbital (48)
`Phenytoin (49)
`Rifampin (46)
`St JohnÕs wort (50)
`
`Carbamazepine (46)
`
`Phenobarbital (48)
`Phenytoin (46)
`Rifabutin (46)
`Rifampin (52)
`Rifapentine (51)
`St JohnÕs wort (50)
`
`Carbamazepine (55)
`Lamotrigine (weak) (55)
`Phenobarbital (55)
`Phenytoin (55)
`Rifampin (52)
`
`BZD, benzodiazepine; CYP, cytochrome P-450; MHD, monohydroxy derivative; HIV,
`human immunodeficiency virus; UGT, uridine diphosphate glucuronosyltransferase.
`
`without the need for frequent dosing (56). Short-
`acting BZDs are preferred for intermittent hyp-
`notic therapy, when the duration of action of the
`drug should be restricted to night-time, allowing
`patients
`to awaken feeling refreshed, without
`hangover effects (56).
`
`Drug–drug interactions
`
`Benzodiazepines interact with other drugs such as
`certain antidepressants, AEDs (e.g. phenobarbital,
`
`72
`
`phenytoin and carbamazepine), sedative antihista-
`mines, opiates, antipsychotics and alcohol (44, 57,
`59), which may result in additive sedative effects.
`As discussed earlier, BZD metabolism is com-
`plex and largely catalyzed by CYP isoenzymes.
`Consequently, there is potential for interactions
`between BZDs and drugs that induce or inhibit
`CYP isoenzymes. The clinical importance of these
`interactions depends on the net effect of inhibition
`or induction on the metabolic pathway of a
`particular BZD. For example,
`inhibition of a
`minor pathway may have little impact on drug
`concentration, whereas
`inhibition of a major
`pathway may result in enhanced clinical effect or
`toxicity. By contrast, addition of an enzyme-
`inducing drug that affects even a relatively minor
`pathway may lead to a clinically important reduc-
`tion in plasma BZD concentration. For BZDs with
`active metabolites, the addition of an inhibitor or
`inducer may affect only the parent drug, only the
`metabolite, or both. Clinicians should exercise
`particular caution when using BZDs with selective
`serotonin reuptake inhibitors, cimetidine, macro-
`lide antibiotics and antimycotics; these drugs may
`inhibit reactions catalyzed by certain CYP isoen-
`zymes and, therefore, inhibit the metabolism of
`many BZDs, which results in increased plasma
`BZD concentrations
`(44). Conversely, potent
`enzyme inducers (e.g. phenytoin, phenobarbital
`and carbamazepine) substantially increase clear-
`ance and reduce the t1 ⁄ 2 of certain BZDs (44). For
`a detailed review of pharmacokinetic drug interac-
`tions involving BZDs, see Tanaka (44).
`the
`Oral contraceptive steroids may inhibit
`metabolism of some BZDs that undergo oxidative
`metabolism or nitroreduction and accelerate
`the metabolism of some BZDs that are conjugated.
`Interactions between BZDs and oral contraceptives
`are described in detail by Back and Orme (60).
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`Special populations
`
`Special formulations of BZDs
`
`Benzodiazepines in the treatment of epilepsy
`
`Elderly patients – Pharmacokinetics in older indi-
`viduals differ from those in younger individuals
`because of age-related changes in physiology and the
`likelihood of concurrent diseases. Elderly individu-
`als often have variable drug absorption, decreased
`plasma protein–drug binding due to lower albumin
`concentrations, and reduced hepatic and renal
`clearance (61). Additionally, many elderly individ-
`uals take multiple medications, which increase their
`risk of drug–drug interactions. Therefore, treatment
`of the elderly can be challenging.
`Increased sensitivity of older patients to BZDs is
`partly due to reduced drug metabolism (when
`compared with younger adults), which can result in
`drug accumulation (62). Furthermore, BZD phar-
`macologic effects appear to be greater in elderly
`patients than in younger patients even at similar
`plasma BZD concentrations (63, 64), possibly
`because of age-related changes in drug–receptor
`interactions, post-receptor mechanisms and organ
`function. When a BZD is prescribed for an elderly
`patient, the initial maintenance dose should be half
`that recommended for younger adults (57), and
`BZD use should be only short term (limited to
`2 weeks) (65). A short-acting BZD may be prefer-
`able for treating an elderly patient because such a
`drug is better tolerated than is a BZD or BZD
`active metabolite with a long t1 ⁄ 2 (64).
`
`Pediatric patients – Limited information is available
`regarding BZD absorption in children. Often,
`before children are administered medications, the
`tablet is crushed or the capsule is opened, and the
`contents are mixed with food or drink. Food and
`beverages may affect BZD bioavailability, but
`studies investigating this issue in children are
`lacking.
`Drug metabolism is variable in children and
`depends on the biotransformation pathway. Cyto-
`chrome P-450-catalyzed metabolism tends to be
`low at birth, but exceeds adult values by age
`2–3 years; thereafter, CYP-catalyzed metabolism
`decreases, reaching adult levels around puberty
`(66). Metabolism via glucuronidation tends to be
`low in neonates, reaching adult
`levels by age
`3–4 years (66). In neonates, the t1 ⁄ 2 of clorazepate
`is prolonged and clearance is decreased (67).
`Infants have reduced hydroxylation metabolism,
`which results in a decreased clearance of diazepam
`(68). The t1 ⁄ 2 of midazolam is shorter in children
`than in adults: 0.79–2.83 h in children (69) vs 1.36–
`4 h in adults (24). Clinicians should consider how
`patient age may affect BZD clearance, because
`clearance will affect BZD dosing.
`
`can help
`Extended-release drug formulations
`patients with epilepsy achieve their primary treat-
`ment goals of controlling seizures while reducing
`side effects by minimizing fluctuations in drug
`concentration and by improving compliance.
`Extended-release formulations may also improve
`quality of
`life and patient
`satisfaction with
`treatment, in part by simplifying dosage regimens
`(70). Currently, clorazepate is the only BZD
`available in both a sustained-release, single-dose
`(TranxeneÒ-SD, Ovation Pharmaceuticals, Deer-
`field, IL, USA) formulation (11.25- and 22.5-mg
`tablets) and an immediate-release formulation that
`requires multiple doses per day (TranxeneÒ T-Tab;
`3.75-, 7.5- and 15-mg tablets) that is approved in
`the USA for the treatment of seizures. Some BZDs
`are also available as oral liquids [diazepam (Diaz-
`epam Intensol; 5 mg ⁄ ml), lorazepam (Lorazepam
`Intensol; 2 mg ⁄ ml) and midazolam (generic only;
`2 mg ⁄ ml)], disintegrating
`tablets
`[clonazepam
`(KlonopinÒ Wafer; Roche
`Pharmaceuticals,
`Nutley, NJ, USA; 0.125-, 0.25-, 0.5-, 1- and 2-mg
`[diazepam (DiastatÒ
`tablets)] or a rectal gel
`AcuDial; Valeant Pharmaceuticals International,
`Costa Mesa, CA, USA; 2.5-, 10- and 20-mg
`delivery systems)].
`
`Effect of extended-release formulations on plasma
`BZD concentrations: pharmacokinetic
`simulations
`with clorazepate – Our group has performed simu-
`lation studies of plasma DMD concentrations over
`time to investigate differences between clorazepate
`formulations and to characterize the effect of
`missed doses with or without replacement doses
`under
`steady-state conditions when using the
`sustained-release and immediate-release formula-
`tions (71). These simulations were briefly described
`by Kaplan and DuPont (72), but detailed results
`are reported herein. The following simulations
`were performed for both formulations using
`WinNonlinÒ (Pharsight Corporation, version 4.1:
`Mountain View, CA, USA) software and a two-
`compartment, first-order, oral-absorption pharma-
`cokinetic model: (1) steady-state conditions, (2)
`missed dose(s) without replacement and (3) missed
`dose(s) with replacement at the next scheduled
`dose. The following dosing schedules for the
`sustained-release and immediate-release formula-
`tions were entered to attain steady-state conditions
`(>7 days): clorazepate sustained-release 22.5 mg –
`one tablet orally every 24 h for 20 days; and
`clorazepate immediate-release 7.5 mg – one tablet
`orally three times daily (given 6 h apart)
`for
`20 days. The resulting simulated plasma DMD
`
`73
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`concentrations over time are depicted in Figs 2–4.
`The mean steady-state plasma DMD concentration
`was approximately 0.71 lg ⁄ ml for the immediate-
`release formulation and 0.73 lg ⁄ ml for the sus-
`tained-release formulation. The time to maximum
`concentration was 2.43 and 9.20 h for the imme-
`diate-release and sustained-release formulations
`respectively. Table 4 shows the effects of missed
`doses on plasma DMD concentrations. When a
`missed dayÕs dose of the immediate-release formu-
`lation was simulated, peak-to-trough differences
`(compared with steady
`state)
`increased by
`0.05 lg ⁄ ml
`(38.5%) with no dose replacement
`and increased by 0.29 lg ⁄ ml
`(223.1%) with
`replacement of the missed doses (Table 4). How-
`ever, when a missed dayÕs dose of the sustained-
`release formulation was simulated, peak-to-trough
`differences increased by 0.01 lg ⁄ ml (5.9%) with no
`dose replacement and increased by 0.24 lg ⁄ ml
`(141.2%) with replacement of the missed dose
`(Table 4).
`Despite the long half-life of DMD, a missed
`dayÕs dosing can result in altered peak-to-trough
`concentration ratios. Overall,
`the differences
`between peak and trough plasma DMD concen-
`trations after a missed daily dose of clorazepate
`increased more with the immediate-release formu-
`lation than with the sustained-release formulation
`(Table 4). There was little difference between the
`two formulations in peak and trough concentra-
`tions following a missed dayÕs dose. Although the
`effect was modest,
`the sustained-release tablet
`maintained higher trough concentrations after a
`missed daily dose and replacement of the missed
`dose. This effect may prevent breakthrough sei-
`zures in some patients. Additionally, when a
`missed daily dose was replaced, the sustained-
`release formulation resulted in a smaller change in
`peak concentrations, which may reduce the risk of
`drug toxicity.
`
`Use of benzodiazepines in epilepsy
`
`Efficacy
`
`Benzodiazepines are among the most useful AEDs
`available for treating status epilepticus and acute
`repetitive seizures and for febrile seizure prophy-
`laxis. Benzodiazepines were used in epilepsy man-
`agement as early as 1965, when Gastaut et al. (73,
`74) administered i.v. diazepam to treat status
`epilepticus. Since then, several other BZDs have
`been used for a variety of
`seizure disorders
`(Table 5).
`Several randomized controlled trials support the
`use of BZDs (particularly diazepam and loraze-
`
`Riss et al.
`
`0
`
`50
`
`100
`
`150
`
`300
`200
`250
`Time (h)
`
`350
`
`400
`
`450
`
`500
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Predicted DMD concentration (µg/ml)
`
`Figure 2. Simulated plasma N-desmethyldiazepam (DMD)
`concentration over time for immediate-release clorazepate
`(solid line, 7.5 mg given every 6 h for three daily doses) and
`sustained-release clorazepate (dashed line, 22.5 mg once daily)
`with no missed doses.
`
`Missed dose
`
`Missed doses
`
`0
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`350
`
`400
`
`450
`
`500
`
`Time (h)
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Predicted DMD concentration (µg/ml)
`
`Figure 3. Simulated plasma N-desmethyldiazepam (DMD)
`concentration over time for immediate-release clorazepate
`(solid line, 7.5 mg given every 6 h for three daily doses) and
`sustained-release clorazepate (dashed line, 22.5 mg once daily),
`missed daily dose(s) without replacement.
`
`Missed dose
`
`Replaced dose
`Replaced doses
`
`Missed
`doses
`
`0
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`350
`
`400
`
`450
`
`500
`
`Time (h)
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Predicted DMD concentration (µg/ml)
`
`Figure 4. Simulated plasma N-desmethyldiazepam (DMD)
`concentration over time for immediate-release clorazepate
`(solid line, 7.5 mg given every 6 h for three daily doses) and
`sustained-release clorazepate (dashed line, 22.5 mg once daily),
`missed daily dose(s) replaced at next dayÕs dose.
`
`74
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`Benzodiazepines in the treatment of epilepsy
`
`Table 4 Effects of missed doses on simulated plasma DMD concentrations (71)
`
`Clorazepate sustained-release
`
`Clorazepate immediate-release
`
`Formulation
`
`Simulation schemea
`
`Peak ⁄ trough differenceb at steady state (lg ⁄ ml)
`Peak ⁄ trough differenceb after missed dose(s) (lg ⁄ ml)
`Change in peak plasma DMD concentrations (%)
`Change in trough plasma DMD concentrations (%)
`
`1
`
`0.17
`NA
`NA
`NA
`
`2
`
`NA
`0.18
`)29.6
`)44.1
`
`3
`
`NA
`0.41
`6.0
`)41.9
`
`1
`
`0.13
`NA
`NA
`NA
`
`2
`
`NA
`0.18
`)27.1
`)48.8
`
`3
`
`NA
`0.42
`9.6
`)48.8
`
`DMD, N-desmethyldiazepam; NA, not applicable.
`aSimulation scheme: 1 = no missed doses; 2 = missed daily dose(s) without replacement; 3 = missed daily dose(s) with replacement.
`bDifference between maximum plasma DMD concentration and minimum plasma DMD concentration.
`
`Table 5 Recommended clinical uses of benzodiazepines
`
`Drug
`
`Trade Name
`
`Generic availability
`
`Product-labeled uses (75)
`
`Common uses in epilepsy (76)
`
`Clobazam
`
`Frisium
`
`Clonazepam
`
`Klonopin
`
`Clorazepate
`
`Diazepam
`
`Tranxene T-Tab
`Tranxene-SD
`Valium
`
`Lorazepam
`
`Midazolam
`
`Ativan
`
`Versed
`
`NA
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Not FDA approved
`
`Panic disorder, epileptic
`seizures (alone or adjunct)
`
`Anxiety, alcohol withdrawal,
`adjunctive treatment of partial seizures
`Anxiety, alcohol withdrawal,
`muscle relaxant, epileptic seizures
`
`Anxiety, pre-anesthetic to induce amnesia,
`antiemetic adjunct, status epilepticus
`Anesthesia, preoperative and procedural
`sedation
`
`First-line adjunctive treatment for treatment-resistant
`partial and generalized seizures, intermittent
`therapy and non-convulsive status epilepticus
`Second-line adjunctive treatment for partial and
`generalized (particularly absence and myoclonic)
`seizures, early status epilepticus and
`Lennox–Gastaut syndrome; second-line
`treatment of status epilepticus
`Adjunctive treatment of partial seizures (75)
`
`First-line treatment for early status epilepticus;
`second-line therapy for established
`status epilepticus; treatment of
`non-convulsive status epilepticus;
`intermittent prophylactic therapy for
`febrile seizures; and at-home treatment of ARS
`First-line treatment for early status epilepticus
`and out-of-hospital status epilepticus
`Second-line therapy for early status epilepticus
`
`NA, not applicable; FDA, US Food and Drug Administration; ARS, acute repetitive seizures.
`
`pam) as initial drug therapy in patients with status
`epilepticus (77–80). Intermittent use of BZDs is
`especially suitable for patients with clusters of
`repetitive seizures (81). Fewer studies have evalu-
`ated the clinical efficacy of BZDs in chronic
`epilepsy. Nevertheless, BZDs are useful as adjunc-
`tive agents in treating certain patients with both
`partial and primary generalized seizures
`(81).
`Benzodiazepines are versatile drugs that can be
`employed in a variety of clinical settings because of
`their broad spectrum of activity and multiple
`formulations and because they can be administered
`by several routes (81).
`
`Diazepam – Diazepam is a drug of first choice for
`treatment of early status epilepticus and acute
`repetitive seizures and for febrile seizure prophy-
`laxis. It can be administered as an i.v. bolus, as a
`continuous infusion, or rectally, which enhances its
`
`utility in managing seizure emergencies. Four
`randomized controlled trials support diazepam as
`a drug of first choice for managing status epilep-
`ticus (77–80). Success rates of i.v. diazepam for
`treating status epilepticus vary. In a randomized
`double-blind study comparing diazepam and
`lorazepam, Leppik et al. (80) found that 76% of
`status epilepticus episodes (25 of 33) were termi-
`nated by one or two diazepam doses (5 mg ⁄ min).
`In a randomized, non-blinded trial of patients
`>15 years of age with status epilepticus, Shaner
`et al. (78) reported that seizures were aborted in
`<10 min in 55.6% of patients (10 of 18) treated
`diazepam (2 mg ⁄ min)
`with
`and
`phenytoin
`(40 mg ⁄ min). A randomized, double-blind, multi-
`center Veterans Affairs cooperative study was
`designed to compare the effectiveness of
`four
`treatments for overt or subtle status epilepticus
`(79). Three hundred eighty-four patients with overt
`
`75
`
`Neurelis - EX. 2013
`Aquestive Therapeutics, Inv. v. Neurelis, Inc. - IPR2019-00451
`
`

`

`Riss et al.
`
`status epilepticus and 134 patients with subtle
`status epilepticus were randomly assigned to
`receive either diazepam (0.15 mg ⁄ kg)
`followed
`by phenytoin (18 mg ⁄ kg), lorazepam (0.1 mg ⁄ kg)
`(15 mg ⁄ kg)
`alone,
`phenobarbital
`alone
`or
`phenytoin (18 mg ⁄ kg) alone. Treatment with diaz-
`epam plus phenytoin was successful in 55.8% of
`patients (53 of 95) with overt status epilepticus and
`8.3% of patients (three of 36) with subtle status
`epilepticus (79). Alldredge et al. (77) conducted a
`randomized double-blind trial to determine the
`effectiveness of
`i.v. diazepam,
`lorazepam and
`placebo on status epilepticus when the drugs were
`administered by paramedics before patients arrived
`at the hospital. They found that status epilepticus
`was terminated by the time of arrival
`in the
`emergency department in 42.6% of the 68 patients
`treated with one or two 5-mg doses of
`i.v.
`diazepam (infused over 1–2 min). Limited pub-
`lished data indicate that continuous i.v. infusions
`of diazepam are safe and effective (82–84).
`Use of i.v. diazepam can result in seizure relapse
`within 2 h of a single injection in approximately
`50% of patients (76). Therefore, multiple injections
`or continuous

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