Methods and compositions particularly for treatment of attention deficit disorder

Information

  • Patent Grant
  • 11896722
  • Patent Number
    11,896,722
  • Date Filed
    Wednesday, January 5, 2022
    2 years ago
  • Date Issued
    Tuesday, February 13, 2024
    3 months ago
Abstract
There is described, inter alia, a coated bead comprising: (a) a granule; (b) a first layer coated over the granule, the first layer comprising a first amount of an active pharmaceutical ingredient comprising a central nervous system stimulant; and (c) a second layer coated over the first layer, the second layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer until after the coated bead reaches a distal intestine portion of a subject to whom the coated bead is administered; and (d) the third layer coated over the second layer, the third layer comprising a second amount of the active pharmaceutical ingredient, the third layer being configured to permit substantially immediate release of the active pharmaceutical ingredient comprised therein. Embodiments related to a solid oral pharmaceutical composition are also described.
Description
BACKGROUND OF THE INVENTION
Field of the Invention

The present invention relates to methods and compositions, particularly for treatment of attention deficit disorder.


Description of the Prior Art

Sustained release dosage forms are important in the search for improved therapy, both through improved patient compliance and decreased incidences of adverse drug reactions.


It is the intent of sustained release formulations to provide a longer period of pharmacologic action after administration than is ordinarily obtained after administration of immediate release dosage forms. Sustained release compositions may be used to delay absorption of a medicament until it has reached certain portions of the alimentary tract, and maintain a desired concentration of the medicament in the blood stream for a longer duration than would occur if conventional rapid release dosage forms are administered. Such longer periods of response provide for many therapeutic benefits that are not achieved with corresponding short acting, immediate release preparations. Thus, therapy may be continued without interrupting the sleep of the patient, which is of special importance, for example, when treating a patient for moderate to severe pain (e.g., a post-surgery patient, a cancer patient, etc.), or for those patients who experience migraine headaches on awakening, as well as for the debilitated patient for whom sleep is essential. A further general advantage of longer acting drug preparations is improved patient compliance resulting from the avoidance of missed doses through patient forgetfulness.


Unless conventional rapid acting drug therapy is carefully administered at frequent intervals to maintain effective steady state blood levels of the drug, peaks and valleys in the blood level of the active drug occur due to rapid absorption, systemic excretion of the compound and through metabolic inactivation, thereby producing special problems in maintenance therapy of the patient.


In view of this, it is considered a goal of many skilled in the art that a controlled release dosage form will ideally provide therapeutic concentration of the drug in blood that is maintained throughout the dosing interval with a reduction in the peak/trough concentration ratio. Central to the development process are the many variables that influence the in vivo release and subsequent absorption of the active ingredients from the gastrointestinal tract.


It is known in the pharmaceutical art to prepare compositions which provide for sustained release of pharmacologically active substances contained in the compositions after oral administration to humans and animals. Sustained release formulations known in the art include specially coated pellets, coated tablets and capsules, and ion exchange resins, wherein the slow release of the active medicament is brought about through selective breakdown of the coating of the preparation or through compounding with a special matrix to affect the release of a drug. Some sustained release formulations provide for related sequential release of a single dose of an active compound at predetermined periods after administration.


Thus, sustained release dosage forms are important in the search for improved therapy, both through improved patient compliance and decreased incidences of adverse drug reactions.


While controlled and/or sustained release compositions have constituted a definite advance in the art, improvements in these compositions have been sought, particularly for preparations available for conditions such as Attention Deficit Hyperactivity Disorder (ADHD), diabetes etc.


Attention Deficit Disorders are the most common psychiatric disorders in children (Campbell et al. 1992) with reported rates ranging from 4% to 9% (Aman et al. 1983).


Attention Deficit Disorder (ADD) is characterized by inattention and impulsivity and may be present with hyperactivity (ADHD) (Shaywitz et al. 1984). Other characteristics may include aggressiveness, stealing, lying, truancy, setting fires, running away, explosiveness, cognitive and learning problems as well as poor social skills (Campbell et al. 1992). It is four to five times more frequent in boys than girls (Campbell et al. 1992).


Stimulant medication, such as amphetamines, have been shown to be the most effective agents in the treatment of children with disorders of activity modulation and attention regulation and result in significant improvement in 70 to 80 percent of affected children (Shaywitz et al. 1984). Positive effects of stimulants have been documented in a variety of areas including behavioral, social, perceptual performance, motor activity, impulse control, attention regulation and cognitive performance (Barkley 1977, Kavale 1983, Offenbacher et al. 1983, Rosenthalet al 1978).


Long thought of as a childhood disorder, ADHD is now known to persist into adolescence and adulthood (Practice Parameter for the Use of Stimulant Medications in the treatment of Children, Adolescents, and Adults. J. AM. ACAD. CHILD ADOLESC. PSYCHIATRY, 41:2 SUPPLEMENT, FEBRUARY 2002)


Methylphenidate [dl-threo-methyl-2-phenyl-2-(2-piperidyl)acetate] is the psycho-stimulant used most frequently in the treatment of hyperactivity and attention deficit disorder. It appears to have a higher incidence of positive effects and a lower incidence of adverse effects than other psychostimulants. The efficacy of methylphenidate (“MPH”) in improving attention and behavioral symptoms has been supported by many studies.


Immediate release methylphenidate preparations, because of their short half-life, require frequent administration at short intervals to ensure adequate treatment throughout a child's school day, adolescence's school day (high school, college, university) and adult working day. The rapid onset and offset of immediate release methylphenidate preparations means that a medicated person with attention deficit disorder will be maximally affected only for relatively brief periods during the day. Due to its short half-life, it has been known to administer MPH given twice per day, usually once after breakfast and once during the day, an event that some children and some school personnel apparently avoid, resulting in poor compliance with prescribed regimens (Brown et al., 1985; Firestone 1982).


Compliance is a major problem for children, adolescences and adults. Poor compliance in taking medication may explain, in part, the variable and conflicting results reported in many studies of the effect of medication on improving the behavior of hyperactive children, adolescents and adults. These limitations of immediate release methylphenidate led to interest in products with longer effective periods of action.


Thus, much of the prior art has focused on development of formulations for treatment of ADHD with a focus on administration to children and improving patient compliance in the patient population. This has led to commercialization of a number of sustained release formulations of methylphenidate—e.g., Ritalin SR™, Concerta™ and Biphentin™.


Duration of efficacy with long-acting methylphenidate formulations was maintained from one hour to 12 hours post-dosing for osmotically controlled-release oral delivery systems (four trials), 1.5 hours to 7.5 hours for methylphenidate extended release in one trial, one hour to 12 hours post-dosing for methylphenidate spheroidal oral drug absorption systems (two trials) and 30 minutes to 12 hours post-dosing for dexmethylphenidate extended release (five trials). Most long-acting stimulants conferred benefits on ADHD symptoms in patients across the age spectrum for up to 12 hours after a single morning dose as measured by the permanent product measure of performance mathematics test (PERMP). Formulations may differ in time to peak effect and maintenance of effect as well as magnitude of effect at different time points during the day (Brams M, Moon E, Pucci M, Lopez F A. Duration of effect of oral long-acting stimulant medications for ADHD throughout the day. Curr Med Res Opin. 2010 August; 26(8):1809-25. doi: 10.1185/03007995.2010.488553).


Despite the advances in the art, there is still room for improvement.


First, some or all of the commercially available sustained release formulations of methylphenidate do not have, in combination, a rapid onset of action and a duration of action that exceeds 12 hours. The provision of a sustained release formulation having this combination of features would be highly desirable for adolescents or adults whose daily activities require them to have a rapid onset of therapeutic effect and duration of action that lasts at least 14 hours to get them through the day and will into the evening without the need of another dose of the medication.


Second, some or all of the commercially available sustained release formulations of methylphenidate are susceptible to premature release of the active ingredient in a gastric environment that contains alcohol (e.g., ethanol). This can be a significant problem if the subject taking the formulation is an alcohol abuser.


This, it would be highly desirable to have a pharmaceutical composition that obviates or mitigates one or both of these problems in the prior art.


SUMMARY OF THE INVENTION

It is an object of the present invention to obviate or mitigate at least one of the above-mentioned disadvantages of the prior art.


It is another object of the present invention to provide a novel coated bead.


It is another object of the present invention to provide a novel solid oral pharmaceutical composition.


Accordingly, in one of its aspects, the present invention provides a coated bead comprising:

    • (a) a granule;
    • (b) a first layer coated over the granule, the first layer comprising a first amount of an active pharmaceutical ingredient comprising a central nervous system stimulant;
    • (c) a second layer coated over the first layer, the second layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer until after the coated bead reaches a distal intestine portion of a subject to whom the coated bead is administered; and
    • (d) a third layer coated over the second layer, the third layer comprising a second amount of the active pharmaceutical ingredient, the third layer being configured to permit substantially immediate release of the active pharmaceutical ingredient comprised therein.


In another of its aspects, the present invention provides a an oral solid pharmaceutical composition comprising a first plurality of coated beads and a second plurality of coated beads, wherein:

    • each coated bead in the first plurality of coated beads comprising: a first granule and a first layer coated over the first granule, the first layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant, the first plurality of coated beads being configured to provide substantially immediate release of the active pharmaceutical ingredient; and
    • each coated bead in the second plurality of coated beads comprising: a second granule; a first layer coated over the granule, the first layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant; and a second layer coated over the first layer, the second layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer until after the coated bead reaches a distal intestine portion of a subject to whom the coated bead is administered, the coated bead being substantially free of an outer layer configured to provide substantially immediate release of the active pharmaceutical ingredient.


In yet another of its aspects, the present invention provides an oral solid pharmaceutical composition comprising a first plurality of coated beads, a second plurality of coated beads and a third plurality of coated bead, wherein:

    • each coated bead in the first plurality of coated beads comprising: a first granule and a first layer coated over the first granule, the first layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant, the first plurality of coated beads being configured to provide substantially immediate release of the active pharmaceutical ingredient;
    • each coated bead in the second plurality of coated beads comprising: a second granule; a first layer coated over the granule, the first layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant; and a second layer coated over the first layer, the second layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer; and
    • each coated bead in the third plurality of coated beads comprising: a third granule; a first layer coated over the granule, the first layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant, a second layer coated over the first layer, the second layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer until after the coated bead reaches a distal intestine portion of a subject to whom the coated bead is administered.


In yet another of its aspects, the present invention provides a coated bead comprising:

    • (a) a granule;
    • (b) an inner layer coated over the granule, the inner layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant; and
    • (c) an outer delayed release layer coated over inner layer which is substantially free of a salt of alginic acid;
    • wherein release of the active pharmaceutical ingredient is not more than 20% when measured under in vitro conditions with stirring at 100 rpm at pH 1.2 for 2 hours in 900 mL of a medium comprising up to about 35% v/v ethanol.


In yet another of its aspects, the present invention provides a coated bead comprising:

    • (a) a granule;
    • (b) an inner layer coated over the granule, the inner layer comprising an active pharmaceutical ingredient comprising a central nervous system stimulant; and
    • (c) an outer delayed release layer coated over inner layer, the outer delayed release coating comprises an anionic copolymer based on methyl acrylate, methyl methacrylate and methacrylic acid with the proviso that the outer delayed release layer is substantially free of a salt of alginic acid, the outer delayed release layer being present at an average thickness in the range of from about 5 μm to about 50 μm.


In yet another of its aspects, the present invention provides a coated bead comprising:

    • (a) a granule comprising a first amount of an active pharmaceutical ingredient comprising a central nervous system stimulant (e.g., the granule can comprise a granule substrate in admixture with the active pharmaceutical ingredient or the active pharmaceutical ingredient could be coated over the granule substrate); and
    • (b) a first layer coated over the granule, the first layer being present in an amount sufficient to substantially delay release of the active pharmaceutical ingredient in the first layer until after the coated bead reaches a distal intestine portion of a subject to whom the coated bead is administered; and
    • (c) a second layer coated over the first layer, the second layer comprising a second amount of the active pharmaceutical ingredient, the second layer being configured to permit substantially immediate release of the active pharmaceutical ingredient comprised therein.


Throughout this specification, the term “coated over” (or the functional equivalent thereof) is used to describe a first layer of material disposed exteriorly with respect to a second layer of material. It should be clearly understood that, in such a case, the first layer layer of material may be directly coated over (i.e., in contacting relation with) the second layer of material or indirectly coated over (i.e., in non-contacting relation with) the second layer of material. An example of the “indirectly coated over” would be when the first layer of material and the second layer of material has disposed between them one or more intermediate layers of material. The point is the term “coated over” (or the functional equivalent thereof), when used on its own encompasses both “directly coated over” and “indirectly coated over” described above.


The present inventors have developed a novel coated bead and a novel pharmaceutical composition which are believed to obviate or mitigate one or both of the above-mentioned disadvantages described above with reference to some or all of the commercially available sustained release formulations of methylphenidate. The present coated bead and pharmaceutical composition are believed to be highly advantageous in that they have a rapid on set of action (e.g., approximately 1 hour after administration) and a long duration of action (e.g., approximately 16 hours or more) after reaching steady state in the subject. While not wishing to be bound by any particular theory or mode of action, it is believed that the long duration of action results in a blood plasma concentration of the active ingredient at 24 hours after administration which allows for a rapid onset of action when another dose of the active ingredient is taken—i.e., there appears to be a baseline blood plasma concentration of the active ingredient when it is time to take a subsequent dose to allow for a rapid onset of action of that subsequent dose.


The present coated bead and pharmaceutical composition are believed to address a limitation of some or all current commercially available long-acting methylphenidate formulations which are not reported to provide and maintain duration of action beyond 12 hours. The present coated bead and pharmaceutical composition are also believed to address the limitation with long-acting lisdexamfetamine dimesylate that is reported to last up to 14 hours but do not have a rapid onset of action. These two characteristics (rapid onset of action and long duration of action) of the present coated bead and pharmaceutical composition are believed to address a significant limitation for adolescences or adults whose daily activities require them to have a rapid onset of therapeutic effect and duration of action that lasts at least 14 hours to get them through the day and will into the evening without the need of another dose of the medication.


In one preferred embodiment, the present coated bead and pharmaceutical composition are characterized by having a resistance release of the active ingredient in an aqueous composition comprising up to about 35% by volume of an alcohol (e.g., ethanol)—i.e., release of the active pharmaceutical ingredient is not more than 20% when measured under in vitro conditions with stirring at 100 rpm at pH 1.2 for 2 hours in 900 mL of a medium comprising up to about 35% v/v ethanol. This resistance to alcohol-related release of the active pharmaceutical ingredient can be achieved without the need to use coating layer comprising one or more salts of aglinic acid thereby simplifying manufacturing costs and the like.





BRIEF DESCRIPTION OF THE DRAWINGS

Embodiments of the present invention will be described with reference to the accompanying drawings, wherein like reference numerals denote like parts, and in which:



FIGS. 1-10 illustrate results on testing done on formulations produced in the examples described below.





DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

The coated bead and solid oral pharmaceutical compositions of the present invention include a central nervous system stimulant which can be generally defined as a chemical entity that affects the dopamine or norepinephrine neural pathways. Preferred pharmaceutically active ingredients include, but are not limited to amphetamine, dextroamphetamine, the active isomers of amphetamines and amphetamine salts including salts of dextroamphetamine, methylphenidate and its active salts, or combinations thereof, all of which can be used as racemic mixtures or pure isomers such as d-threo methylphenidate, or a prodrug or pharmaceutical salt, or mixed pharmaceutical salts of any thereof alone or in combination. The disclosed coated bead and solid oral pharmaceutical compositions can also include a prodrug, including but not limited to amino acid conjugated active ingredients such as 1-lysine-d-amphetamine.


Conditions or disorders that can be treated using the present coated bead or solid oral pharmaceutical compositions include, but are not limited to attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD), excessive daytime sleepiness, major depressive disorder, bipolar depression, negative symptoms in schizophrenia, chronic fatigue, fatigue associated with chemotherapy or binge eating disorder. Attention deficit disorders are characterized by hyperactive, impulsive or inattentive symptoms that cause impairment in social, academic, or occupational functioning, and are often present in two or more settings, school (or work) and at home, for example. For the Inattentive Type, at least 6 (5 for adults≥18 years of age) of the following symptoms have persisted for at least 6 months: lack of attention to details/careless mistakes; lack of sustained attention; poor listener; failure to follow through on tasks; poor organization; avoids tasks requiring sustained mental effort; loses things; easily distracted; and forgetful. For the Hyperactive-Impulsive Type, at least 6 (5 for adults≥18 years of age) of the following symptoms have persisted for at least 6 months: fidgeting/squirming; leaving seat; inappropriate running/climbing; difficulty with quiet activities; “on the go;” excessive talking; blurting answers; can't wait turn, and intrusive. The combined type includes both inattentive and hyperactive-impulsive behaviors.


It is understood that the term treatment as used herein is not limited to the cure or elimination of any condition or disorder nor is that term limited to the achievement of certain milestones or improvement criteria in a particular subject, but includes the administration of an agent for the purpose of achieving positive effects in terms of cognitive or behavioral function, reduction of symptoms or side effects. All such activities are considered to be treatment whether or not any improvement is immediately observable or measureable.


In a highly preferred embodiment, the present invention relates to a controlled release oral formulation of methylphenidate (or a pharmaceutically acceptable salt thereof) that provides a rapid onset of therapeutic effect and a gradual drop in plasma concentration after a prolonged period of therapeutic effect (e.g., 16 hours). This oral formulation comprises a plurality of substrates, preferably in the form of coated beads. Preferably, the coated bead comprises: (i) an initial portion of an effective dose of methylphenidate (or a pharmaceutically acceptable salt thereof) in immediate release form coated over granule; (ii) a controlled release (e.g., hydrophobic) material, preferably in the form of an acrylic polymer, coated over (i); (iii) a delayed (or distal) release (e.g., colonic delivery) coating over (ii) in an amount sufficient to substantially delay the release of the drug from the substrate until after coated bead passes through the stomach and the distal part of the gastrointestinal tract; and, optionally, (iv) a remaining portion of the effective dose of methylphenidate (or a pharmaceutically acceptable salt thereof) in immediate release form coated over (iii).


Preferably, the colonic delivery coating is derived from an aqueous dispersion of an anionic copolymer based on methyl acrylate, methyl methacrylate and methacrylic acid, a plasticizer and a glidant. The contents of the encapsulated product may be sprinkled on soft foods before administration.


The substrate (e.g., granules) can be chosen from spheres, also referred as pellets, also referred as beads, made of microcrystalline cellulose, mannitol-PVP, silica, starch, lactose, calcium carbonate or combination thereof. The preferred substrate to be used is sugar spheres 14/18 mesh to 18/20 mesh.


It can be preferred to use sugar spheres 14/18 mesh to 18/20 mesh amount of about 20% to about 70% by weight, of about 25% to about 65% by weight, of about 40% to about 64% by weight, of about 41% to about 63% by weight, of about 42% to about 62% by weight, of about 43% to about 61% by weight, based on the weight of the pharmaceutical composition. An amount of about 44.0% to about 53.5% by weight based on the weight of the pharmaceutical composition can be preferred of the nonpareil substrate, particularly of sugar spheres 14/18 mesh to 18/20 mesh.


The controlled release polymer can include ethylcellulose polymers, cellulose ethers (e.g., hydroxypropyl methylcellulose, hydroxypropylcellulose, methylcellulose, hydroxy-ethylcellulose, etc.), polyethylene oxide, polyvinyl alcohol derivates, methacrylic acid copolymers (e.g., poly(ethylene glycol) diacrylate, poly(ethylene glycol) triacrylate, poly(ethylene glycol) dimethacrylate, poly(ethylene glycol) trimethacrylate, polymulti (meth)acrylates], polyethylene glycol, polyglycolic acid, polylactic acid, poly caprolactone, poly(n-hydroxybutyrate), polyamino acids, poly(amide-enamines), poly(esters)1, ethylene-vinyl acetate (EVA), polyvinyl pyrrolidone (PVP), poly acrylic acid (PAA), poly methacrylic acid (PMAA) or combinations thereof in amounts that would deliver the active pharmaceutical ingredient at the desired release rate. Preferably, the controlled release polymer is derived from a mixture copolymer of ethyl acrylate, methyl methacrylate and methacrylic acid ester with quaternary ammonium groups (Ammonio Methacrylate Copolymer, Type B USP/NF). 1Chem. Rev. 1999, 99, 2118-3198 Polymeric Systems for Controlled Drug Release (Uhrich et al.)


It can be preferred to use ammonio methacrylate copolymer, Type B USP/NF as a controlled release material. Such a material is commercially available from Evonik under the tradement name Eudragit® RS30D.


It thus can be preferred to use a controlled release polymer amount of about 3% to about 16% by weight, of about 4% to about 15% by weight, of about 5% to about 14% by weight, of about 5.1% to about 13.5% by weight, such as of about 8.0% by weight, of about 8.1% by weight, of about 8.2% by weight, of about 8.3% by weight, of about 8.4% by weight, of about 8.5% by weight, of about 8.6% by weight, of about 8.7% by weight, of about 8.8% by weight, of about 8.9% by weight or about 9.0% by weight, of about 9.1% by weight, of about 9.2% by weight, of about 9.3% by weight, of about 9.4% by weight, of about 9.5% by weight, of about 9.6% by weight, of about 9.7% by weight, of about 9.8% by weight, of about 9.9% by weight of about 10.0%, of about 10.1% by weight, of about 10.2% by weight, of about 10.3% by weight, of about 10.4% by weight, of about 10.5% by weight, of about 10.6% by weight or of about 10.7% by weight, based on the weight of the pharmaceutical composition and the coated bead.


An amount of about 10.0% to about 10.7% by weight based on the weight of the pharmaceutical composition can be preferred, particularly of ammonio methacrylate copolymer, Type B USP/NF is used as controlled release modifier. The aforementioned amounts refer to the amount of all controlled release (e.g., hydrophobic) materials in the pharmaceutical composition or coated bead.


The delayed (or distal) release (e.g., colonic delivery) coating material can include guar gum, pectin, hydroxypropyl methylcellulose phthalate, cellulose acetate phthalate, cellulose acetate trimelliate, biodegradable polysaccharides (amylose, arabinogalactan, chitosan, chondroitin sulfate, cyclodextrine, dextran, guar gum, pectin, xanthan gum, xylan), poly(methacylic acid-co-methyl methacrylate) 1:2, poly(methacylic acid-co-methyl methacrylate) 1:1, polyvinyl acetate phthalate, covalent linkage of the drug with carrier (azo conjugates, cyclodextrine conjugates, glycoside conjugates, glucuronate conjugates, dextran conjugates, polypeptide conjugates, polymeric drugs), acidic comonomers, methacryloyloxy azobenzene and 2-hydroxyethyl methacrylate (HEMA), dextran hydrogels, and combinations thereof in amounts that would control the delivery of the product to the distal part of the GI tract. The preferred system to be used is the anionic copolymer based on methyl acrylate, methyl methacrylate and methacrylic acid (IUPAC name: Poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) 7:3:1).


It can be preferred to use poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) 7:3:1 as the delayed (or distal) release (e.g., colonic delivery) material. Such a material is commercially available from Evonik under the tradement name Eudragit® FS30D.


It thus can be preferred to use a distal release modifier amount of about 3% to about 20% by weight, of about 8% to about 18% by weight, of about 10% to about 17% by weight, of about 10.1% to about 16.5 by weight, such as of about 15.0% by weight, of about 15.1% by weight, of about 15.2% by weight, of about 15.3% by weight, of about 15.4% by weight, of about 15.5% by weight, of about 15.6% by weight, of about 15.7% by weight, of about 15.8% by weight, of about 15.9% by weight or about 16.0% by weight, of about 16.1% by weight, of about 16.2% by weight, of about 16.3% by weight, of about 16.4% by weight based on the weight of the pharmaceutical composition.


An amount of about 15.0% to about 16.0% by weight based on the weight of the pharmaceutical composition can be preferred, particularly of poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) 7:3:1 is used as distal release modifier. The aforementioned amounts refer to the amount of all delayed (or distal) release (e.g., colonic delivery) materials (i.e., including mixtures) in the pharmaceutical composition and the coated bead.


Plasticizers can may optionally be used. Examples of useful plasticizers include citrates (triethyl citrate, acetyl Triethyl citrate, tributyl citrate and acetyl tributyl citrate, acetyl tributyl citrate), triacetin, dibutyl sebacate, sebacate and azelate esters (di-2-ethylhexyl sebacate, di-2-ethylhexyl azelate, diisodecyl sebacate), ester of glycol and polyhydric alcohol [propylene glycol, glycerol (glycerin), polyethylene glycol, glyceryl triacetate], glyceryl monostearate (GMS), polysorbate 80, phthalates (di-octyl phthalate, diisodecyl phthalate, diisononyl phthalate), dibutyl phthalate, diethyl phthalate, adipates, phosphate esters, Polymerics, trimelliates (tris-2-ethylhexyltrimelliate), glutarates, castor oil, acetylated monoglycerides, fractionated coconut oil and mixtures of any two or more thereof. The preferred plasticizers to be used are triethyl citrate, glyceryl monostearate in combination with polyoxyethylene (20) sorbitan monooleate (Polysorbate 80™).


It can be preferred to use triethyl citrate and glyceryl monostearate emulsion as plasticizer system.


It thus can be preferred to use a plasticizer system amount of about 0.1% to about 10% by weight, of about 0.5% to about 9% by weight, of about 1% to about 7% by weight, of about 2% to about 6% by weight, of about 2.5% to about 5.5% by weight, of about 3.5% to about 4.5% by weight such as of about 3.6% by weight, of about 3.7% by weight, of about 3.8% by weight, of about 3.9% by weight, of about 4.0% by weight, of about 4.1% by weight, of about 4.2% by weight, of about 4.3% by weight, of about 4.4% by weight, of about 4.5% by weight based on the weight of the pharmaceutical composition.


An amount of about 4.0% to about 4.5% by weight based on the weight of the pharmaceutical composition can be preferred, particularly of triethyl citrate and glyceryl monostearate emulsion is used as plasticizer system. The aforementioned amounts refer to the amount of all plasticizers (i.e., including mixtures) in the composition.


Glidants can include talc, fumed silica, lecithin. The preferred glidants to be used are talc and fumed silica.


Binders can include hydroxypropyl cellulose (HPMC), hydroxypropyl cellulose (HPC), polyvinyl pyrollidone, carbopol, and combinations thereof. It can be preferred to use HMPC as a binder.


It thus can be preferred to use a binder amount of about 1% to about 10% by weight, of about 2% to about 9% by weight, of about 3% to about 7% by weight, of about 3% to about 6% by weight, such as of about 3.0% by weight, of about 3.1% by weight, of about 3.2% by weight, of about 3.3% by weight, of about 3.4% by weight, of about 3.5% by weight, of about 3.6% by weight, of about 3.7% by weight, of about 3.8% by weight, of about 3.9% by weight, of about 4.0% by weight, of about 4.1% by weight, of about 4.2% by weight, of about 4.3% by weight, of about 4.4% by weight, of about 4.5% by weight, of about 4.6% by weight, of about 4.7% by weight, of about 4.8% by weight, of about 4.9% by weight or of about 5.0% by weight, of about 5.1% by weight, of about 5.2% by weight, of about 5.3% by weight, of about 5.4% by weight, of about 5.5% by weight, of about 5.6% by weight, of about 5.7% by weight, of about 5.8% by weight, of about 5.9% by weight or of about 6.0% by weight, based on the weight of the pharmaceutical composition.


An amount of about 3.8% to about 6.0% by weight based on the weight of the pharmaceutical composition can be preferred, particularly of HPMC is used as binder. The aforementioned amounts refer to the amount of all binders (i.e., including mixtures) in the composition.


It is preferred to use an outer layer of immediate release methylphenidate HCl amount of about 1% to about 30% by weight, of about 5% to about 28% by weight, of about 15% to about 27% by weight, of about 18% to about 25% by weight, such as of about 19.0% to about 25.0% by weight, of about 19.1% by weight, of about 19.2% by weight, of about 19.3% by weight, of about 19.4% by weight, of about 19.5% by weight, of about 19.6% by weight, of about 19.7% by weight, of about 19.8% by weight, of about 19.9% by weight, of about 20.0% by weight, of about 20.1% by weight, of about 20.2% by weight, of about 20.3% by weight, of about 20.4% by weight, of about 20.5% by weight, of about 20.6% by weight, of about 20.7% by weight, of about 20.8% by weight, of about 20.9% by weight or of about 21.0% by weight, of about 21.1% by weight, of about 21.2% by weight, of about 21.3% by weight, of about 21.4% by weight, of about 21.5% by weight, of about 21.6% by weight, of about 21.7% by weight, of about 21.8% by weight, of about 21.9% by weight or of about 22.0% by weight, based on the weight of the pharmaceutical composition. An amount of about 18.0% to about 22.0% by weight based on the weight of the pharmaceutical composition can be preferred. The aforementioned amounts refer to the amount of methylphenidate hydrochloride or its respective amount of the base or any of its salts in the outer immediate release layer composition.


It is also preferred to use an inner core layer of immediate release methylphenidate HCl amount of about 1% to about 99% by weight, of about 5% to about 95% by weight, of about 60 to about 90% by weight, of about 70% to about 85% by weight, such as of about 73.0% to about 83.0% by weight, of about 79.0% by weight, of about 79.1% by weight, of about 79.2% by weight, of about 79.3% by weight, of about 79.4% by weight, of about 79.5% by weight, of about 79.6% by weight, of about 79.7% by weight, of about 79.8% by weight, of about 79.9% by weight, of about 80.0% by weight, of about 80.1% by weight, of about 80.2% by weight, of about 80.3% by weight, of about 80.4% by weight, of about 80.5% by weight, of about 80.6% by weight, of about 80.7% by weight, of about 80.8% by weight, of about 80.9% by weight or of about 81.0% by weight, of about 81.1% by weight, of about 81.2% by weight, of about 81.3% by weight, of about 81.4% by weight, of about 81.5% by weight, of about 81.6% by weight, of about 81.7% by weight, of about 81.8% by weight, of about 81.9% by weight or of about 82.0% by weight, based on the weight of the pharmaceutical composition. An amount of about 78.0% to about 82.0% by weight based on the weight of the pharmaceutical composition can be preferred. The aforementioned amounts refer to the amount of methylphenidate hydrochloride or its respective amount of the base or any of its salts in the inner core immediate release layer composition.


As described above, several solid dose controlled-release formulations of some or all commercially available methylphenidate are commercially available in the market. However, the therapeutic effect of some or all of those formulations is not expected to last for more than 12 hours after administration.


An advantage of the highly preferred embodiment of the present invention is believed to be that the formulation that will have a therapeutic effect of at least 14 hours of duration or more. To achieve this, the delivery of methylphenidate in the distal part of the GI tract was investigated to prolong the duration of action of the drug. The present inventors are unaware of an example of actual reported example the delivery of methylphenidate in the distal part of the GI tract. Also, no relevant in vivo data was found reporting the release of methylphenidate or its pharmaceutically acceptable salts in the distal part of the GI tract.


Methylphenidate hydrochloride is freely soluble in water and methanol, soluble in alcohol, slightly soluble in chloroform and acetone; melts between 224-226° C.; and has a pKa of approximately 8.8. Methylphenidate is relatively stable in acidic solutions but it is degraded extensively in basic solutions. The degradation occurs via the hydrolysis of the methyl ester to the free acid, α-phenyl 1-2-piperidineacetic acid. Therefore, the degradation amount increases up to 100% as the pH increases to 8.9. See Chemical Stability of Pharmaceuticals a Handbook for Pharmacists 1986, 587-590 (Kenneth A. Connors, Gordon L. Amidon and Valentino J. Stella) and Analytical Profiles of Drug Substances. 1981, 473-497. Gandharva R. Padmanabhan for additional information.


As a result, it was not known whether methylphenidate could be absorbed systematically in sufficient amount to have therapeutic effect at distal locations within the GI tract where the pH is known to be above pH 6.0. Thus, the present inventors performed in vitro and in vivo studies to determine the amount released and the extent of absorption of several methylphenidate controlled release formulations. Table 1 shows some of the different formulations explored; Table 2 shows their correspondent in vitro dissolution data and FIGS. 1-4 shows their respective in vivo results.


The preferred oral dosage form of the present invention is a capsule containing multilayer release (MLR) beads which have multiple layers to achieve the desire release rate profile of methylphenidate hydrochloride. Some of those layers have immediate release and controlled release components. It is made up of a controlled release bead which is coated to delay dissolution until it has reached the distal part of the GI tract. The distal coated controlled release bead has an immediate release topcoat to provide an initial rate of absorption needed to have the desired therapeutic effect. In a highly preferred embodiment, the immediate release component represents 20% of the total dose per bead and the controlled release component represents 80% of the total dose per bead. This formulation is designed to produce rapid rise to therapeutic plasma levels after oral administration, due to the rapid dissolution and absorption of the outer immediate release layer, followed by a period of minimum absorption and then controlled release of the immediate release core, plasma levels would then gradually decrease according to the elimination kinetics of methylphenidate.


In a preferred embodiment, the pharmaceutical dosage forms comprise methylphenidate or a pharmaceutical acceptable salt or derivate thereof as the sole pharmaceutically active agent.


The pharmaceutical composition of methylphenidate HCl, Controlled Release Capsules (e.g., Formulation I and J 80:20 described below) MPH IR distal beads may comprise about 1 to 150 mg such as about 15 mg, 25 mg, 30 mg, 35 mg, 45 mg, 55 mg 70 mg, 85 mg 100 mg and 120 mg


The present coated beads are preferably formulated as a single multilayer coated bead into an oral solid pharceutical compositions, preferably in the form of a capsule. The capsule material is preferably a hard gelatin capsule or a hard HPMC capsule. Other capsule materials may also be used and the selection thereof is within the purview of a person of ordinary skill in the art.


In other embodiments of the present invention, it may be preferred to provide an outer layer on the coated bead, wherein the outer layer comprises one or more salts of alginic acid. The salts of alginic acid may be selected from sodium alginate, potassium alginate, magnesium alginate, lithium alginate or ammonium alginate or mixtures thereof. Preferably, the salts of alginic acid may have a viscosity of 30 to 720, preferably 40 to 450, preferably 40 to 400 or preferably 50 to 300 centipoise (cp) of a 1% aqueous solution (weight/weight). The provision of such an outer layer can improve the coated bead resistance to alcohol (e.g., ethanol) in concentrations of greater than 35% (volume/volume)—e.g., 40% (volume/volume)—since such the presence of alcohol (e.g., ethanol) in the gastric fluid usually leads to an increase to the release rates in the stomach. Due to distribution effect, the effect of ingested ethanol in the intestine is not of the same importance as in the stomach. A preferred embodiment of the invention relates to the use of coating layers described above in the coated bead to confer resistance to alcohol (e.g., ethanol) in concentrations of up to about 35% (volume/volume) in the gastric fluid without the need to use a coating comprising one or more salts of alginic acid. Thus an effective protection against the influence of ethanol should prevent such an undesired increase of pharmaceutical or nutraceutical active ingredient in the stomach in the first place. Furthermore it may be desired that protection against the influence of ethanol shall at least not influence the comparably fast release rates at pH 6.8 in media without ethanol.


Embodiments of the present invention will be illustrated with references to the following examples which should not be used to limit or otherwise construe the invention.


In the examples the following abbreviations are used:

    • IR beads—these are beads coated with the methylphenidate hydrochloride (MPH) and having no controlled or delayed release coating;
    • CRIR beads—these are IR beads which have been coated with a controlled release coating;
    • ECCRIR—these are CRIR beads which have been coated with an enteric coating (similar to commercially available Biphentin® product);
    • DRIR beads—these are IR beads which have been coated with a delayed or distal release coating;
    • DRCRIR beads—these are CRIR beads which have been coated with a delayed (enteric coating [ECCRIR]) or distal release coating; and
    • MPH IR distal beads—these are DRCRIR beads which have been coated with an immediate release layer of MPH.


Various of these beads were coated with sodium alginate which can confer resistance to 40% alcohol.


The general method of manufacture of Formulation I or Formulation J is described below followed by the various studies and findings.


The MPH IR distal beads were manufactured in four different stages involving separate coating process at each stage—immediate-release coating (drug layering [IR]), controlled-release coating [CR], distal release coating [DR] and top immediate release coating [CRDRIR]. All four stages are carried out in a fluid bed dryer with Wurster column. More detailed information of the process parameters used at each stage of the manufacturing process is found in Tables 9-12, respectively.


In some cases, a layer of sodium alginate-talc is also applied as a fifth stage on top of the top immediate release coating to improve dose dumping of methylphenidate HCl in hydro-ethanolic solutions compared with coated bead formulations that do not have a layer comprising one or more salts of alginic acid without affecting the immediate release performance of the top immediate release coating. More detailed information of the process parameters used at this stage of the manufacturing process is found in Table 13.


The following is a description of the manufacturing process.


Example 1—IR Beads

The following protocol was used to produce the IR beads.


Add Opadry clear YS-1-7006 to water and mix, then add methylphenidate hydrochloride and stir until a clear solution is obtained.


Charge fluid bed dryer (FBD) equipped with Wurster column with sugar spheres 14-18 or 18-20 mesh.


Coat the beads at an inlet temperature of 65°±10° C. and product temperature of 37.5°±3.5° C. by spraying the solution of methylphenidate hydrochloride.


After completion of the spraying process, allow the beads to dry at 41°±4° C. for approximately 3 minutes.


Cool the beads to 30°±1° C. product temperature and weigh.


Example 2—CRIR Beads

The following protocol was used to produce the methylphenidate CR beads.


Prepare a coating dispersion by mixing Plasacryl T20, filtered (250 micrometer screen) Eudragit RS 30 D and Triethyl Citrate in a container for at least 60 minutes.


Charge fluid bed dryer (FBD) equipped with Wurster column with IR beads.


Coat the IR beads at product temperature of 25°±5° C. by spraying the coating dispersion.


After completion of the coating dispersion, spray rinse water at a product temperature of 25°±5° C.


Example 3—DRCRIR Beads, Uncured

Charge fluid bed dryer (FBD) equipped with Wurster column with CRIR beads.


Prepare a coating dispersion by mixing Plasacryl T20, filtered (250 micrometer screen) Eudragit FS 30 D and water in a container for at least 60 minutes.


Coat the CRIR beads with Eudragit FS30D dispersion.


Note: In case the manufacturing process is interrupted, Syloid 244FP, quantity based on 0.43% of the theoretical yield of CRDR beads, is added to the beads and blended.


Example 4—DRCRIR Beads, Cured

The following protocol was used to produce the methylphenidate DRCR beads.


Charge fluid bed dryer (FBD) equipped with Wurster column with CRIR beads.


Prepare a coating dispersion by mixing Plasacryl T20, filtered (250 micrometer screen) Eudragit FS 30D and water in a container for at least 60 minutes.


Coat the CRIR beads with Eudragit FS30D dispersion at product temperature of 25°±3° C. by spraying the coating dispersion.


After completion of the coating dispersion, spray rinse water at a product temperature of 25°±3° C.


Suck into the FBD, Syloid 244FP, quantity based on 0.43% of the theoretical yield of CRDR beads and blend.


Cure the beads at a product temperature of 40°±2° C. for 60 minutes.


Cool the beads to product temperature of 25°±3° C.


Screen the beads on 0.85 mm screen and remove fines if any.


Note: In case the manufacturing process is interrupted, Syloid 244FP, quantity based on 0.43% of the theoretical yield of CRDR beads, is added to the beads and blended.


Example 5—MPH IR Distal Beads

The following protocol was used to produce the MPH IR distal beads.


Add Opadry clear YS-1-7006 to water and mix, then add methylphenidate hydrochloride and stir until a clear solution is obtained.


Charge fluid bed dryer (FBD) equipped with Wurster column with DRCRIR beads


Coat the beads at an inlet temperature of 56°±15° C. and product temperature of 37.5°±3.5° C. by spraying the solution of methylphenidate hydrochloride.


After completion of the solution, spray rinse water at a product temperature of 37.5°±3.5° C.


Allow the beads to dry at product temperature of 41°±4° C. for 5 minutes.


Cool the beads to 30°±1° C. product temperature and weigh.


Screen the beads and collect the beads passing through 1.8 mm screen and retained on 0.85 mm screen.


Example 6—MPH IR Distal Beads Coated with Sodium Alginate Beads

The following protocol was used to produce the MPH IR distal beads coated with sodium alginate beads.


Add talc to water and mix; stir using a homogenizer until a uniform dispersion is obtained.


Add sodium alginate to water and mix, stir until a uniform dispersion is obtained.


Add the talc dispersion on the sodium alginate and mix until a uniform dispersion is obtained.


Charge fluid bed dryer (FBD) equipped with Wurster column with MPH IR distal beads.


Coat the beads at an inlet temperature of 70°±15° C. and product temperature of 50°±5° C. by spraying the solution of sodium alginate.


Allow the beads to dry at product temperature of 41°±4° C. for 5 minutes.


Cool the beads to 30°±1° C. product temperature and weigh.


Screen the beads and collect the beads passing through 1.8 mm screen and retained on 0.85 mm screen.


Example 7—MPH IR Distal Beads (with or without Sodium Aginate) with Silicon Dioxide

The following protocol was used to produce these beads.


Charge V blender with approximately half the total quantity of MPH IR Distal Beads (with or without sodium aginate).


Screen Syloid FP 244 through 20 mesh screen and add to the V blender.


Load remaining quantity of MPH IR Distal Beads (with or without sodium aginate) into the V blender.


Blend for 3 minutes.


Discharge the blend into plastic drums lined with polyethylene bags.


Example 8—Encapsulation of MPH IR Distal Beads (with or without Sodium Aginate) with Silicon Dioxide

The following equipment is used during the capsule filling process of the MPH IR Distal Beads (with or without sodium aginate) with silicon dioxide in either hard gelatin capsules (used in these Examples) or hard hypromellose (HPMC) capsules (an alternative to hard gelatin capsules):

    • Bosch GKF 1400 Encapsulator & Checkweigher
    • Metal Detector
    • Empty Capsule Conveying Bin


Example 9—Testing (PK Studies and Preliminary Studies on Alcohol Resistance)

The following methodology was used.


The dissolution of various formulations was performed using USP paddle method at 100 rpm in 900 ml at 37° C. of simulated gastric fluid (without enzyme) for 2 hours, 900 ml phosphate buffer pH 6.0 for 4 hours and 7th hour onwards, 900 mL of phosphate buffer pH 7.4. The samples were withdrawn at the respective time points and analysed on HPLC using UV detector. The in vitro release data is indicated as percentage dissolved based on the label content of the active tested.


The results of a bioavailability study of this formulation indicate a biphasic release profile (FIG. 5).


It can be concluded from the in vitro dissolution data and its correspondent in vivo plasma concentration that methylphenidate can be absorbed in the distal part of the GI tract. It can also be concluded that the amount and extent of methylphenidate being absorbed depends on the excipients used in the formulation.



FIG. 1 shows that 7 to 10% of the controlled release polymer might be sufficient to provide a therapeutic effect that lasts for more than 14 hours but without the desired rapid on set of action and distinctive biphasic or triphasic pattern shown in FIG. 3 or 4. However, the next study showed that increasing the amount of the controlled release polymer up to 20% as shown in FIG. 2 prolonged the extent of release of methylphenidate. Nonetheless, the higher the amount of controlled release polymer, the less the total amount of methylphenidate is absorbed. This could be due to the degradation of methylphenidate at higher pH environments, thus less amount of methylphenidate is available at distal part of the GI tract to be absorbed in systemic circulation.


Therefore, the amount of controlled release polymer needs to be adjusted accordingly to achieve the desired distinctive in vivo plasma concentration pattern. In the case of the preferred embodiments of the present invention, the longer duration of action and distinctive pattern might be achieved between 7% to 20% weight gain of the controlled release polymer, more specifically about 16% weight gain of the controlled polymer. The 16% would provide the desired total amount and extent of methylphenidate in plasma concentration over time in a distinctive pattern that differentiates this formulation from any other long acting solid dose methylphenidate formulation available in the market. Moreover, it achieves duration of action of no less than 14 hours.


Formulation I also has the property that does not undergo food effect as shown in FIG. 5 and Table 3. It can also be sprinkled on apple sauce, yogurt or ice cream for up to 10 minutes without affecting its bioavailability performance as shown in FIG. 6 and Table 5. Compared to three equivalent doses of immediate-release methylphenidate administered separately at 4 hourly intervals, Formulation I has greater residual levels of methylphenidate at hour 24 post-administration and different partial AUCs during the dosing interval, particularly in the 12-16 hour period where the pAUC is significantly larger than immediate-release methylphenidate (Table 4). In addition, the second peak of methylphenidate occurs more than 2 hours after the third peak of immediate-release methylphenidate (FIG. 5 and Table 4). As a result of the significant residual methylphenidate plasma levels at hour 24 post-administration (FIG. 5), the pharmacokinetic profile changes after multiple days of dosing resulting in an overall increase in plasma levels (FIG. 7 and Table 6) and higher peak concentrations. Formulation J has a similar pharmacokinetic profile and properties as Formulation I (FIG. 8 and Table 7).


The in vitro dissolution specifications of the drug at various time points for formulations in accordance with Formulations A-J are shown in Table 2. Based on these results and the correlation between in vivo and in vitro data, the present inventors developed the target specification should in Table 8 for preferred embodiments of the present coated bead.


Based on the studies excipients were identified and adjusted to obtain a finished product that is stable within a product shelf life of at least 24 months and provides no less than 14 hours of therapeutic effect. Stability testing of the above formulation showed that the total related substances at 6 months 40° C./75% RH are within 2.0% and no individual unknown is higher than 0.2%.


In vitro dissolution testing at 40% ethanol in SFG dissolution media was performed. As will be illustrated in Example 10, Formulation I and Formulation J were found to be resistant up to about 35% v/v and up to about 32% v/v, respectively, ethanol in SGF. Therefore, different trials were performed with immediate release excipients to be applied to the outer immediate release layer methylphenidate hydrochloride to increase the ethanol resistance to 40% ethanol v/v. Some of the excipients that were investigated individually or in combination are: sodium alginate, Kollicoat™ IR, hypromellose, Lycoat™, pectin, lactose, methylcellulose, ethylcellulose and talc. An outer layer of these excipients was applied on top of the desired methylphenidate CRDRIR formulation or formulation I/Formulation J and a test to determine in vitro alcohol resistance (see Example 10 for details of the test) was performed to determine the impact of the excipients in the formulation.


The experiment with sodium alginate in combination with talc showed that above 40% weight gain of sodium alginate the dissolution rate in 40% v/v ethanol would impart alcohol resistance to Formulation I under the prescribed test conditions. At weight gains between 55 to 75% the dissolution rate in the first two hours of the product would meet the criteria at 40% v/v ethanol and released about 20% of the IR component. Therefore, an improved CRDRIRR methylphenidate formulation (Formulation I or Formulation J) was developed. This formulation would have a most outer layer of about 55-75% weight gain of sodium alginate. This layer is applied on top of the external IR layer to provide a 40% v/v ethanol resistant formulation without affecting the original release rate of the formulation.


Example 10—Testing (Further Studies on Alcohol Resistance)

Generally dose dumping is observed as a result of a compromise of the release-rate controlling mechanism in a pharmaceutical product. Some products can be expected to exhibit a more rapid drug dissolution and release rate in the presence of ethanol. Therefore, when a modified—release product is consumed with alcohol, the modified-release mechanism could be adversely affected, which could lead to dose dumping.


The following study was performed to evaluate the alcohol induced dose dumping in IRDR Methylphenidate HCl capsules. The effect of varying concentrations of ethanol on the drug release was evaluated at 0% (no ethanol added), 5%, 20% and 40% ethanol which are considered to be representative of consumption of beer (5% ethanol), mixed drinks (20% ethanol), and neat liquor (40% ethanol). The dissolution evaluation was also carried out in 35% ethanol to understand the effect of ethanol concentrations from 20% to 40% and at what level the alcohol induced dose dumping becomes significant.


The dissolution profiles showed that even though in the presence of 40% ethanol, the rate of dissolution rapidly increased as compared to that observed in control, the release was never considered to be dose dumping of methylphenidate HCl. Furthermore, in the presence of 35% ethanol the rate of release increased but the average percentage release amount was determined to be similar when the f2, similarity factor, was calculated against the control sample. The calculated value was 50. An f2 value of 50-100 suggests similar dissolution profiles.


The experiments were carried out on 12 units as following: The ethanolic dissolution media used were 5%, 20%, 35% and 40% USP anhydrous ethanol in 0.1N HCl (v/v). The experiments were performed in 900 ml of respective media using USP apparatus 1 (baskets) at 100 rpm and 37° C. The control (0% ethanol) was also run using 900 ml of 0.1 N HCl. The 0.1 N HCl was selected to approximate the conditions in the stomach.


The samples were collected every 15 minutes up to 2 hours to understand the release profile starting as early as 15 minutes. Since the dissolution experiments were run for 2 hours, and the vessels were covered at all times, the media evaporation had no impact on the results. The samples were analyzed on HPLC as per specified IRDR Methylphenidate HCl capsule dissolution method and the percent released methylphenidate HCl at each time point was calculated. The dissolution and HPLC parameters are reported in Table 14.


Resistance to ethanol means that the release of the pharmaceutical active ingredient is in the presence of ethanol not more than 20% to be measured under in-vitro conditions at pH 1.2 for 2 hours in 900 mL medium according to USP with the addition of 5, 10, 20 or 40% (v/v) ethanol at 100 rpm using USP. Dose Dumping is defined as unintended, rapid drug release in a short period of time of a significant amount of the drug contained in a modified release dosage form. Dose dumping shall mean that, the release of the pharmaceutical active ingredient is faster but does not release more than 25%, no more than 20% to be measured under in-vitro conditions at pH 1.2 for 60 minutes in medium according to USP with the addition of 5, 10, 20 or 40%) (v/v) ethanol.


In this study, the focus was on coating application as a function of theoretical weight gain of coating applied to the nonpareil beads. Since it is also common to quantify film coating amount as mass/surface area, film coating amount (mg/cm2) was determined using a calculation for surface area, assuming the bead is a perfect sphere:

SA=4(πr2)

wherein SA is the surface area and r is the radius of the bead.


Conventional round nonpareil beads with diameters ranging from 0.85 to 1.4 mm with an average of 1.125 mm were used in this ethanol resistance study. The surface area of beads of this average diameter was calculated as follows:

SA=4(π(0.56252))
SA=3.98 mm2


Conventional round nonpareil beads with diameters ranging from 0.85 to 1.4 mm with an average of 1.125 mm were used in this ethanol resistance study.


Since a certain layer thickness is describable in film coating with Eudragit FS30D to impart alcohol resistant properties up to 30%, up to 32%, up to 35% without the capsules possessing dose dumping characteristics of the active pharmaceutical ingredient within the first 60 minutes; the amount of coating material is related to the surface area of the substrate per cm2 of surface area. Thus, the inventors divided the surface area of a substrate A (mm2) by its weight gain w (mg), to obtain the desired coating quantity in % (w/w), i.e. as shown in the following equation:

Coating weight (%)=[Amm2)/w(mg)]*1(mg/cm2)


The total amount of the delayed (or distal) release (e.g., colonic delivery) material may be in the range of from about 5% to about 35% by weight, preferably from about 10% to 30% by weight, most preferably from about 15 to about 25% by weight, in relation to the weight of the core.


The absolute amount of the delayed (or distal) release (e.g., colonic delivery) material described above (prior to the examples) may, in the case of pellets or granules with a diameter size in the range of from about 840 to 1410 μm, be present at an average thickness in the range in from about 5 μm to about 50 μm, preferably from about 10 μm to about 50 μm, more preferably from about 33 μm to about 47 μm, most preferably about 40 μm.


It can be preferred to use poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) 7:3:1 as the delayed (or distal) release (e.g., colonic delivery) material. Such a material is commercially available from Evonik under the tradement name Eudragit® FS30D.


The presence of ethanol in concentrations of 5, 10, 20 or 40% volume/volume) in the gastric fluid usually leads to an increase to the release rates in the stomach. Due to distribution effect the effect of ingested ethanol is in the intestine not of that importance as in the stomach. Thus an effective protection against the influence of ethanol should prevent such an undesired increase of pharmaceutical active ingredient in the stomach in the first place. Furthermore it may be desired that protection against the influence of ethanol shall at least not influence the comparably fast release rates at pH 6.8 in media without ethanol.


International Patent Publication WO 2012/0224998 describes a gastric resistant pharmaceutical or nutraceutical composition, comprising a core, comprising a pharmaceutical or nutraceutical active ingredient and a gastric resistant coating layer onto the core, wherein the release of the pharmaceutical active ingredient is not more than 15% under in-vitro conditions at pH 1 0.2 for 2 hours in a buffered medium according to USP with and without the addition of 40% (v/v) ethanol, wherein the gastric resistant coating layer comprises 10 to 100% by weight of one or more salts of alginic acid with a viscosity of 30 to 720 cP of a 1% aqueous solution. The one layer system as described is stated to solve the problem of protection against the influence of ethanol. However, there is no reference of ethanol protection provided by coating layer containing Eudragit™ FS30D at any ethanol concentration by itself or when the coatings which include the ammonium alginate, coatings which employ other alginate salts, like sodium or potassium alginate, are deposited in the most outer layer of the bead.


The data demonstrated that the in-vitro rate of dissolution of methylphenidate HCl did not increase in the presence of 5% and 20% ethanol within two hours as compared to that in control; and the in-vitro rate of dissolution of methylphenidate HCl did not increase in the presence of 5%, 20%, 35% and 40% ethanol within 30 minutes as compared to that in control. Nevertheless, in the presence of 35% and 40% ethanol, more rapid increase was observed in the dissolution release rate after 30 minutes. Even though a faster release was observed, the release of the active pharmaceutical ingredients was still in a controlled release manner. Dose dumping of methylphenidate hydrochloride did not occur at any time of the release in presence of different concentration of ethanol up to 40%.


The results of this study are believed to be a reasonable basis for the present inventors to predict similar resistance to alcohol (e.g., ethanol) would be observed clinically and for active ingredients other than methylphenidate HCl.


Example 11—A Randomized, Double-Blind Study of the Time Course of Response of MPH-IR Distal Bead (Formulation I) in Adult with ADHD in a Simulated Adult Workplace Environment (AWE)
Objectives

The purpose of this randomized, double-blind, crossover, placebo-controlled, optimized-dose study was to assess the clinical efficacy, time of onset and time course of efficacy over 16 hours of MPH-IR Distal Bead compared to placebo in adults diagnosed with ADHD in an AWE setting.


Methodology

This study (063-008) was a randomized, double-blind, placebo-controlled cross-over study in adult, male and female ADHD subjects conducted to assess clinical efficacy, the time of onset and time course of efficacy of MPH-IR Distal Bead measured by the Permanent Product Measure of Performance (PERMP) (an objective, skill-adjusted math test that measures attention in ADHD) score. Subjects were titrated to an optimal dose in an open-label phase of between 2 and 7 weeks, familiarized with study procedures in a practice AWE session and then randomized to one of two sequences (ACTIVE to PLACEBO or PLACEBO to ACTIVE) and received one treatment for one week, followed by an AWE session, then crossed over to the other treatment for one week, followed by a second AWE session.


Number of Subjects


Planned: 60 subjects. Randomised: 59 subjects. Completed: 46 subjects.


Test Treatment, Dose, and Mode of Administration

Active or matching placebo MPH-IR Distal Bead (methylphenidate hydrochloride controlled-release—Formulation I in Table 1) 25, 35, 45, 55, 70, 85 or 100 mg oral capsules were administered once-daily in the morning.


Duration of Treatment


Subjects received open label medication during a 2 to 9 week dose titration, followed by a double-blind crossover of one week of placebo treatment and one week of active treatment.


Criteria for Evaluation


The primary outcome measure was the mean between-treatment PERMP Total score across the AWE sessions. Secondary outcome measures included the onset and time course of efficacy of MPH-IR Distal Bead compared to placebo as measured by the PERMP Total Score (PERMP-T), PERMP Attempted Score (PERMP-A) and PERMP Correct Score (PERMP-C) at pre-dose and 1.0, 2.0, 5.0, 8.0, 11.0, 14.0 and 16.0 hours post-dose and the onset and time course of efficacy of MPH-IR Distal Bead compared to placebo as measured by the SKAMP (a subjective measure of behaviour), using the combined score (SKAMP-C), the SKAMP-Deportment (SKAMP-D) subscale and SKAMP Attention (SKAMP-A) subscale at pre-dose and 0.5, 1.0, 2.0, 4.0, 5.0, 7.0, 8.0, 11.0, 13.0, 14.0 and 16.0 hours post-dose.


Efficacy & Safety Results


The study met the primary endpoint in that subjects treated with MPH-IR Distal Bead had improved attention compared to subjects receiving placebo, as measured by the mean change from pre-dose PERMP-Total Scores (FIG. 9).


Subjects receiving MPH-IR Distal Bead showed improvement in attention with an onset of action within 1.0 hour of receiving active medication compared to placebo with duration of effect continuing for up to and including 16.0 hours post-dose, based on change from pre-dose LS mean difference from placebo PERMP-Total Scores (FIG. 9).


Subjects receiving MPH-IR Distal Bead showed improvement in behaviour with an onset of action within 1.0 hour of receiving active medication compared to placebo with duration of effect continuing for up to and including 16.0 hours post-dose based on change from pre-dose LS mean difference from placebo SKAMP-C Scores (FIG. 10).


MPH-IR Distal Bead was relatively safe and well-tolerated medication.


Overall Conclusions

MPH-IR Distal Bead was safe and effective in the treatment of adults with ADHD, demonstrating efficacy from one hour to 16 hours post administration on both objective and subjective measures. Subjects demonstrated significant improvement in the primary endpoint—an objective measure of attention (the PERMP)—and the secondary endpoint—a subjective measure of behaviour (the SKAMP)—during the double-blind phase of the study when treated with MPH-IR Distal Bead compared to when treated with placebo.


In addition, the study medication was well-tolerated, with no serious adverse events. Patients reported satisfaction with ability to fall asleep, appetite for lunch, appetite for dinner or overall adverse effects and no significant differences compared to placebo in sleep quality.


The onset and duration of action at one hour and 16 hours post-administration respectively, is the result of the pharmacokinetic profile of MPH-IR Distal Bead. The residual methylphenidate plasma concentration at hour 24 post-administration leads to an increase in the first peak of methylphenidate following multiple days of dosing, resulting in an onset of action with one hour. In addition, the second peak of methylphenidate is also increased following multiple days of dosing, providing sufficient plasma levels of methylphenidate late in the day that results in a prolonged duration of action extending to 16 hours post-administration. The pharmacokinetic profile of this formulation provides a combination of rapid onset and a prolonged duration of action in a single daily administration.


Example 12—Preferred Formulations

Based on the exemplary work described above, Formulations I and J were identified as the most preferred for the present coated bead. Tables 15 and 16 provide complete formulation specifications for oral solid pharmaceutical composition based on Formulations I and J, respectively for the following dosage strengths of methylphenidate HCl: 25 mg, 30, mg, 35 mg, 45 mg, 55 mg, 70 mg, 85 mg and 100 mg.


While this invention has been described with reference to illustrative embodiments and examples, the description is not intended to be construed in a limiting sense. Thus, various modifications of the illustrative embodiments, as well as other embodiments of the invention, will be apparent to persons skilled in the art upon reference to this description. It is therefore contemplated that the appended claims will cover any such modifications or embodiments.


All publications, patents and patent applications referred to herein are incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.











TABLE 1








Strength (label claim) 20 mg
Strength (label claim) 20 mg












Formulation A - 7%
Formulation B - 10%
Formulation C - 16%
Formulation D - 20%



063-001
063-001
063-002
063-002



DRCRIR bead
DRCRIR bead
DRCRIR bead
DRCRIR bead
















Quantity

Quantity

Quantity

Quantity



Ingredient
per unit
%
per unit
%
per unit
%
per unit
%





Methylphenidate HCl, USP
20.0
11.25
20.0
10.88
20.0
10.19
20.0
9.79


Sugar spheres 14/18 mesh, USP/NF
107.0
60.2
107.0
58.2
107.0
54.5
106.8
52.3


Opadry Clear YS-1-7006, USP
6.4
3.6
6.4
3.5
6.5
3.3
6.4
3.1


Ammonio methacrylate copolymer
9.4
5.26
13.3
7.24
21.3
10.86
26.7
13.05


dispersion, Type B, 30% dispersion










(Eudragit RS30D Solids), NF










Triethyl Citrate, USP/NF
1.4
0.80
2.0
1.10
3.3
1.67
4.1
2.00


Glyceryl monostearate emulsion
3.8
2.18
4.3
2.37
5.3
2.74
6.0
2.94


(Plasacryl T20), HS










Silicon dioxide, (Syloid 244FP), NF
0.7
0.4
0.7
0.4
0.8
0.4
0.8
0.4


Eudragit FS30D Liquid, HS
29.0
16.32
30.0
16.32
32.0
16.32
33.3
16.32


Total weigh in capsule (mg)
~178
~100
~184
~100
~196
~100
~204
~100













Strength (label claim) 30 mg
Strength (label claim) 30 mg



Formulation E - 70:30 063-002
Formulation F - 80:20 063-003



ECCRIR bead (70%) + DRCRIR bead (30%)
DRCRIR bead (80%) + IR bead (20%)


Ingredient
Quantity per unit (%)
Quantity per unit (%)












1. IR bead










IR bead (0%)
IR bead (20%)





Methylphenidate HCl, USP

15.0


Sugar spheres 14/18 mesh, USP/NF

80.2


Opadry Clear YS-1-7006, USP

4.8


Total IR bead

~100%












7. CR/EC/IR bead










ECCRIR bead (70%)
ECCRIR bead (0%)





Methylphenidate HCl, USP
12.78



Sugar spheres 14/18 mesh, USP/NF
62.02



Opadry Clear YS-1-7006, USP
4.04



Ammonio methacrylate copolymer
5.44



dispersion, Type B, 30% dispersion




(Eudragit RS30D Solids), NF




Methacrylic Acid copolymer dispersion, 30%
8.15



dispersion (Eudragit L30 D-55 solids), USP NF




Triethyl Citrate, USP/NF
2.70



Talc Ph. Eur/USP
4.87



Total CR/EC/IR bead
~100%
~100%












6. CR/DR bead










DRCRIR bead (30%)
DRCRIR bead (80%)





Methylphenidate HCl, USP
10.19
10.19


Sugar spheres 14/18 mesh, USP/NF
54.5
54.5


Opadry Clear YS-1-7006, USP
3.3
3.3


Ammonio methacrylate copolymer
10.86
10.86


dispersion, Type B, 30% dispersion




(Eudragit RS30D Solids), NF




Triethyl Citrate, USP/NF
1.67
1.67


Glyceryl monostearate emulsion
2.74
2.74


(Plasacryl T20), HS




Silicon dioxide, (Syloid 244FP), NF
0.4
0.4


Eudragit FS30D Liquid, HS
16.32
16.32


Total CR/DR bead (30%)
~100%
~100%


Total weight in capsule
~211 mg
~275 mg






Strength (label claim) 30 mg
Strength (label claim) 30 mg



Formulation G - 30:55:15 063-003
Formulation H - 35:55:10 063-003



ECCRIR bead (30%) + DRCRIR bead
ECCRIR bead (35%) + DRCRIR bead



(55%) + IR bead (15%)
(55%) + IR bead (10%)


Ingredient
Quantity per unit (%)
Quantity per unit (%)












1. IR bead










IR bead (15%)
IR bead (10%)





Methylphenidate HCl, USP
15.0
15.0


Sugar spheres 14/18 mesh, USP/NF
80.2
80.2


Opadry Clear YS-1-7006, USP
4.8
4.8


Total IR bead
~100%
~100%












7. CR/EC/IR bead










ECCRIR bead (30%)
ECCRIR bead (35%)





Methylphenidate HCl, USP
12.78
12.78


Sugar spheres 14/18 mesh, USP/NF
62.02
62.02


Opadry Clear YS-1-7006, USP
4.04
4.04


Ammonio methacrylate copolymer
5.44
5.44


dispersion, Type B, 30% dispersion




(Eudragit RS30D Solids), NF




Methacrylic Acid copolymer dispersion,
8.15
8.15


30% dispersion




(Eudragit L30 D-55 solids), USP NF




Triethyl Citrate, USP/NF
2.70
2.70


Talc Ph. Eur/USP
4.87
4.87


Total CR/EC/IR bead
~100%
~100%












6. CR/DR bead










DRCRIR bead (55%)
DRCRIR bead (55%)





Methylphenidate HCl, USP
10.19
10.19


Sugar spheres 14/18 mesh, USP/NF
54.5
54.5


Opadry Clear YS-1-7006, USP
3.3
3.3


Ammonio methacrylate copolymer
10.86
10.86


dispersion, Type B, 30% dispersion




(Eudragit RS30D Solids), NF




Triethyl Citrate, USP/NF
1.67
1.67


Glyceryl monostearate emulsion
2.74
2.74


(Plasacryl T20), HS




Silicon dioxide, (Syloid 244FP), NF
0.4
0.4


Eudragit FS30D Liquid, HS
16.32
16.32


Total CR/DR bead (30%)
~100%
~100%


Total weight in capsule
~262 mg
~264 mg















Strength (label claim)





100 mg





Formulation I 80:20
Strength (label claim)



Strength (label claim) 100 mg
063-004, 063-005,
100 mg



Formulation F 80:20
063-007
Formulation J 80:20



063-003
063-008
063-011



DRCRIR bead + IR bead
MPH IR distal bead
MPH IR distal bead


Ingredient
Quantity per unit (%)
Quantity per unit (%)
Quantity per unit (%)












1. IR bead











IR bead (20%)
IR bead (0%)
IR bead (0%)





Methylphenidate HCl, USP
15.0




Sugar spheres 14/18 mesh, USP/NF
80.2




Opadry Clear YS-1-7006, USP
4.8




Total IR bead
~100%














6. CR/DR bead











DRCRIR bead (80%)
DRCRIR bead (0%)
DRCRIR bead (0%)





Methylphenidate HCl, USP
10.19




Sugar spheres 14/18 mesh, USP/NF
54.5




Opadry Clear YS-1-7006, USP
3.3




Ammonio methacrylate copolymer
10.86




dispersion, Type B, 30% dispersion





(Eudragit RS30D Solids), NF





Triethyl Citrate, USP/NF
1.67




Glyceryl monostearate emulsion
2.74




(Plasacryl T20), HS





Silicon dioxide, (Syloid 244FP), NF
0.4




Eudragit FS30D Liquid, HS
16.32




Total CR/DR bead
~100%














8. CR/DR/IR bead












MPH IR distal bead
MPH IR distal bead













MPH IR
Quantity per

Quantity per




distal bead
unit (mg)
%
unit (mg)
%





Methylphenidate HCl, USP

100
12.3
100
18.4


Sugar spheres 14/18 mesh or 18/20 mesh,

428.5
52.7
243.1
44.7


USP/NF







Opadry Clear YS-1-7006, USP

31.7
3.9
32.2
5.9


Ammonio methacrylate copolymer

85.4
10.5
55.55
10.3


dispersion, Type B, 30% dispersion







(Eudragit RS30D Solids), NF







Triethyl Citrate, USP/NF

13.0
1.6
8.61
1.6


Glyceryl monostearate emulsion

22.0
2.7
14.1
2.6


(Plasacryl T20), HS







Silicon dioxide, (Syloid 244FP), NF

4.1
0.5
4.52
0.8


Eudragit FS30D Liquid, HS

128.5
15.8
84.61
15.6


Total weight in capsule
~1100 mg
~813 mg
~100%
~543 mg
~100%
















TABLE 2







Dissolution results









% Methylphenidate HCl dissolved



















Time
Formu-
Formu-
Formu-
Formu-
Formu-
Formu-
Formu-
Formu-
Formu-
Formu-


Dissolution
(hours)
lation A
lation B
lation C
lation D
lation E
lation F
lation G
lation H
lation I
lation J





















USP paddle method,
1
4
4
1
1
25
18
25
24
20
20


100 rpm, at 37° C., 900 ml
4
11
10
3
4
31
22
29
28
21
21


simulated gastric fluid for 2
8
41
49
28
38
54
46
61
61
44
41


hours, 900 ml phosphate buffer
12 
68
76
44
40
64
58
71
71
83
73


pH 6.0 for 4 hours and 7th hour
161
79
78
54
41
68
67
74
73
78
79


onwards, 900 mL of phosphate













buffer pH 7.4













USP<711>Acceptance Table 2






1The total amount of Methylphenidate HCl decreases as it degrades at pH 7.4.














TABLE 3







Ratios, 90% Geometric CI for non-dose-normalized


Parameters Study 063-004









90% Geometric C.I.











Parameter
Treatment Comparisons
Ratio
Lower
Upper














AUC0-t
Formulation I
102.73%
97.69%
108.03%



(fed) vs IR-MPH (fed)






Formulation I
123.92%
117.87%
130.29%



(fast) vs IR-MPH (fast)






Formulation I
98.02%
93.21%
103.07%



(fed) vs (fast)





AUC0-inf
Formulation I
128.07%
122.18%
134.24%



(fed) vs IR-MPH (fed)






Formulation I
147.91%
141.31%
154.83%



(fast) vs IR-MPH (fast)






Formulation I
102.09%
97.41%
107.01%



(fed) vs (fast)





Cmax
Formulation I
71.17%
65.92%
76.83%



(fed) vs IR-MPH (fed)






Formulation I
87.06%
80.66%
93.96%



(fast) vs IR-MPH (fast)






Formulation I
89.31%
82.73%
96.41%



(fed) vs (fast)
















TABLE 5







Ratios, 90% Geometric CI for non-dose-normalized


Parameters Study 063-005









90% Geometric C.I.











Parameter
Treatment Comparisons
Ratio
Lower
Upper














AUC0-t
Test(A) - Reference(D)
98.78%
95.30%
102.40%



Test(B) - Reference(D)
98.60%
95.11%
102.22%



Test(C) - Reference(D)
101.08%
97.52%
104.78%


AUC0-inf
Test(A) - Reference(D)
97.87%
94.45%
101.41%



Test(B) - Reference(D)
98.45%
95.01%
102.02%



Test(C) - Reference(D)
101.35%
97.82%
105.01%


Cmax
Test(A) - Reference(D)
108.31%
100.05%
117.26%



Test(B) - Reference(D)
100.08%
92.43%
108.37%



Test(C) - Reference(D)
105.84%
97.77%
114.57%
















TABLE 4







Summary of non-dose-normalized Pharmacokinetic Results Study 063-004












Formulation I
Formulation I
Ritalin
Ritalin



(100 mg Fast)
(100 mg Fed)
(20 mg × 3 Fast)
(20 mg × 3 Fed)





AUC0-t
167783.86 ± 46487.66 
161271.48 ± 40500.38
132957.12 ± 43955.82
155290.78 ± 37540.22


(pg · hr/mL)






AUC0-inf
205610.43 ± 61472.88 
202964.28 ± 57449.88
136436.27 ± 45902.96
159381.72 ± 39469.43


(pg · hr/mL)






Residual Area (%)
17.62 ± 11.06
18.63 ± 8.51
 2.42 ± 0.96
 2.45 ± 0.92


Cmax (pg/mL)
12875.81 ± 4590.85 
11088.11 ± 2699.06
14105.39 ± 3770.36
15247.79 ± 3288.76


Tmax (hr)
11.5 
12.5 
9.50
6.04


Kel (1/hr)
0.1173 ± 0.0430
 0.1074 ± 0.0296
 0.1968 ± 0.0218
 0.1976 ± 0.0247


T1/2 el (hr)
6.95 ± 3.25
 7.03 ± 2.28
 3.57 ± 0.40
 3.56 ± 0.45


AUC0-4
24818.34 ± 7976.76 
21160.21 ± 6420.56
22955.21 ± 8292.78
26886.10 ± 7606.67


(pg · hr/mL)






AUC8-12
36457.19 ± 18489.84
29392.15 ± 8453.72
 41094.69 ± 12181.51
44914.86 ± 8753.20


(pg · hr/mL)






AUC12-16
39322.95 ± 10236.29
 36653.88 ± 11521.14
20364.91 ± 9558.52
24467.89 ± 8948.65


(pg · hr/mL)






AUC0-8
48626.93 ± 16709.44
47422.42 ± 9947.71
 58725.69 ± 17067.12
 70056.85 ± 16031.58


(pg · hr/mL)






AUC0-12
85084.12 ± 33395.48
 76814.57 ± 16722.84
 99820.38 ± 28132.26
114971.70 ± 23607.37


(pg · hr/mL)






AUC0-16
124407.07 ± 40902.66 
113468.45 ± 26835.93
120185.29 ± 37077.85
139439.59 ± 30402.02


(pg · hr/mL)






AUC0-24
167740.82 ± 46495.35 
161217.02 ± 40475.17
132949.12 ± 43953.43
155281.82 ± 37538.47


(pg · hr/mL)






AUC4-8
23500.00 ± 10293.79
25814.81 ± 6376.73
35446.96 ± 9454.91
 42739.42 ± 10194.06


(pg · hr/mL)






AUC12-t
82699.74 ± 21862.39
 84456.90 ± 26718.88
 33136.74 ± 16892.19
 40319.08 ± 17657.25


(pg · hr/mL)






Cmax0-4 (pg/mL)
9365.42 ± 3213.96
 9248.95 ± 1886.65
 9206.46 ± 3371.78
10951.60 ± 3222.66


Cmax4-8 (pg/mL)
7927.79 ± 4347.57
 8162.71 ± 1932.67
12684.06 ± 3583.22
14454.13 ± 3450.37


Cmax8-16
12413.97 ± 4546.66 
10667.64 ± 3017.29
13650.77 ± 3689.34
14174.43 ± 3158.31


(pg/mL)






Tmax0-4 (hr)
1.63
3.00
2.00
2.07


Tmax4-8 (hr)
4.00
3.95
6.00
6.00


Tmax8-16 (hr)
12.5 
13.5 
9.52
10.0 
















TABLE 6







Ratios, 90% Geometric CI for non-dose-normalized


Parameters Study 063-007









90% Geometric C.I.











Parameter
Treatment Comparisons
Ratio
Lower
Upper














AUC0-24
Formulation I
147.61%
143.02%
152.34%



vs IR-MPH





Cmax
Formulation I
98.92%
92.48%
105.81%



vs IR-MPH





Cmin
Formulation I
456.91%
404.67%
515.90%



vs IR-MPH
















TABLE 7







Ratios, 90% Geometric CI Parameters for Study 063-011









90% Geometric C.I.











Parameter
Treatment Comparisons
Ratio
Lower
Upper














AUC0-t
Formulation I
99.15%
91.82%
107.08%



vs Formulation J (fed)






Formulation I
99.64%
92.27%
107.61%



vs Formulation J (fast)





AUC0-inf
Formulation I
98.66%
91.67%
106.20%



vs Formulation J (fed)






Formulation I
98.76%
91.75%
106.29%



vs Formulation J (fast)





Cmax
Formulation I
98.34%
83.67%
115.60%



vs Formulation J (fed)






Formulation I
90.87%
77.31%
106.81%



vs Formulation J (fast)



















TABLE 8







Time (hours)
% Methylphenidate HCl dissolved









 1
NLT 15%



 4
18-38%



 8
35-55%



12
68-98



16
NLT 68

















TABLE 9







Production of Methylphenidate Immediate Release (IR) Beads








Process Parameters
Recommended Parameters












Coating sugar spheres with

Fluid bed


methylphenidate solution
Preheating
coating





Inlet air volume, cmh (to be
800 ± 300
1100 ± 200 


adjusted as necessary)




Inlet air temperature, ° C.
56 ± 5 
65 ± 10


Atomization air pressure, bar
  1 ± 0.2
3 ± 1


Inlet dew point, ° C.
8 ± 4
8 ± 4


Product temperature, ° C.
35 ± 2 
37.5 ± 3.5 


Spraying rate, g/min (to be
N/A
150-400


adjusted as necessary)













Rinsing with purified water
Rinsing
Drying
Cooling





Amount of water used (kg)
1
N/A
N/A


Fluidized air volume, cmh (to
1100 ± 200 
1000 ± 200 
1000 ± 300 


be adjusted as necessary)





Inlet air temperature, ° C. (to
65 ± 10
60 ± 6 
Minimum


be adjusted as necessary)





Atomization air pressure, bar
3 ± 1
  1 ± 0.2
  1 ± 0.2


Inlet dew point, ° C.
8 ± 4
8 ± 4
8 ± 4


Product temperature, ° C.
36 ± 3 
41 ± 4 
30 ± 1 


Spraying rate, g/min (to be
150-350
N/A
N/A


adjusted as necessary)





Time, min
N/A
~3
N/A
















TABLE 10







Production of Methylphenidate Controlled Release (CRIR) Beads








Process Parameters



Coating IR beads



with Controlled Release
Recommended Parameters










coating dispersion
Preheating
CR Coating
Rinsing





Amount of purified water
N/A
N/A
1


used (Kg)





Inlet air volume, cmh (to be
1000 ± 250
1250 ± 350 
1250 ± 250 


adjusted as necessary)





Inlet air temperature, ° C.
45 ± 5
45 ± 15
45 ± 15


Atomization air pressure, bar
  1 ± 0.2
2.8 ± 0.5
2.8 ± 0.5


Inlet dew point, ° C.
 8 ± 4
8 ± 4
8 ± 4


Product temperature, ° C.
25 ± 3
25 ± 5 
25 ± 5 


Spraying rate, g/min (to be
N/A
100-400
100-350


adjusted as necessary)
















TABLE 11







Production of Methylphenidate Distal (DRCRIR) Beads, cured


Process Parameters


Coating CRIR beads








with Distal Release
Recommended Parameters











coating dispersion
DR Coating
Rinsing
Curing
Cooling





Amount of purified water
N/A
1
N/A
N/A


used (Kg)






Inlet air volume, cmh (to be
1400 ± 400 
1400 ± 400 
800 ± 400
800 ± 100


adjusted as necessary)






Inlet air temperature, ° C.
45 ± 15
45 ± 15
To be
15 ± 10





adjusted



Atomization air pressure, bar
2.8 ± 0.5
2.8 ± 0.5

1 ± 0.2


1 ± 0.2



Inlet dew point, ° C.
8 ± 4
8 ± 4
8 ± 4
8 ± 4


Product temperature, ° C.
25 ± 3 
25 ± 3 
40 ± 2 
25 ± 3 


Spraying rate, g/min (to be
150-400
150-350
N/A
N/A


adjusted as necessary)






Time, min.
N/A
N/A
60
N/A
















TABLE 12







Production of Methylphenidate IR Distal (MPH IR distal) Beads








Process Parameters



Coating DRCRIR beads
Recommended Parameters












with Immediate Release layer
Preheating
IR coating
Rinsing
Drying
Cooling





Amount of purified water
N/A
N/A
1
N/A
N/A


used (Kg)







Inlet air volume, cmh (to be
1100 ± 400
1400 ± 400
1400 ± 400
1100 ± 400
1100 ± 400


adjusted as necessary)







Inlet air temperature, ° C.
 56 ± 15
 56 ± 15
 56 ± 15
To be
 20 ± 10






adjusted



Atomization air pressure, bar
  3 ± 0.5
  3 ± 0.5
  3 ± 0.5
  1 ± 0.2
  1 ± 0.2


Inlet dew point, ° C.
 8 ± 4
 8 ± 4
 8 ± 4
 8 ± 4
 8 ± 4


Product temperature, ° C.
36 ± 3
37.5 ± 3.5
37.5 ± 3.5
41 ± 4
30 ± 1


Spraying rate, g/min (to be
N/A
150-400
150-350
N/A
N/A


adjusted as necessary)







Time, min.
5-10
N/A
N/A
5
N/A
















TABLE 13







Production of Methylphenidate IR (MPH IR


distal) Beads coated with Sodium Alginate








Process Parameters
Recommended Parameters











Coating MPH IR distal beads

Sodium Alginate




with sodium alginate layer
Preheating
coating
Drying
Cooling





Amount of purified water
N/A
N/A
N/A
N/A


used (Kg)






Inlet air volume, cmh (to be
1100 ± 400
1400 ± 400
1100 ± 400
1100 ± 400


adjusted as necessary)






Inlet air temperature, ° C.
 70 ± 15
 70 ± 15
To be
 20 ± 10





adjusted



Atomization air pressure, bar
  3 ± 0.5
  3 ± 0.5
  1 ± 0.2
  1 ± 0.2


Inlet dew point, ° C.
 8 ± 4
 8 ± 4
 8 ± 4
 8 ± 4


Product temperature, ° C.
52 ± 5
52 ± 5
41 ± 4
30 ± 1


Spraying rate, g/min (to be
N/A
50-300
N/A
N/A


adjusted as necessary)






Time, min.
5-10
N/A
5
N/A
















TABLE 14







Dissolution Parameters (Example 10)








Equipment
Conditions





Apparatus
apparatus 1


Basket, USP



Speed
100 rpm


Bath
temperature: 37° C.


Dissolution
0%, 5%, 20%, 35% or 40% ethanol v/v in 0.1N HCl


media 900 ml



Sampling
15, 30, 45, 60, 75, 90, 105, 120 minutes


time points
















TABLE 15







Composition of the dosage form (Formulation I 80:20) MPH IR distal:









Quantity per capsule (mg) per Strength (label claim) approx:
















Ingredient
25 mg
30 mg
35 mg
45 mg
55 mg
70 mg
85 mg
100 mg
%



















Methylphenidate HCl, USP
25
30
35
45
55
70
85
100
12.3


Sugar spheres 14/18 mesh, USP/NF
107.1
128.5
150.0
192.8
235.7
299.9
364.2
428.5
52.7


Opadry Clear YS-1-7006
7.9
9.5
11.1
14.3
17.4
22.2
27.0
31.7
3.9


Ammonio methacrylate copolymer
21.3
25.6
29.9
38.4
47.0
59.8
72.6
85.4
10.5


dispersion, Type B, 30% dispersion











(Eudragit RS30D Solids), NF











Triethyl Citrate, USP/NF
3.3
3.9
4.6
5.9
7.2
9.1
11.1
13.0
1.6


Glyceryl monostearate emulsion
5.5
6.6
7.7
9.9
12.1
15.4
18.7
22.0
2.7


(Plasacryl T20), HS, solids











Silicon dioxide, (Syloid 244FP), NF
1.0
1.2
1.4
1.8
2.2
2.8
3.5
4.1
0.5


Eudragit FS30D, HS, solids
32.1
38.5
45.0
57.8
70.7
89.9
109.2
128.5
15.8


TOTAL (approx.)
203
244
285
366
447
569
691
813
100
















TABLE 16







Composition of the dosage form (Formulation J 80:20) MPH IR distal:









Quantity per capsule (mg) per Strength (label claim) approx:
















Ingredient
25 mg
30 mg
35 mg
45 mg
55 mg
70 mg
85 mg
100 mg
%



















Methylphenidate HCl, USP
25
30
35
45
55
70
85
100
18.4


Sugar spheres 14/18 mesh, USP/NF
60.8
72.9
84.9
109.5
133.7
169.9
206.5
243.1
44.7


Opadry Clear YS-1-7006
8.0
9.6
11.2
14.5
17.6
22.4
27.3
32.2
5.9


Ammonio methacrylate copolymer
14.0
16.8
19.6
25.2
30.8
39.1
47.6
55.55
10.3


dispersion, Type B, 30% dispersion











(Eudragit RS30D Solids), NF











Triethyl Citrate, USP/NF
2.2
2.6
3.0
3.9
4.8
6.1
7.4
8.61
1.6


Glyceryl monostearate emulsion
3.5
4.3
4.9
6.4
7.8
9.9
12.0
14.1
2.6


(Plasacryl T20), HS, solids











Silicon dioxide, (Syloid 244FP), NF
1.1
1.3
1.5
2.0
2.4
3.0
3.7
4.52
0.8


Eudragit FS30D, HS, solids
21.2
25.4
29.6
38.2
46.6
59.3
72.1
84.61
15.6


TOTAL (approx.)
136
163
190
245
299
380
462
543
100








Claims
  • 1. A method of treating attention deficit hyperactivity disorder (ADHD) in a subject in need thereof, the method comprising orally administering to the subject a dosage form providing 25, 35, 45, 55, 70, 85, or 100 mg of methylphenidate hydrochloride, wherein the dosage form provides: a first in vivo methylphenidate Tmax and a second in vivo methylphenidate Tmax, wherein in a fed state, the second in vivo methylphenidate Tmax is a Tmax8-16 of about 13.5 hours; and efficacious treatment of ADHD for about 16 hours after a single oral administration, further wherein the dosage form has an in vitro methylphenidate hydrochloride dissolution rate that does not increase in the presence of about either 5% or 20% ethanol when measured under in vitro conditions with stirring at 100 rpm at pH 1.2 for 2 hours in 900 mL of a medium comprising up to about either 5% or 20% v/v ethanol.
  • 2. The method of claim 1, wherein not more than 20% of the methylphenidate hydrochloride in the dosage form is released after two hours when measured under in vitro conditions with stirring at 100 rpm at pH 1.2 for 2 hours in 900 mL of a medium comprising up to about 20% v/v ethanol.
  • 3. The method of claim 1, wherein not more than 20% of the methylphenidate hydrochloride in the dosage form is released after two hours when measured under in vitro conditions with stirring at 100 rpm at pH 1.2 for 2 hours in 900 mL of a medium comprising up to about 5% v/v ethanol.
  • 4. The method of claim 1, wherein the dosage form comprises an immediate release methylphenidate hydrochloride component and a controlled/delayed release methylphenidate hydrochloride component, wherein the immediate release methylphenidate hydrochloride component and the controlled/delayed release methylphenidate hydrochloride component, together, provide 25, 35, 45, 55, 70, 85, or 100 mg of methylphenidate hydrochloride.
  • 5. The method of claim 4, wherein the dosage form has the following in vitro methylphenidate dissolution profile:
  • 6. The method of claim 4, wherein the controlled/delayed release methylphenidate hydrochloride component and the immediate release methylphenidate hydrochloride component are provided via a plurality of coated beads.
  • 7. The method of claim 6, wherein each bead comprises a portion of the controlled/delayed release methylphenidate hydrochloride component and a portion of the immediate release methylphenidate hydrochloride component.
  • 8. The method of claim 7, wherein each bead in the plurality of beads comprises a core comprising a first amount of methylphenidate hydrochloride corresponding to the portion of the controlled/delayed release methylphenidate hydrochloride component.
  • 9. The method of claim 8, wherein the core comprises a granule coated with the first amount of methylphenidate hydrochloride or wherein the core comprises a granule substrate in admixture with the first amount of methylphenidate hydrochloride.
  • 10. The method of claim 9, wherein the first amount of methylphenidate hydrochloride is coated with an inner controlled release coating and an outer delayed release coating coated over the inner controlled release coating.
  • 11. The method of claim 10, wherein a second amount of methylphenidate hydrochloride, corresponding to the portion of the immediate release methylphenidate hydrochloride component, is coated over the outer delayed release coating.
  • 12. The method of claim 11, wherein the controlled/delayed release methylphenidate hydrochloride component comprises about 78 weight percent to about 82 weight percent of the 25, 35, 45, 55, 70, 85, or 100 mg of methylphenidate hydrochloride in the pharmaceutical composition.
  • 13. The method of claim 12, wherein the inner controlled release coating is present in an amount of about 3 percent to about 16 percent by weight of each bead in the plurality of coated beads, and wherein the outer delayed release coating is present in an amount of from about 3 percent to about 20 percent by weight of each bead in the plurality of coated beads.
  • 14. The method of claim 13, wherein the inner controlled release coating comprises ammonio methacrylate copolymer, Type B USP/NF.
  • 15. The method of claim 14, wherein the outer delayed release coating comprises poly(methyl acrylate-co-methyl methacrylate-co-methacrylic acid) 7:3:1.
Priority Claims (1)
Number Date Country Kind
CA 2902911 Aug 2015 CA national
US Referenced Citations (363)
Number Name Date Kind
2377237 James May 1945 A
2507631 Hartmann et al. May 1950 A
2676169 Baldoni Apr 1954 A
2772488 Meltzer Dec 1956 A
2791509 Cosler May 1957 A
3365365 Butler et al. Jan 1968 A
3370054 Loew Feb 1968 A
3371015 Sjogren et al. Feb 1968 A
3424842 Eberhard Jan 1969 A
3623997 Thomas Nov 1971 A
3629393 Atsushi et al. Dec 1971 A
3845770 Theeuwes et al. Nov 1974 A
3870790 Lowey et al. Mar 1975 A
3883647 Geller May 1975 A
3901968 Cohen et al. Aug 1975 A
3916899 Theeuwes et al. Nov 1975 A
3927205 Ohno et al. Dec 1975 A
3935326 Groppenbacher et al. Jan 1976 A
3983233 Brattsand et al. Sep 1976 A
3996356 Grunberg Dec 1976 A
4000254 Gordon et al. Dec 1976 A
4060598 Groppenbacher et al. Nov 1977 A
4083949 Benedikt Apr 1978 A
4088798 Michaelis May 1978 A
4093709 Choi et al. Jun 1978 A
4140755 Sheth et al. Feb 1979 A
4167558 Sheth et al. Sep 1979 A
4173626 Dempski et al. Nov 1979 A
4182756 Guzek et al. Jan 1980 A
4226849 Schor Oct 1980 A
4234565 Flodin et al. Nov 1980 A
4252786 Weiss et al. Feb 1981 A
4256108 Theeuwes Mar 1981 A
4259314 Lowey Mar 1981 A
4318400 Peery et al. Mar 1982 A
4341759 Bogentoft et al. Jul 1982 A
4357469 Schor Nov 1982 A
4369172 Schor et al. Jan 1983 A
4389393 Schor et al. Jun 1983 A
4439453 Vogel Mar 1984 A
4443497 Samejima et al. Apr 1984 A
4454108 Iida et al. Jun 1984 A
4464170 Clemens et al. Aug 1984 A
4503031 Glassman Mar 1985 A
4520172 Lehmann et al. May 1985 A
4526777 Blume et al. Jul 1985 A
4540566 Davis et al. Sep 1985 A
4548990 Mueller et al. Oct 1985 A
4574080 Roswall et al. Mar 1986 A
4592753 Panoz Jun 1986 A
4600645 Ghebre-Sellassie et al. Jul 1986 A
4606909 Bechgaard et al. Aug 1986 A
4606940 Frank et al. Aug 1986 A
4609542 Panoz et al. Sep 1986 A
4610870 Jain et al. Sep 1986 A
4611008 Heinzelmann Sep 1986 A
4634587 Hsiao Jan 1987 A
4666703 Kopf May 1987 A
4693895 Wong et al. Sep 1987 A
4695467 Uemura et al. Sep 1987 A
4705695 Lehmann et al. Nov 1987 A
4708867 Hsiao Nov 1987 A
4708874 De et al. Nov 1987 A
4710519 Finnan et al. Dec 1987 A
4716040 Panoz Dec 1987 A
4716041 Kjornaes et al. Dec 1987 A
4721619 Panoz et al. Jan 1988 A
4724148 Sonobe et al. Feb 1988 A
4728513 Ventouras Mar 1988 A
4729190 Lee Mar 1988 A
4740365 Yukimatsu et al. Apr 1988 A
4748023 Tamas et al. May 1988 A
4765988 Sonobe et al. Aug 1988 A
4766012 Valenti Aug 1988 A
4770809 Heidenrich et al. Sep 1988 A
4772475 Fukui et al. Sep 1988 A
4783337 Wong et al. Nov 1988 A
4786503 Edgren et al. Nov 1988 A
4786505 Lovgren et al. Nov 1988 A
4786506 Fontanelli Nov 1988 A
4794001 Mehta et al. Dec 1988 A
4798724 Khanna Jan 1989 A
4800084 Zerbe Jan 1989 A
4811845 Baggett Mar 1989 A
4814178 Bolton et al. Mar 1989 A
4816264 Phillips et al. Mar 1989 A
4820522 Radebaugh et al. Apr 1989 A
4826688 Panoz et al. May 1989 A
4828840 Sakamoto et al. May 1989 A
4837004 Wu et al. Jun 1989 A
4839177 Colombo et al. Jun 1989 A
4842867 Ayer et al. Jun 1989 A
4853230 Lovgren et al. Aug 1989 A
4861599 Springolo et al. Aug 1989 A
4863744 Urquhart et al. Sep 1989 A
4867984 Patel Sep 1989 A
4867987 Seth Sep 1989 A
4880830 Rhodes Nov 1989 A
4889238 Batchelor Dec 1989 A
4891230 Geoghegan et al. Jan 1990 A
4892739 Shah et al. Jan 1990 A
4892742 Shah Jan 1990 A
4894240 Geoghegan et al. Jan 1990 A
4900557 Dell et al. Feb 1990 A
4917899 Geoghegan et al. Apr 1990 A
4931295 Courtright et al. Jun 1990 A
4940586 Cheng et al. Jul 1990 A
4946685 Edgren et al. Aug 1990 A
4950486 Ayer et al. Aug 1990 A
4952402 Sparks et al. Aug 1990 A
4954350 Jones et al. Sep 1990 A
4966769 Guittard et al. Oct 1990 A
4966770 Giannini et al. Oct 1990 A
4968509 Radebaugh et al. Nov 1990 A
4971805 Kitaneshi et al. Nov 1990 A
4981468 Benefiel et al. Jan 1991 A
4981693 Higashi et al. Jan 1991 A
4983401 Eichel et al. Jan 1991 A
4983403 Ardaillon et al. Jan 1991 A
4984592 Hellein Jan 1991 A
4994279 Aoki et al. Feb 1991 A
5004613 Radebaugh et al. Apr 1991 A
5008118 Iwanami et al. Apr 1991 A
5011694 Nuernberg et al. Apr 1991 A
5015479 Mulligan et al. May 1991 A
5019397 Wong et al. May 1991 A
5024842 Edgren et al. Jun 1991 A
5026560 Makino et al. Jun 1991 A
5026709 Harwood et al. Jun 1991 A
5032406 Dansereau et al. Jul 1991 A
5047007 Mcnichols et al. Sep 1991 A
5047258 Belanger et al. Sep 1991 A
5051262 Panoz et al. Sep 1991 A
5055306 Barry et al. Oct 1991 A
5068110 Fawzi et al. Nov 1991 A
5073380 Babu et al. Dec 1991 A
5084278 Mehta et al. Jan 1992 A
5085865 Nayak Feb 1992 A
5085866 Cowsar et al. Feb 1992 A
5091175 Imondi et al. Feb 1992 A
5093200 Watanabe et al. Mar 1992 A
5095054 Lay et al. Mar 1992 A
5096717 Wirth et al. Mar 1992 A
5112384 Miura et al. May 1992 A
5112621 Stevens et al. May 1992 A
5128142 Mulligan et al. Jul 1992 A
5130140 Urban et al. Jul 1992 A
5130171 Prudhomme et al. Jul 1992 A
5133974 Paradissis et al. Jul 1992 A
5158777 Abramowitz et al. Oct 1992 A
5160737 Friedman et al. Nov 1992 A
5160742 Mazer et al. Nov 1992 A
5167964 Muhammed et al. Dec 1992 A
5171580 Iamartino et al. Dec 1992 A
5178866 Wright et al. Jan 1993 A
5186943 Okada et al. Feb 1993 A
5198228 Urban et al. Mar 1993 A
5200193 Radebaugh et al. Apr 1993 A
5202128 Morella et al. Apr 1993 A
5202159 Chen et al. Apr 1993 A
5219621 Geoghegan et al. Jun 1993 A
5229131 Amidon et al. Jul 1993 A
5229134 Mention et al. Jul 1993 A
5233987 Fabian et al. Aug 1993 A
5252341 Sauerbier et al. Oct 1993 A
5262169 Sauerbier et al. Nov 1993 A
5266331 Oshlack et al. Nov 1993 A
5273760 Oshlack et al. Dec 1993 A
5275824 Carli et al. Jan 1994 A
5275825 Okada et al. Jan 1994 A
5286493 Oshlack et al. Feb 1994 A
5288505 Deboeck et al. Feb 1994 A
5310558 Pozzi et al. May 1994 A
5316774 Eury et al. May 1994 A
5320853 Noda et al. Jun 1994 A
5324717 Berglindh et al. Jun 1994 A
5324718 Loftsson Jun 1994 A
5326570 Rudnic et al. Jul 1994 A
5330766 Morella et al. Jul 1994 A
5334372 Kawamata et al. Aug 1994 A
5342627 Chopra et al. Aug 1994 A
5356467 Oshlack et al. Oct 1994 A
5358718 Sauerbier et al. Oct 1994 A
5368861 Ushimaru et al. Nov 1994 A
5370878 Shah Dec 1994 A
5374430 Newton Dec 1994 A
5374759 Imperante et al. Dec 1994 A
5378474 Morella et al. Jan 1995 A
5384130 Kamada Jan 1995 A
5395626 Kotwal et al. Mar 1995 A
5407687 Coffin et al. Apr 1995 A
5439689 Hendrickson et al. Aug 1995 A
5445829 Paradissis et al. Aug 1995 A
5451409 Rencher et al. Sep 1995 A
5472708 Chen Dec 1995 A
5472711 Baichwal Dec 1995 A
5474786 Kotwal et al. Dec 1995 A
5478573 Eichel et al. Dec 1995 A
5489436 Hoy Feb 1996 A
5494678 Paradissis et al. Feb 1996 A
5500227 Oshlack et al. Mar 1996 A
5521208 York May 1996 A
5547878 Kell Aug 1996 A
5576022 Yang et al. Nov 1996 A
5580578 Oshlack et al. Dec 1996 A
5597072 Lieberman et al. Jan 1997 A
5639476 Oshlack et al. Jun 1997 A
5643602 Ulmius Jul 1997 A
5652146 Kell Jul 1997 A
5658590 Heiligenstein et al. Aug 1997 A
5661123 Stalker et al. Aug 1997 A
5672360 Sackler et al. Sep 1997 A
5711967 Juch Jan 1998 A
5763396 Weiner et al. Jun 1998 A
5773031 Shah et al. Jun 1998 A
5773478 Richards et al. Jun 1998 A
5785994 Wong et al. Jul 1998 A
5837284 Mehta et al. Nov 1998 A
5840329 Bai Nov 1998 A
5874090 Baker et al. Feb 1999 A
5885616 Hsiao et al. Mar 1999 A
5945123 Hermelin Aug 1999 A
6093420 Baichwal Jul 2000 A
6143322 Sackler et al. Nov 2000 A
6214379 Hermelin Apr 2001 B1
6228398 Devane et al. May 2001 B1
6255325 Dariani et al. Jul 2001 B1
6322819 Burnside et al. Nov 2001 B1
6372254 Ting et al. Apr 2002 B1
6419960 Krishnamurthy et al. Jul 2002 B1
6602887 Dariani et al. Aug 2003 B2
6605300 Burnside et al. Aug 2003 B1
6627223 Percel et al. Sep 2003 B2
6632454 Beckert et al. Oct 2003 B2
6673367 Goldenheim et al. Jan 2004 B1
6730325 Devane et al. May 2004 B2
6761904 Bertelsen et al. Jul 2004 B2
6793936 Devane et al. Sep 2004 B2
6878387 Petereit et al. Apr 2005 B1
6897205 Beckert et al. May 2005 B2
6902742 Devane et al. Jun 2005 B2
7048945 Percel et al. May 2006 B2
7070803 Skinhoj et al. Jul 2006 B2
7083808 Goldenheim et al. Aug 2006 B2
7195778 Fleshner-Barak et al. Mar 2007 B2
7247318 Krishnamurthy et al. Jul 2007 B2
7438929 Beckert et al. Oct 2008 B2
7438930 Krishnamurthy et al. Oct 2008 B2
RE41148 Burnside et al. Feb 2010 E
RE42096 Burnside et al. Feb 2011 E
8124653 Matalon et al. Feb 2012 B2
8133507 Yum et al. Mar 2012 B2
8147870 Yum et al. Apr 2012 B2
8153152 Yum et al. Apr 2012 B2
8168217 Yum et al. May 2012 B2
8354124 Yum et al. Jan 2013 B2
8420120 Yum et al. Apr 2013 B2
8580310 Krishnamurthy et al. Nov 2013 B2
8846100 Shojael et al. Sep 2014 B2
8911777 Coulter Dec 2014 B2
8945614 Yum et al. Feb 2015 B2
8951556 Yum et al. Feb 2015 B2
8974821 Yum et al. Mar 2015 B2
9066869 Krishnamurthy et al. Jun 2015 B2
9107804 Rubino et al. Aug 2015 B2
9161918 Venkatesh Oct 2015 B2
9161919 Venkatesh Oct 2015 B2
9233160 Yum et al. Jan 2016 B2
9364430 Babul Jun 2016 B2
9517271 Yum et al. Dec 2016 B2
9522119 Odidi Dec 2016 B2
9566249 Venkatesh Feb 2017 B2
9579293 Venkatesh Feb 2017 B2
9700515 Odidi Jul 2017 B2
9700516 Odidi Jul 2017 B2
9801823 Krishnamurthy et al. Oct 2017 B2
9801939 Odidi Oct 2017 B2
9884022 Deshmukh et al. Feb 2018 B2
10722473 Donnelly Jul 2020 B2
20010012847 Lam et al. Aug 2001 A1
20010055613 Burnside et al. Dec 2001 A1
20020034544 Skinhoj et al. Mar 2002 A1
20020054907 Devane et al. May 2002 A1
20020147232 Sundgreen et al. Oct 2002 A1
20020192282 Beckert et al. Dec 2002 A1
20020193445 Bertelsen et al. Dec 2002 A1
20030054033 Krishnamurthy et al. Mar 2003 A1
20030124188 Burnside et al. Jul 2003 A1
20030129229 Krsek Jul 2003 A1
20030129237 Devane et al. Jul 2003 A1
20030153607 Glinecke et al. Aug 2003 A1
20030170304 Devane et al. Sep 2003 A1
20040091531 Glinecke et al. May 2004 A1
20040091532 Mehta May 2004 A1
20040131680 Goldenheim et al. Jul 2004 A1
20040161382 Yum et al. Aug 2004 A1
20040197405 Devane et al. Oct 2004 A1
20040219213 Burnside et al. Nov 2004 A1
20040228802 Chang et al. Nov 2004 A1
20050089571 Beckert et al. Apr 2005 A1
20050136111 Glinecke et al. Jun 2005 A1
20060046008 Wildfang Mar 2006 A1
20060159757 Payne Jul 2006 A1
20060204576 Petereit et al. Sep 2006 A1
20060240105 Devane et al. Oct 2006 A1
20070104789 Spector May 2007 A1
20070212414 Baichwal Sep 2007 A1
20070264323 Shojaei et al. Nov 2007 A1
20070264325 Krishnamurthy et al. Nov 2007 A1
20070281023 Glinecke et al. Dec 2007 A1
20080069872 Rubio et al. Mar 2008 A1
20080118556 Devane et al. May 2008 A1
20080193522 Meier et al. Aug 2008 A1
20080206321 Yum et al. Aug 2008 A1
20080206324 Gryczke et al. Aug 2008 A1
20090023689 Yum et al. Jan 2009 A1
20090023690 Yum et al. Jan 2009 A1
20090123554 Krishnamurthy et al. May 2009 A1
20090280176 Vieira et al. Nov 2009 A1
20090297597 Liversidge et al. Dec 2009 A1
20090297602 Devane et al. Dec 2009 A1
20090324716 Shen et al. Dec 2009 A1
20100136106 Liversidge et al. Jun 2010 A1
20100151010 Petereit et al. Jun 2010 A1
20100151020 Rosenberger et al. Jun 2010 A1
20100226978 Petereit et al. Sep 2010 A1
20100260844 Scicinski Oct 2010 A1
20100297221 Coulter Nov 2010 A1
20100316712 Nangia et al. Dec 2010 A1
20110064803 Devane et al. Mar 2011 A1
20110212175 Kim et al. Sep 2011 A1
20110223244 Liversidge Sep 2011 A1
20110262496 Desai Oct 2011 A1
20110268799 Manesh Nov 2011 A1
20120015007 Bredenberg et al. Jan 2012 A1
20120065221 Babul Mar 2012 A1
20120128771 Venkatesh May 2012 A1
20120135072 Yum et al. May 2012 A1
20120135073 Yum et al. May 2012 A1
20120178771 Babul et al. Jul 2012 A1
20120276017 Lickrish Nov 2012 A1
20130022654 Deshmukh et al. Jan 2013 A1
20130236554 Tengler Sep 2013 A1
20130287845 Yum et al. Oct 2013 A1
20130295168 Yum et al. Nov 2013 A1
20140099361 Krishnamurthy et al. Apr 2014 A1
20140120185 Hirose et al. May 2014 A1
20140200237 Babul Jul 2014 A1
20140212483 Lickrish et al. Jul 2014 A1
20140255594 Rubino et al. Sep 2014 A1
20150196644 Yum et al. Jul 2015 A1
20150313850 Krishnamurthy et al. Nov 2015 A1
20150366850 Jain Dec 2015 A1
20160193345 Yum et al. Jul 2016 A1
20160215069 Stern et al. Jul 2016 A1
20160220491 Rubino et al. Aug 2016 A1
20170000783 Devane et al. Jan 2017 A1
20170071926 Krishnamurthy et al. Mar 2017 A1
20170079921 Krishnamurthy et al. Mar 2017 A1
20170112774 Venkatesh Apr 2017 A1
20170135999 Krishnamurthy et al. May 2017 A1
20170165255 Yum et al. Jun 2017 A1
20170258912 Odidi Sep 2017 A1
Foreign Referenced Citations (90)
Number Date Country
1632692 Nov 1992 AU
653223 Sep 1994 AU
1183665 Mar 1985 CA
1297368 Mar 1992 CA
2348090 Apr 2000 CA
2355644 Jun 2000 CA
2368367 Oct 2000 CA
2373909 Sep 2001 CA
2426883 May 2002 CA
2502371 May 2004 CA
2507631 Jun 2004 CA
2537103 May 2005 CA
2566497 Dec 2005 CA
2683409 Oct 2008 CA
2754604 Sep 2010 CA
2830788 Sep 2012 CA
2877190 Jan 2014 CA
2926082 Apr 2015 CA
2933587 Jun 2015 CA
0054010 Jun 1982 EP
0040590 Jan 1983 EP
0074584 Mar 1983 EP
0122077 Oct 1984 EP
0232690 Aug 1987 EP
0239361 Sep 1987 EP
0248548 Dec 1987 EP
0278174 Aug 1988 EP
0325086 Jul 1989 EP
0325843 Aug 1989 EP
0327295 Aug 1989 EP
0377517 Oct 1990 EP
0250374 May 1991 EP
0463877 Jan 1992 EP
0514814 Nov 1992 EP
0631781 Jan 1995 EP
377518 Jul 1932 GB
1182124 Feb 1970 GB
1245467 Sep 1971 GB
2098867 Dec 1982 GB
2141342 Dec 1984 GB
2159715 Dec 1985 GB
2178313 Feb 1987 GB
2209280 May 1989 GB
2253348 Sep 1992 GB
S55149211 Nov 1980 JP
H01165518 Jun 1989 JP
H0291028 Mar 1990 JP
2000516610 Dec 2000 JP
2016006608 Jan 2016 JP
WO-198300435 Feb 1983 WO
WO-198500481 Jan 1985 WO
WO-198603676 Jul 1986 WO
WO-198908448 Sep 1989 WO
WO-199221333 Jan 1993 WO
WO-199317673 Sep 1993 WO
WO-199703671 Feb 1997 WO
WO-199703672 Feb 1997 WO
WO-199703673 Feb 1997 WO
WO-199806380 Feb 1998 WO
WO-199814168 Apr 1998 WO
WO-199818454 May 1998 WO
WO-199823263 Jun 1998 WO
WO-199848782 Nov 1998 WO
WO-199908662 Feb 1999 WO
WO-199962496 Dec 1999 WO
WO-200023055 Apr 2000 WO
WO-200025752 May 2000 WO
WO-200028990 May 2000 WO
WO-200035426 Jun 2000 WO
WO-200035450 Jun 2000 WO
WO-200168058 Sep 2001 WO
WO-200174334 Oct 2001 WO
WO-200174335 Oct 2001 WO
WO-200188092 Nov 2001 WO
WO-200189473 Nov 2001 WO
WO-200235426 May 2002 WO
WO-2003000032 Jan 2003 WO
WO-2003080032 Oct 2003 WO
WO-2004054542 Jul 2004 WO
WO-2006078811 Jul 2006 WO
WO-2006132752 Dec 2006 WO
WO-2007011473 Jan 2007 WO
WO-2007037790 Apr 2007 WO
WO-2007070082 Jun 2007 WO
WO-2007093642 Aug 2007 WO
WO-2008079102 Jul 2008 WO
WO-2008083442 Jul 2008 WO
WO-2012080834 Jun 2012 WO
WO-2014174387 Oct 2014 WO
WO-2015188092 Dec 2015 WO
Non-Patent Literature Citations (112)
Entry
Andersson, P., et al., “In Vitro Biotransformation of Glucocorticoids in Liver and Skin Homogenate Fraction from Man, Rat and Hairless Mouse,” Steroid Biochemistry 16(6):787-795, Pergamon Press, England (Jun. 1982).
Aoyama, T., et al., “Pharmacokinetics and Pharmacodynamics of (+)-Threo-Methylphenidate Enantiomer In Patients With Hypersomnia,” Clinical Pharmacology and Therapeutics 55(3):270-276, Wiley, United States (Mar. 1994).
Aqua coat (Ethylcellulose ), pp. 17-36, 1985, Manufacturer's Info.
Aqua coat, (Ethylcellulose), 1987, Manufacturer's Info.
Ariens, E.J, “Racemic Therapeutics—Ethical and Regulatory Aspects,” European Journal of Clinical Pharmacology 41(2):89-93, Springer, Germany (1991).
Ariens, E.J, “Stereoselectivity in Pharmacodynamics and Pharmacokinetics,” Swiss Medical Weekly 120(5): 131-134, EMH Swiss Medical Publishers, Switzerland (Feb. 1990).
Biederman, J., et al., “A Double-Blind Placebo Controlled Study of Desipramine in the Treatment of ADD: I. Efficacy,” American Academy of Child and Adolescent Psychiatry 28(5):777-784, Elsevier, United States (Sep. 1989).
Biederman, J., et al., “Comorbidity of Attention Deficit Hyperactivity Disorder with Conduct, Depressive, Anxiety, and Other Disorders,” American Journal of Psychiatry 148(5):564-577, American Psychiatric Association, United States (May 1991).
Biederman, J., et al., “Patterns of Psychiatric Comorbidity, Cognition, and Psychosocial Functioning In Adults with Attention Deficit Hyperactivity Disorder,” American Journal of Psychiatry 150(12):1792-1798, American Psychiatric Association, United States (Dec. 1993).
Birmaher, B., et al., “Sustained Release Methylphenidate: Pharmacokinetic Studies in ADDH Males,” American Academy of Child and Adolescent Psychiatry 28(5):768-772, Elsevier, United States (Sep. 1989).
Castle, W.E., et al., “Linkage Studies of the Rat (Rattus Norvegicus),” Proceedings of the National Academy of Sciences 27(8):394-398, National Academy of Sciences, United States (Aug. 1941).
Catapres (Cionidine). Physicians' Desk Reference, 48th Edition. ed. Montvale, NJ: Thomson Healthcare, 1994, pp. 612-614.
Child, A.H., et al., “Joint Hypermobility Syndrome: Inherited Disorder of Collagen Synthesis,” Rheumatology 13(2):239-243, Journal Of Rheumatology Publishing Co, Canada (1986).
Chouinard, G., et al., “An Early Phase II Clinical Trial of Tomoxetine (LY139603) in the Treatment of Newly Admitted Depressed Patients,” Psychopharmacology (Berl) 83(1):126-128, Springer-Verlag, Germany (1984).
Chouinard, G., et al., “An Early Phase II Clinical Trial with Followup of Tomoxetine (LY139603) in the Treatment of Newly Admitted Depressed Patients,” Psychopharmacology Bulletin 21(1):73-76, MedWorks Media, United States (1985).
Chumpradit, S., et al., “Iodinated Tomoxetine Derivatives as Selective Ligands for Serotonin and Norepinephrine Uptake Sites,” Medicinal Chemistry 35 (23):4492-4497 , American Chemical Society, United States (Nov. 1992).
Corrigan, O.I., et al., “The Biophannaceutic Drug Classification and Drugs Administered in Extended Release (ER) Formulations,” Advances in Experimental Medicine and Biology 423: 111-128, Kluwer Academic/Plenum Publishers, United States (1997).
Covera—HS, Phvsicians' Desk Reference. 51st Edition. Montvale. NJ: Thomson Healthcare, 1997, pp. 2573-2576.
Cusack, B., et al., “Binding of Antidepressants to Human Brain Receptors: Focus on Newer Generation Compounds,” Psychopharmacology 114(4):559-565, Springer-Verlag, Germany (May 1994).
Danielsson, A., et al., “A Controlled Randomized Trial of Budesonide Versus Prednisolone Retention Enemas In Active Distal Ulcerative Colitis,” Scandinavian Journal of Gastroenterology 22(8):987-992, Informa Healthcare, England (Oct. 1987).
De Haan, P., et al., “Oral Controlled Release Dosage Forms. A Review,” Pharmaceutisch Weekblad 6(2):57-67, Bohn, Scheltema En Holkema, Netherlands (Apr. 1984).
De Scalzi, M., et al., “Circadian Rhythms of Arterial Blood Pressure,” Chronobiologia 13(3):239-244, Associated Chronobiologia Researchers, Italy (Jul.-Sep. 1986).
Depakote Tablets, Physicians' Desk Reference.51st Edition, Montvale, NJ: Thomson Healthcare, 1997, pp. 418-422.
Drimmer, E.J., et al., “Desipramine and Methylphenidate Combination Treatment for Depression: Case Report,” American Journal of Psychiatry 140(2):241-242, American Psychiatric Association, United States (Feb. 1983).
Eckerman, D.A., et al., “Enantioselective Behavioral Effects of Threo-Methylphenidate In Rats,” Pharmacology Biochemistry and Behavior 40(4):875-880, Elsevier, United States (Dec. 1991).
Elavil (Amitriptyline), Physicians' Desk Reference, 47th Edition, Montvale, NJ: Thomson Healthcare, 1993, pp. 2372-2374.
Erramouspe, J., et al., “Effect on Dissolution from Halving Methylphenidate Extended-Release Tablets,” Annals of Pharmacotherapy 31(10):1123-1126, Sage, United States (Oct. 1997).
Farid, N.A., et al., “Single-Dose and Steady-State Pharmacokinetics of Tomoxetine in Normal Subjects,” Clinical Pharmacology 25(4):296-301, Wiley, England (May-Jun. 1985).
Fuller, R.W., et al., “Antagonism By Tomoxetine of the Depletion of Norepinephrine and Epinephrine In Rat Brain By Alpha-Methyl-M-Tyrosine,” Research Communications in Chemical Pathology and Pharmacology 41(1):169-172, P. J. D. Publications, United States (Jul. 1983).
Fuller, R.W., et al., “Effects of Duloxetine, an Antidepressant Drug Candidate, On Concentrations of Monoamines and Their Metabolites in Rats and Mice,” Journal of Pharmacology and Experimental Therapeutics 269(1):132-136, American Society for Pharmacology and Experimental Therapeutics, United States (Apr. 1994).
Fuller, R.W., et al., “Serotonin Reuptake Blockers in Vitro and In Vivo,” Clinical Psychopharmacology 7(6 Suppl):36S-43S, Williams And Wilkins, United States (Dec. 1987).
Gamstedt, A., et al., “Effect of Betamethasone Treatment on Iodothyronines and Thyroid Hormone-Binding Proteins During controlled nutrition. A study on patients with chronic inflammatory bowel disease,” Acta Endocrinol (Copenh) 103(2):188-191, Periodica, Denmark (Jun. 1983).
Gehlert, D.R., et al., “Localization of Rat Brain Binding Sites for [3H]Tomoxetine, An Enantiomerically Pure Ligand for Norepinephrine Reuptake Sites,” Neuroscience Letters 157(2):203-206, Elsevier Scientific Publishers Ireland, Ireland (Jul. 1993).
Gilman, V., et al., “The Top Pharmaceuticals that Changed the World: Ritalin,” Chemical & Engineering News 83:25, (2005).
Green, W.H., et al., “Nonstimulant Drugs in the Treatment of Attention Deficit Hyperactivity Disorder,” Child and adolescent psychiatric clinics of North America 1:449-465, (1992).
Grob, C.S., et al., “Suspected Adverse Methylphenidate-Imipramine Interactions in Children,” Developmental & Behavioral Pediatrics 7(4):265-267, Lippincott Williams & Wilkins, United States (Aug. 1986).
Heile, B, “Critical Factors Influencing Gastrointestinal Absorption13 What is the Role of Pellets?” Acta Pharmaceutica Technologica 28 (2):149-157, (1982).
International Search Report and Written Opinion for Application No. PCT/US2015/34466 dated Aug. 26, 2015.
Jewell D.P, “Corticosteroids for the Management of Ulcerative Colitis and Crohn's Disease,” Gastroenterology Clinics of North America 18(1):21-34, ElseVier Health Science Division, United States (Mar. 1989).
Johanson, C.E., et al., “The Discriminative Stimulus Effects of Cocaine in Pigeons,” Pharmacology and Experimental Therapeutics 267(1):1-8, American Society for Pharmacology and Experimental Therapeutics, United States (Oct. 1993).
Johansson, S.A., et al., “Topical and Systemic Glucocorticoid Potencies of Budesonide, Beclomethasone Dipropionate and Prednisolone in Man,” European journal of respiratory diseases (Supplement 122):74-82, Munksgaard International Publishers, Denmark (1982).
Kapseals Dilantin, Physicians' Desk Reference. 51st Edition, Montvale, NJ: Thomson Healthcare, 1997, pp. 1965-1970.
Keshavarzian, A., et al., “MPTP-Induced Duodenal Ulcers in Rat. Prevention by Reuptake Blockers for Serotonin and Norepinephrine, but Not Dopamine,” Gastroenterology 98(3):554-560, W.B. Saunders, United States (1990).
Khutoryanskiy, V.V., “Supramolecular Materials: Longer and Safer Gastric Residence,” Nature Materials 14(10): 963-964, London, Nature Pub. Group, United Kingdom (2015).
Krasznai, A., et al., “Decreased Number of Steroid Receptors of Circulating Lymphocytes in Crohn's Disease and Ulcerative Colitis,” Haematologia (Budapest) 19(4):299-301, Utrecht: Vsp, Netherlands (1986).
Kumana, C.R., et al., “Beclomethasone Dipropionate Enemas for Treating Inflammatory Bowel Disease without Producing Cushing's Syndrome Or Hypothalamic Pituitary Adrenal Suppression,” Lancet 1(8272):579-583, Elsevier, England (Mar. 1982).
Lehmann, K., et al., Practical Course in Lacquer Coating. N.p.: K. Lehmann, 1989.
Levine, D.S., et al., “Coating of Oral Beclomethasone Dipropionate Capsules with Cellulose Acetate Phthalate Enhances Delivery of Topically Active Anti-inflammatory Drug to the Terminal Ileum,” Gastroenterology 92(4):1037-1044, W.B. Saunders, United States (Apr. 1987).
Licamele, W.L., et al., “The Concurrent Use of Lithium and Methylphenidate in a Child,” American Academy of Child and Adolescent Psychiatry 28(5):785-787, Elsevier, United States (Sep. 1989).
Ismo, Physicians' Desk Reference, 51st Edition, Montvale, NJ: Thomson Healthcare, 1997, pp. 2844-2845.
Malchow, H., et al., “Therapy of Crohn's disease,” Deutsche Medizinische Wochenschrift 109(47):1811-1816, Thieme, Germany (Nov. 1984).
Manley, R.H., et al., “Binary Solvents for Zein,” Industrial & Engineering Chemistry 35(6):661-665, American Chemical Society, United States (1943).
McGinity, Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms, Second Edition, pp. 124-125, 272-273, 280-285.
Medical Economics Company. “Anafranil (Clomipramine)” Physicians' Desk Reference. 47th Edition, Montvale, NJ: Thomson Healthcare, 1993, pp. 671-675.
Munday, D.L., et al., “Changes in Drug Release Rate: Effect of Stress Storage Conditions on Film Coated Mini-Tablets,” Drug Development and Industrial Pharmacy 17(15):2135-2143, Inforrna Pharmaceutical Science, United Kingdom (1991).
Norpramin (Desipramine), Physicians' Desk Reference. 47th Edition, Montvale, NJ: Thomson Healthcare, 1993, pp. 1389-1390.
Oberlender, R., et al., “Tomoxetine and the Stereoselectivity of Drug Action,” Pharmacy and Pharmacology 39(12):1055-1066, Pharmaceutical Society of Great Britain, England (Dec. 1987).
Pamelor (Nortriptyline), Physicians' Desk Reference. 47th Edition, Montvale, NJ: Thomson Healthcare, 1993; pp. 2115-2116.
Patrick, K.S., et al., “Pharmacology of the Enantiomers of Threo-Methylphenidate,” Journal of Pharmacology and Experimental Therapeutics 241(1):152-158, American Society for Pharmacology and Experimental Therapeutics, United States (Apr. 1987).
Patrick, K.S., et al., “The Absorption of Sustained-Release Methylphenidate Formulations Compared To an Immediate-Release Formulation,” Biopharmaceutics & Drug Disposition 10(2):165-171, Wiley, England (Mar.-Apr. 1989).
Pelham, W.E., et al., “Relative Efficacy of Long-Acting Stimulants on Children with Attention Deficit-Hyperactivity Disorder: A Comparison of Standard Methylphenidate, Sustained-Release Methylphenidate, Sustained-Release Dextroamphetamine, and Pemoline,” Pediatrics 86(2):226-237, American Academy of Pediatrics, United States (Aug. 1990).
Product Monograph Biphentin, prepared on Mar. 8, 2006, and revised on Jul. 27, 2007.
Prozac (Fiuoxetine), Physicians' Desk Reference, 48th Edition, Montvale, NJ: Thomson Healthcare, 1994, pp. 877-880.
Purdue Pharma. “Biphentin” package insert, Pickering, ON, 2012.
Quinn, D., et al., “Single-Dose Pharmacokinetics of Multilayer-Release Methylphenidate and Immediate-Release Methylphenidate in Children with Attention-Deficit/Hyperactivity Disorder,” Clinical Pharmacology 47(6):760-766, Wiley, England, (Jun. 2007).
Rapport, M.D., et al., “Methylphenidate and Desipramine In Hospitalized Children: Separate and Combined Effects On Cognitive Function,” American Academy of Child and Adolescent Psychiatry 32(2):333-342, Elsevier, United States (Mar. 1993).
Remington: The Science and Practice of Pharmacy, Adejare, A., ed., pp. 1592, Elsevier, Netherlands, 1980.
Ritalin S.R. Physicians' Desk Reference. 46th Edition, Thomson Healthcare, pp. 880-881, New Jersey, United States, 2005.
Ryan N.D, “Heterocyclic Antidepressants in Children and Adolescents,” Child and Adolescent Psychopharmacology 1(1):21-31, Mary Ann Liebert, Inc., United States (1990).
Sinemet, Physicians' Desk Reference, 51st Edition, 1997, pp. 959-963.
Spencer, T., et al., “Nortriptyline Treatment of Children with Attention-Deficit Hyperactivity Disorder and Tic Disorder or Tourette's Syndrome,” American Academy of Child and Adolescent Psychiatry 32(1):205-210, New York : Elsevier, United States (Jan. 1993).
Springer, J.P., et al., “Facilitatory and Inhibitory Effects of Selective Norepinephrine Reuptake Inhibitors on Hypogastric Nerve-Evoked Urethral Contractions in the Cat: A Prominent Role of Urethral Beta-Adrenergic Receptors,” Journal of Urology 152(2 Pt 1):515-519, Elsevier, United States (Aug. 1994).
Steele, M., et al., “A Randomized, Controlled Effectiveness Trial of OROS-Methylphenidate Compared to Usual Care with Immediate-Release Methylphenidate in Attention Deficit-Hyperactivity Disorder,” Clinical Pharmacology 13(1): e50-e62, Pulsus Group [for the] Canadian Society for Clinical Pharmacology, Canada (Winter 2006).
Steinberg, S., et al., “A Case of Mania Associated with Tomoxetine,” American Journal of Psychiatry 142(12):1517-1518, American Psychiatric Association, United States (Dec. 1985).
Swallen, L.C., et al., “Zein. A new industrial protein,” Industrial and Engineering Chemistry 33(3):394-397, American Chemical Society, United States (1941).
Swanson, J.M. et al., “The Use of a Laboratory School Protocol to Evaluate Concepts about Efficacy and Side Effects of New Formulations of Stimulant Medications,” Attention Disorders 6 Suppl 1:S73-S88, SAGE Publications, United States (2002).
Terry, P., et al., “Pharmacological Characterization of the Novel Discriminative Stimulus Effects of a Low Dose of Cocaine,” Pharmacology and Experimental Therapeutics 270(3):1041-1048, American Society for Pharmacology and Experimental Therapeutics, United States (Sep. 1994).
Thomas, P., et al., “Absorption of Delayed-Release Prednisolone in Ulcerative Colitis and Crohn's Disease,” Pharmacy and Pharmacology 37(10):757-758, Wiley, England (Oct. 1985).
Thorazine (Chlorpromazine), Physicians' Desk Reference. 47th Edition, Montvale, NJ: Thomson Healthcare, 1993. pp. 2327-2330.
Tofranil (Imipramine), Physicians' Desk Reference. 47th Edition, Montvale, NJ: Thomson Healthcare, 1993, pp. 1070-1074.
Tyndale, R.F., et al., “Neuronal Cytochrome P45011D1 (debrisoquinelsparteine-Type): Potent Inhibition of Activity by (−)−Cocaine and Nucleotide Sequence Identity to Human Hepatic P450 Gene CYP2D6,” Molecular pharmacology 40(1):63-68, American Society for Pharmacology and Experimental Therapeutics, United States (Jul. 1991).
United States Pharmacopeia Convention. “<2040> Disintegration and Dissolution of Nutritional Supplements.” United States Pharmacopeia I National Formulary. USP231NF18 vol. Rockville, MD: United States Pharmacopeia! Convention, Inc., 1995. 2184-5, 2577-8, 2833-4, 3794-5.
United States Pharmacopeia Convention. “<711> Dissolution.” United States Pharmacopeia I National Formulary. USP231NF18 vol. Rockville, MD: United States Pharmacopeia Convention, Inc., 1995. 1791-4, 2185, 2577-8, 2833-4, 3208-9, 3794-5.
United States Pharmacopeia Convention. “<724> Drug Release.” United States Pharmacopeia I National Formulary. USP231NF18 vol. Rockville, MD: United States Pharmacopeia! Convention, Inc., 1995. 1793-9, 2534-6, 2709-15, 3012-7, 3209-15, 3468-74.
Vitiello, B., et al., “Methylphenidate Dosage for Children with ADHD Over Time Under Controlled Conditions: Lessons From the MTA,” American Academy of Child and Adolescent Psychiatry 40(2):188-196, Elsevier, United States (Feb. 2001).
Wellbutrin (Bupropion), Physicians' Desk Reference, 47th Edition, Montvale, NJ: Thomson Healthcare, 1993, pp. 842-844.
Wilens, T.E., et al., “Nortriptyline in the Treatment of ADHD: A Chart Review of 58 Cases,” American Academy of Child & Adolescent Psychiatry 32(2):343-349, Elsevier, United States (Mar. 1993).
Wolman, S.L., et al., “Use of Oral Budesonide in a Patient with Small Bowel Crohn's Disease and Previous Pseudotumor Cerebri Secondary to Steroids,” Scandinavian Journal of Gastroenterology 24 Suppl 158:146-147, (1989).
Wong, D.T., et al., “A New Inhibitor of Norepinephrine Uptake Devoid of Affinity for Receptors in Rat Brain,” Pharmacology and Experimental Therapeutics 222(1):61-65, Williams & Wilkins, United States (Jul. 1982).
Wong, D.T., et al., “LY248686, a New Inhibitor of Serotonin and Norepinephrine Uptake,” Neuropsychopharmacology 8(1):23-33, Nature Publishing Group, England (Jan. 1993).
Wong, D.T., et al., “The Comparison of Fluoxetine and Nisoxetine with Tricyclic Antidepressants in Blocking the Neurotoxicity of P-Chloroamphetamine and 6-Hydroxydopamine in the Rat Brain,” Research Communications in Chemical Pathology and Pharmacology 15(2):221-231, P. J. D. Publications, United States (Oct. 1976).
Zerbe, R.L., et al., “Clinical Pharmacology of Tomoxetine, a Potential Antidepressant,” Pharmacology and Experimental Therapeutics 232(1):139-143, American Society for Pharmacology and Experimental Therapeutics, United States (Jan. 1985).
Zhang, L., et al., “Imipramine as a Discriminative Stimulus,” Pharmacology and Experimental Therapeutics 259(3):1088-1093, American Society for Pharmacology and Experimental Therapeutics, United States (Dec. 1991).
Rhodes Pharmaceuticals L.P. vs. Actavis, Inc., Actavis Elizabeth LLC, Actavis LLC, and Allergan PLC, Defendants' Preliminary Invalidity Contentions for U.S. Pat. Nos. 6,419,960, 7,083,808, 7,438,930, 7,247,318, 8,580,310, and 9,066,869, Civil Action No. 16-1668 (WHW)(CLW), Oct. 7, 2016. 229 pages.
Malchow, H., et al., “Therapy of Crohn's disease,” Deutsche Medizinische Wochenschrift 109(47):1811-1816, Thieme, Germany (Nov. 1984). (English Translation).
Office Action dated Oct. 11, 2018, in U.S. Appl. No. 16/120,999, Vargas Rincon et al., filed Sep. 4, 2018, 18 pages.
Office Action dated Dec. 4, 2018, in U.S. Appl. No. 16/120,900, Vargas Rincon et al., filed Sep. 4, 2018, 7 pages.
Office Action dated Mar. 22, 2017, in U.S. Appl. No. 14/928,276, Vargas Rincon et al., filed Oct. 30, 2015, 13 pages.
Office Action dated Oct. 16, 2017, in U.S. Appl. No. 14/928,276, Vargas Rincon et al., filed Oct. 30, 2015, 24 pages.
Coghill, D., et al., “Long-acting methylphenidate formulations in the treatment of attention-defecit/hyperactivity disorder: a systematic review of head-to-head studies,” BMC Psychiatry13:1-24, Springer Nature, United Kingdom (2013).
Office Action dated Feb. 1, 2019, in U.S. Appl. No. 16/185,730, Vargas Rincon et al., filed Nov. 9, 2018, 13 pages.
Office Action dated Apr. 4, 2019, in U.S. Appl. No. 16/282,858, Vargas Rincon et al., filed Feb. 22, 2019, 10 pages.
Office Action dated Apr. 4, 2019, in U.S. Appl. No. 16/282,876, Vargas Rincon et al., filed Feb. 22, 2019, 8 pages.
Office Action dated May 31, 2019, in U.S. Appl. No. 16/399,580, Vargas Rincon et al., filed Apr. 30, 2019, 9 pages.
Office Action dated Jun. 14, 2019, in U.S. Appl. No. 16/399,574, Vargas Rincon et al., filed Apr. 30, 2019, 12 pages.
Office Action dated Jun. 13, 2019, in U.S. Appl. No. 16/195,418, Donnelly, G., et al., filed Nov. 19, 2018, 7 pages.
Office Action dated Jul. 22, 2019, in U.S. Appl. No. 16/422,177, inventor Vargas Rincon, R.A., et al., filed May 24, 2019, 14 pages.
Office Action dated Aug. 19, 2019, in U.S. Appl. No. 16/399,571, inventor Vargas Rincon, R.A., et al., filed Apr. 30, 2019, 18 pages.
Office Action dated Jan. 30, 2020, in U.S. Appl. No. 16/399,571, inventor Vargas Rincon, R.A., et al., filed Apr. 30, 2019, 10 pages.
Notification of Reexamination for Chinese Patent Application No. 201580059528.9, dated May 26, 2022, The China National Intellectual Property Administration, Beijing, China, 8 pages.
XU, J ., Practical Prescription Pharmacology, Yang, B., ed., pp.352, People's Army Medical Publishing House, Beijing, China (2009).
2014 National Health Professional and Technical Qualification Examination Guidance Book: Pharmacy (Intermediate), Version 4, Chen, Y., ed., pp. 342-343, China Medical Science and Technology Press, Beijing, China (2013).
Related Publications (1)
Number Date Country
20220233456 A1 Jul 2022 US
Provisional Applications (1)
Number Date Country
62122847 Oct 2014 US
Divisions (1)
Number Date Country
Parent 16399571 Apr 2019 US
Child 16880639 US
Continuations (4)
Number Date Country
Parent 16880639 May 2020 US
Child 17647149 US
Parent 16120999 Sep 2018 US
Child 16399571 US
Parent 15958413 Apr 2018 US
Child 16120999 US
Parent 14928276 Oct 2015 US
Child 15958413 US