USRE41148E1 - Oral pulsed dose drug delivery system - Google Patents

Oral pulsed dose drug delivery system Download PDF

Info

Publication number
USRE41148E1
USRE41148E1 US11/091,010 US9101099A USRE41148E US RE41148 E1 USRE41148 E1 US RE41148E1 US 9101099 A US9101099 A US 9101099A US RE41148 E USRE41148 E US RE41148E
Authority
US
United States
Prior art keywords
formulation
release
coating
enteric
amphetamine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US11/091,010
Inventor
Beth A. Burnside
Xiaodi Guo
Kimberly Fiske
Richard A. Couch
Donald J. Treacy
Rong-Kun Chang
Edward M. Rudnic
Charlotte M. McGuinness
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shire LLC
Original Assignee
Shire Laboratories Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22644775&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE41148(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Shire Laboratories Inc filed Critical Shire Laboratories Inc
Priority to US11/091,010 priority Critical patent/USRE41148E1/en
Assigned to SHIRE LABORATORIES, INC. reassignment SHIRE LABORATORIES, INC. RELEASE OF SECURITY INTEREST UNDER THE AMENDED AND RESTATED CREDIT AGREEMENT Assignors: DLJ CAPITAL FUNDING, INC. AS ADMINISTRATIVE AGENT
Assigned to SHIRE LLC reassignment SHIRE LLC MERGER (SEE DOCUMENT FOR DETAILS). Assignors: SHIRE LABORATORIES, INC.
Application granted granted Critical
Publication of USRE41148E1 publication Critical patent/USRE41148E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5026Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • A61K9/5047Cellulose ethers containing no ester groups, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5073Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings
    • A61K9/5078Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings with drug-free core
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • This invention pertains to a multiple dosage form delivery system comprising one or more amphetamine salts for administering the amphetamine salts to a recipient.
  • sustained release delivery is not suitable and is affected by the following factors:
  • Pulsed dose delivery systems prepared as either single unit or multiple unit formulations, and which are capable of releasing the drug after a predetermined time, have been studied to address the aforementioned problematic areas for sustained release preparations. These same factors are also problematic in pulsed dose formulations development. For example, gastrointestinal transit times vary not only from patient to patient but also within patients as a result of food intake, stress, and illness; thus a single-unit pulsed-release system may give higher variability compared to a multiple unit system. Additionally, drug layering or core making for multiple unit systems is a time-consuming and hard-to-optimize process. Particularly challenging for formulation scientists has been overcoming two conflicting hurdles for pulsatile formulation development, i.e., lag time and rapid release.
  • enteric materials e.g., cellulose acetate phtalate, hydroxypropyl methylcellulose phtalate, polyvinyl acetate phthalate, and the EUDRAGIT® acrylic polymers
  • enteric materials which are soluble at higher pH values, are frequently used for colon-specific delivery systems. Due to their pH-dependent attributes and the uncertainty of gastric retention time, in-vivo performance as well as inter- and intra-subject variability are major issues for using enteric, coated systems as a time-controlled release of drugs.
  • a retarding swellable hydrophilic coating has been used for oral delayed release systems (4,5). It was demonstrated that lag time was linearly correlated with coating weight gain and drug release was pH independent.
  • Hydroxypropyl methylcellulose barriers with erodible and/or gellable characteristics formed using press coating technology for tablet dosage forms have been described to achieve time-programmed release of drugs (6).
  • bimodal release is characterized by a rapid initial release, followed by a period of constant release, and finalized by a second rapid drug release.
  • Tablets or capsules coated with a hydrophobic wax-surfactant layer made from an aqueous dispersion of carnauba wax, beeswax, polyoxyethylene sorbitan monooleate, and hydroxypropyl methylcellulose have been used for rapid drug release after a predetermined lag time.
  • a hydrophobic wax-surfactant layer made from an aqueous dispersion of carnauba wax, beeswax, polyoxyethylene sorbitan monooleate, and hydroxypropyl methylcellulose
  • a sustained-release drug delivery system is described in U.S. Pat. No. 4,871,549.
  • This system When this system is placed into dissolution medium or the gastrointestinal tract, water influx and the volume expansion of the swelling agent cause the explosion of the water permeable membrane. The drug thus releases after a predetermined time period.
  • the OROS® push-pull system (Alza Company) has been developed for pulsatile delivery of water-soluble and water-insoluble drugs (9,10), e.g. the OROS-CT® system and is based on the swelling properties of an osmotic core compartment which provides a pH-independent, time-controlled drug release.
  • the PULSINCAP® dosage form releases its drug content at either a predetermined time or at a specific site (e.g., colon) in the gastrointestinal tract (11).
  • the drug formulation is contained within a water-insoluble capsule body and is sealed with a hydrogel plug.
  • the capsule cap dissolves in the gastric juice and the hydrogel plug swells.
  • the swollen plug is ejected from the PULSINCAP® dosage form and the encapsulated drug is released.
  • a pulsatile capsule system containing captopril with release after a nominal 5-hr period was found to perform reproducibly in dissolution and gamma scintigraphy studies. However, in the majority of subjects, no measurable amounts of the drug were observed in the blood, possibly due to instability of the drug in the distal intestine. (12)
  • ADDERALL® comprises a mixture of four amphetamine salts, dextroamphetamine sulfate, dextroamphetamine saccharate, amphetamine asparate monohydrate and amphetamine sulfate, which in combination, are indicated for treatment of Attention Deficity Hyperactivity Disorder in children from 3-10 years of age.
  • One disadvantage of current treatment is that a tablet form is commonly used which many young children have difficulty in swallowing.
  • Another disadvantage of current treatment is that two separate dose are administered, one in the morning and one approximately 4-6 hours later, commonly away from home under other than parental supervision. This current form of treatment, therefore, requires a second treatment which is time-consuming, inconvenient and may be problematic for those children having difficulty in swallowing table formulations.
  • FIG. 1 illustrates the desired target plasma level profile of the pharmaceutical active contained within the delivery system.
  • a pharmaceutical composition for delivering one or more pharmaceutically active amphetamine salts that includes:
  • the immediate release and enteric release portions of the composition are present on the same core.
  • immediate release and enteric release components are present on different cores.
  • composition may include a combination of the hereinabove referred to cores (one or more cores that include both components on the same core and one or more cores that include only one of the two components on the core).
  • the present invention provides a composition in which there is immediate release of drug and enteric release of drug wherein the enteric release is a pulsed release and wherein the drug includes one or more amphetamine salts and mixtures thereof.
  • the immediate release component releases the pharmaceutical agent in a pulsed dose upon oral administration of the delivery system.
  • the enteric release coating layer retards or delays the release of the pharmaceutical active or drug for a specified time period (“lag time”) until a predetermined time, at which time the release of the drug is rapid and complete, i.e., the entire dose is released within about 30-60 minutes under predetermined environmental conditions, i.e. a particular location within the gastrointestinal tract.
  • the delay or lag time will take into consideration factors such as transit times, food effects, inflammatory bowel disease, use of antacids or other medicaments which alter the pH of the GI tract.
  • the lag time period is only time-dependent, i.e., pH independent.
  • the lag time is preferably within 4 to 6 hours after oral administration of the delivery system.
  • the present invention is directed to a composition that provides for enteric release of at least one pharmaceutically active amphetamine salt, including at least one pharmaceutically active amphetamine salt that is coated with an enteric coating wherein (1) the enteric release coating has a defined minimum thickness and/or (2) there is a protective layer between the at least one pharmaceutically active amphetamine salt and the enteric release coating and/or (3) there is a protective layer over the enteric release coating.
  • Typical enteric coating levels did not meet the above requirements for the desired dosage profile of amphetamine salts. Using the typical amount of enteric coating (10-20 ⁇ ) resulted in undesired premature leakage of the drug from the delivery system into the upper gastrointestinal tract and thus no drug delivery at the desired location in the gastrointestinal tract after the appropriate lag time. Thus this coating did not meet the requirements for the drug release profile to provide full beneficial therapeutic activity at the desired time.
  • the pulsed enteric release of the amphetamine salts is accomplished by employing a certain minimum thickness of the enteric coating.
  • the pulsed dose delivery comprises a composition which comprises one or more pharmaceutically active amphetamine salts; an enteric coating over the one or more pharmaceutically active amphetamine salts, wherein the thickness of the enteric coating layer is at least 25 ⁇ ; a further layer of one or more pharmaceutically active amphetamine salts over the enteric coating layer; and an immediate release layer coating.
  • the thicker enteric coating surprisingly provides the required delayed immediate release of the pharmaceutically active amphetamine salt at the desired time in the desired area of the gastrointestinal tract.
  • FIG. 2 illustrates a model of this delivery system.
  • the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed around which the enteric coating is applied.
  • a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
  • a delayed immediate release drug delivery can also be accomplished by coating the drug first with a protective layer prior to applying the enteric coating.
  • the pulsed enteric release is accomplished by employing a protective layer between the drug and the enteric coating.
  • the enteric coating may be of an increased thickness or may be of lower thickness.
  • the object of the invention is met by providing a composition comprising one or more pharmaceutically active amphetamine salts; a protective layer coating over the one or more pharmaceutically active amphetamine salt layer(s), and an enteric coating layer over the protective coating layer; a further pharmaceutically active amphetamine salt layer and an immediate release layer coating.
  • the thickness of the enteric coating is at least 25 ⁇
  • the protective layer comprises an immediate release coating.
  • the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed, during the core seed manufacturing process, around which the protective coating is applied.
  • a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
  • the pulsed enteric release is accomplished by employing a protective layer over the enteric coating.
  • a pulsed dose release drug delivery system comprising one or more pharmaceutically active amphetamine salts; an enteric coating layer over the pharmaceutically active amphetamine salt layer(s); and a protective layer over the enteric coating; a second pharmaceutically active amphetamine salt layer; and an immediate release layer coating.
  • the protective layer comprises a semi-permeable polymer and an immediate release coating layer.
  • the modifying layer comprises a first layer of a semi-permeable polymer which is adjacent to the enteric coating layer and a second coating layer over the semi-permeable polymer coating layer comprising an immediate release polymer coating layer.
  • a semi-permeable polymer which may comprise a low water-permeable pH-insensitive polymer, is layered onto the outer surface of the enteric layer, in order to obtain prolonged delayed release time.
  • This semi-permeable polymer coating controls the erosion of the pH-sensitive enteric polymer in an alkaline pH environment in which a pH-sensitive polymer will dissolve rapidly.
  • Another pH-sensitive layer may be applied onto the surface of a low water-permeability layer to further delay the release time.
  • the composition in addition to a protective layer, comprises an acid which is incorporated into the pharmaceutical active layer or coated onto the surface of the active layer to reduce the pH value of the environment around the enteric polymer layer.
  • the acid layer may also be applied on the outer layer of the pH-sensitive enteric polymer layer, followed by a layer of low water-permeability polymer. The release of the active thus may be delayed and the dissolution rate may be increased in an alkaline environment.
  • the protective coating may be used both over the drug and over the enteric coating.
  • the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed, during the core seed manufacturing process, around which the enteric coating is applied.
  • a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
  • the drug delivery system of the present invention as described herein preferably comprises one or a number of beads or beadlets in a dosage form, either capsule, tablet, sachet or other method of orally administering the beads.
  • FIG. 1 illustrates a multiple pulse drug delivery system target plasma profile of the drug delivery system of the present invention.
  • the profile reflects an immediate-release component followed by a delayed-release component.
  • FIG. 2a graphically illustrates a pulsed dose delivery system.
  • FIGS. 2b and c graphically illustrate the drug release mechanism from the proposed delivery system.
  • FIG. 3 is a plot of the present drug released versus time from the drug-loaded pellets described in Example 1 which exemplifies the immediate release component of the present invention.
  • FIG. 4 is a plot of the percent drug released versus time from the coated pellets described in Example 2 which exemplifies the immediate release component and the delayed release components of the present invention.
  • FIG. 5 is a plot of the percent drug released versus time from the coated pellets described in Example 3 which exemplifies the immediate release component and the delayed release components of the present invention.
  • FIG. 6 illustrates the drug release profile of coated pellets described in Example 4 which exemplifies the immediate release component and the delayed release components of the present invention.
  • FIG. 7 is a plot of a profile of plasma amphetamine concentration after administration of a composite capsule containing the immediate release pellets and delayed release pellets from Examples 1 and 2, respectively.
  • FIG. 8 is a plot of a profile of plasma amphetamine concentration after administration of a composite capsule containing the immediate release pellets and delayed release pellets from Examples 1 and 3, respectively.
  • the present invention comprises a core or starting seed, either prepared or commercially available product.
  • the cores or starting seeds can be sugar spheres; spheres made from microcrystalline cellulose and any suitable drug crystals.
  • the materials that can be employed in making drug-containing pellets are any of those commonly used in pharmaceutics and should be selected on the basis of compatibility with the active drug and the physicochemical properties of the pellets.
  • the additives except active drugs are chosen below as examples.
  • Binders such as cellulose derivatives such as methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer and the like.
  • Disintegration agents such as corn starch, pregelatinized starch, cross-linked carboxymethylcellulose (AC-DI-SOL®), sodium starch glycolate (EXPLOTAB®), cross-linked polyvinylpyrrolidone (PLASDONE XL®), and any disintegration agents used in tablet preparations.
  • Filling agents such as lactose, calcium carbonate, calcium phosphate, calcium sulfate, microcrystalline cellulose, dextran, starches, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • Surfactants such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, bile salts, glyceryl monostearate, PLURONIC® line (BASF), and the like.
  • Solubilizers such as citric acid, succinic acid, fumaric acid, malic acid, tartaric acid, maleic acid, glutaric acid sodium bicarbonate and sodium carbonate and the like.
  • Stabilizers such as any antioxidation agents, buffers, acids, and the like, can also be utilized.
  • Methods of manufacturing the core include
  • These particles can then be coated in a fluidized bed apparatus with an alternating sequence of coating layers.
  • the core may be coated directly with a layer or layers of at least one pharmaceutically active amphetamine salts and/or the pharmaceutically active amphetamine salt may be incorporated into the core material.
  • Pharmaceutical active amphetamine salts contemplated to be within the scope of the present invention include amphetamine base, all chemical and chiral derivatives and salts thereof; methylphenidate, all chemical and chiral derivatives and salts thereof; phenylpropanolamine and its salts; and all other compounds indicated for the treatment of attention deficit hyperactivity disorder (ADHD).
  • ADHD attention deficit hyperactivity disorder
  • a protective layer may be added on top of the pharmaceutical active containing layer and also may be provided between active layers.
  • a separation or protective layer may be added onto the surface of the active-loaded core, and then the enteric layer is coated thereupon.
  • Another active layer may also be added to the enteric layer to deliver an initial dose.
  • the enteric coating layer is applied onto the cores with or without seal coating by conventional coating techniques, such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. All commercially available pH-sensitive polymers are included.
  • the pharmaceutical active is not released in the acidic stomach environment of approximately below pH 4.5, but not limited to this value. The pharmaceutical active should become available when the pH-sensitive layer dissolves at the greater pH, after a certain delayed time; or after the unit passes through the stomach.
  • the preferred delay time is in the range of two to six hours.
  • Enteric polymers include cellulose acetate phthalate, Cellulose acetate trimellitate, hydroxypropyl methylcellulose phtalate, polyvinyl acetate phtalate, carboxymethylethylcellulose, co-polymerized methacrylic acid/methacrylic acid methyl esters such as, for instance, materials known under the trade name EUDRAGIT® L12.5, L100, or EUDRAGIT® S12.5, S100 or similar compounds used to obtain enteric coatings.
  • Aqueous collodial polymer dispersions or re-dispersions can be also applied, e.g.
  • the enteric polymers used in this invention can be modified by mixing with other known coating products that are not pH sensitive.
  • coating pro ducts include the neutral methacrylic acid esters with a small portion of trimethylammonioethyl methacrylate chloride, sold currently under the trade names EUDRAGIT® RS and EUDRAGIT® RL; a neutral ester dispersion without any functional groups, sold under the trade names EUDRAGIT® NE30D; and other pH independent coating products.
  • the modifying component of the protective layer used over the enteric coating can include a water penetration barrier layer (semipermeable polymer) which can be successively coated after the enteric coating to reduce the water penetration rate through the enteric coating layer and thus increase the lag time of the drug release.
  • a water penetration barrier layer semipermeable polymer
  • Sustained-release coatings commonly known to one skilled in the art can be used for this purpose by conventional coating techniques such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions.
  • the following materials can be used, but not limited to: Cellulose acetate, Cellulose acetate butyrate, Cellulose acetate propionate, Ethyl cellulose, Fatty acids and their esters, Waxes, zein, and aqueous polymer dispersions such as EUDRAGIT® RS and SURELEASE®, cellulose acetate latex.
  • aqueous polymer dispersions such as EUDRAGIT® RS and SURELEASE®, cellulose acetate latex.
  • hydrophilic polymers such as Hydroxyethyl cellulose, Hydroxypropyl cellulose (KLUCEL®, Hercules Corp.), Hydroxypropyl methylcellulose (METHOCEL®, Dow Chemical Corp.), Polyvinylpyrrolidone can also be used.
  • An overcoating layer can further optionally be applied to the composition of the present invention.
  • OPADRY®, OPADRY II® (Colorcon) and corresponding color and colorless grades from Colorcon can be used to protect the pellets from being tacky and provide colors to the product.
  • the suggested levels of protective or color coating are from 1 to 6%, preferably 2-3% (w/w).
  • ingredients can be incorporated into the overcoating formula, for example to provide a quicker immediate release, such as plasticizers: acetyltriethyl citrate, triethyl citrate, acetyltributyl citrate; dibutylsebacate, triacetin, polyethylene glycols, propylene glycol and the others; lubricants: talc, colloidal silica dioxide, magnesium stearate, calcium stearate, titanium dioxide, magnesium silicate, and the like.
  • composition preferably in beadlet form, can be incorporated into hard gelatin capsules, either with additional excipients, or alone.
  • Typical excipients to be added to a capsule formulation include, but are not limited to: filters such as microcrystalline cellulose, soy polysaccharides, calcium phosphate dihydrate, calcium sulfate, lactose, sucrose, sorbitol, or any other inert filler.
  • filters such as microcrystalline cellulose, soy polysaccharides, calcium phosphate dihydrate, calcium sulfate, lactose, sucrose, sorbitol, or any other inert filler.
  • there can be flow aids such as fumed silicon dioxide, silica gel, magnesium stearate, calcium stearate or any other material imparting flow to powders.
  • a lubricant can further be added if necessary by using polyethylene glycol, leucine, glyceryl behenate, magnesium stearate or calcium stearate.
  • the composition may also be incorporated into a tablet, in particular by incorporation into a tablet matrix, which rapidly disperses the particles after ingestion.
  • a filler/binder In order to incorporate these particles into such a tablet, a filler/binder must be added to a table that can accept the particles, but will not allow their destruction during the tableting process.
  • Materials that are suitable for this purpose include, but are not limited to, microcrystalline cellulose (AVICEL®), soy polysaccharide (EMCOSOY®), pre-gelatinized starches (STARCH® 1500, NATIONAL® 1551), and polyethylene glycols (CARBOWAX®).
  • the materials should be present in the range of 5-75% (w/w), with a preferred range of 25-50% (w/w).
  • disintegrants are added in order to dispense the beads once the tablet is ingested.
  • Suitable disintegrants include, but are not limited to: cross-linked sodium carboxymethyl cellulose (AC-DI-SOL®), sodium starch glycolate (EXPLOTAB®, PRIMOJEL®), and cross-linked polyvinylpolypyrrolidone (Plasone-XL). These materials should be present in the rate of 3-15% (w/w), with a preferred range of 5-10% (w/w).
  • Lubricants are also added to assure proper tableting, and these can include, but are not limited to: magnesium stearate, calcium stearate, stearic acid, polyethylene glycol, leucine, glyceryl behanate, and hydrogenated vegetable oil. These lubricants should be present in amounts from 0.1-10% (w/w), with a preferred range of 0.3-3.0% (w/w).
  • Tablets are formed, for example, as follows.
  • the particles are introduced into a blender along with AVICEL®, disintegrants and lubricant, mixed for a set number of minutes to provide a homogeneous blend which is then put in the hopper of a tablet press with which tablets are compressed.
  • the compression force used is adequate to form a tablet; however, not sufficient to fracture the beads or coatings.
  • the multiple dosage form of the present invention can deliver rapid and complete dosages of pharmaceutically active amphetamine salts to achieve the desired levels of the drug in a recipient over the course of about 8 hours with a single oral administration.
  • the levels of drug in blood plasma of the pharmaceutically active amphetamine salts will reach a peak fairly rapidly after about 2 hours, and after about 4 hours a second pulse dose is released, wherein a second fairly rapid additive increase of plasma drug levels occurs which slowly decreases over the course of the next 12 hours.
  • Nonpareil seeds (30/35 mesh, Paulaur Corp., NJ), 6.8 kg were put into a FLM-15 fluid bed processor with a 9′′ Wurster column and fluidized at 60° C.
  • the suspension of mixed amphetamine salts (MAS) with 1% HPMC E5 Premium (Dow Chemical) as a binder was sprayed onto the seed under suitable conditions. Almost no agglomeration and no fines were observed with a yield of at least 98%.
  • the drug-loaded cores were used to test enteric coatings and sustained release coatings.
  • the drug release profile of the drug-loaded pellets of this example is shown in FIG. 3 .
  • the following formulation was used to coat the mixed amphetamine salts loaded (MASL) pellets from Example 1 with the EUDRAGIT® L 30D-55 (Rohm Pharma, Germany) coating dispersion.
  • 2 kg of MASL pellets were loaded into a fluid bed processor with a reduced Wurster column equipped with a precision coater (MP 2/3, Niro Inc) (see Examples 3 and 4 ).
  • the coating dispersion was prepared by dispersing Triethyl citrate, Talc and EUDRAGIT® L 30D-55 into water and mixing for at least 30 minutes. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidized MASL pellets. The spraying was continued until the targeted coating level was achieved (20 ⁇ ) .
  • the coated pellets were dried at 30-35° C. for 5 minutes before stopping the process.
  • the enteric coated PPA MASL pellets were tested at different pH buffers by a USP paddle method.
  • the drug content was analyzed using HPLC. The results showed that the enteric coating delayed the drug release from the coated pellets until after exposure to pH 6 or higher. (Reference #AR98125-4)
  • the drug release profile of the coated pellets of this example is shown in FIG. 4 .
  • the following formulation was used to coat the MASL pellets from Example 1 with the EUDRAGIT® 4110D (Rohm Pharma, Germany) coating dispersion.
  • MASL pellets (2 kg) were loaded in a fluid bed processor with a reduced Wurster column (GPGC-15, Glatt).
  • the coating dispersion was prepared by dispersing Triethyl citrate, Talc and EUDRAGIT® 4110D into water and mixing for at least 30 minutes. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidized MASL pellets. The spraying was continued until the targeted coating level was achieved.
  • the coated pellets were dried at 30-35° C. for 5 minutes before stopping the process.
  • the enteric coated MASL pellets were tested using a USP paddle method at different pH buffers. The drug content was analyzed using HPLC. The enteric coating delayed the drug release for several hours from the coated pellets until the pH value reached 6.8 or higher, as shown below in Table 3. (Reference #AR98125-3)
  • the drug release profile of coated pellets of this example is shown in FIG. 5 .
  • the following formulation was selected to coat the enteric coated MASL pellets.
  • Coated MASL pellets from Example 2 or coated MASL pellets from Example 3 (2 kg of either) were loaded into a fluid bed processor with a reduced Wurster column (GPGC-15, Glatt).
  • the coating dispersion was prepared by mixing SURELEASE® (Colorcon) and water for at least 15 minutes prior to spraying. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidizated pellets. The spraying was continued until the targeted coating level was achieved.
  • the coated pellets were coated with a thin layer of OPADRY® white (Colorcon) (2%) to prevent the tackiness of the coated pellets during storage. The coated pellets were then dried at 35-40° C.
  • the drug release profile of the coated pellets from this example is shown in FIG. 6 .
  • a pulsatile delivery system can be achieved by combining the immediate release pellets (Example 1) with delayed release pellets (Example 2 or Example 3).
  • the immediate-release pellets equivalent to half the dose and the delayed-release pellets equivalent to half the dose are filled into a hard gelatin capsule to produce the oral pulsed dose delivery system.
  • the delayed-release portion releases the amphetamine salts rapidly and completely, after a specified lag time.
  • the capsule products containing immediate-release pellets and delayed-release pellets (Example 1 plus Example 2 and Example 1 plus Example 3) were tested in a crossover human study.
  • FIG. 7 and 8 show the typical profiles of plasma amphetamine concentration after administration of a composite capsule containing the immediate-release pellets and delayed-release pellets from Examples 1 and 2 (10 mg dose each pellet type) and a capsule containing the pellets from immediate-release pellets and delayed-release pellets from Examples 1 and 3 (10 mg dose each pellet type), respectively.
  • the general plasma profiles are similar to the desired target plasma level profile shown in FIG. 1 .

Abstract

A multiple pulsed dose drug delivery system for pharmaceutically active amphetamine salts, comprising an immediate-release component and an enteric delayed-release component wherein (1) the enteric release coating has a defined minimum thickness and/or (2) there is a protective layer between the pharmaceutically active amphetamine salt and the enteric release coating and/or (3) there is a protective layer over the enteric release coating. The product can be composed of either one or a number of beads in a dosage form, including either capsule, tablet, or sachet method for administering the beads.

Description

This application is a 371 of PCT/US99/24554 filed Oct. 20, 1999, which is continuation-in-part of application Ser. No. 09/176,542, filed Oct. 21, 1998, now U.S. Pat. No. 6,322,819 the contents of which are incorporated herein by reference.
This invention pertains to a multiple dosage form delivery system comprising one or more amphetamine salts for administering the amphetamine salts to a recipient.
BACKGROUND OF THE INVENTION
Traditionally, drug delivery systems have focused on constant/sustained drug output with the objective of minimizing peaks and valleys of drug concentrations in the body to optimize drug efficacy and to reduce adverse effects. A reduced dosing frequency and improved patient compliance can also be expected for the controlled/sustained release drug delivery systems, compared to immediate release preparations. However, for certain drugs, sustained release delivery is not suitable and is affected by the following factors:
    • First pass metabolism: Some drugs, such as β blockers, β-estradiol, and salicylamide, undergo extensive first pass metabolism and require fast drug input to saturate metabolizing enzymes in order to minimize pre-systemic metabolism. Thus, a constant/sustained oral method of delivery would result in reduced oral bio-availability.
    • Biological tolerance: Continuous release drug plasma profiles are often accompanied by a decline in the pharmacotherapeutic effect of the drug, e.g., biological tolerance of transdermal nitroglycerin.
    • Chronopharmacology and circadian rhythms: Circadian rhythms in certain physiological functions are well established. It has been recognized that many symptoms and onset of disease occur during specific time periods of the 24 hour day, e.g., asthma and angina pectoris attacks are most frequently in the morning hours (1,2).
    • Local therapeutic need: For the treatment of local disorders such as inflammatory bowel disease, the delivery of compounds to the site of inflammation with no loss due to absorption in the small intestine is highly desirable to achieve the therapeutic effect and to minimize side effects.
    • Gastric irritation or drug instability in gastric fluid: For compounds with gastric irritation or chemical instability in gastric fluid, the use of a sustained release preparation may exacerbate gastric irritation and chemical instability in gastric fluid.
    • Drug absorption differences in various gastrointestinal segments: In general, drug absorption is moderately slow in the stomach, rapid in the small intestine, and sharply declining in the large intestine. Compensation for changing absorption characteristics in the gastrointestinal tract may be important for some drugs. For example, it is rational for a delivery system to pump out the drug much faster when the system reaches the distal segment of the intestine, to avoid the entombment of the drug in the feces.
Pulsed dose delivery systems, prepared as either single unit or multiple unit formulations, and which are capable of releasing the drug after a predetermined time, have been studied to address the aforementioned problematic areas for sustained release preparations. These same factors are also problematic in pulsed dose formulations development. For example, gastrointestinal transit times vary not only from patient to patient but also within patients as a result of food intake, stress, and illness; thus a single-unit pulsed-release system may give higher variability compared to a multiple unit system. Additionally, drug layering or core making for multiple unit systems is a time-consuming and hard-to-optimize process. Particularly challenging for formulation scientists has been overcoming two conflicting hurdles for pulsatile formulation development, i.e., lag time and rapid release.
Various enteric materials, e.g., cellulose acetate phtalate, hydroxypropyl methylcellulose phtalate, polyvinyl acetate phthalate, and the EUDRAGIT® acrylic polymers, have been used as gastroresistant, enterosoluble coatings for single drug pulse release in the intestine (3). The enteric materials, which are soluble at higher pH values, are frequently used for colon-specific delivery systems. Due to their pH-dependent attributes and the uncertainty of gastric retention time, in-vivo performance as well as inter- and intra-subject variability are major issues for using enteric, coated systems as a time-controlled release of drugs.
A retarding swellable hydrophilic coating has been used for oral delayed release systems (4,5). It was demonstrated that lag time was linearly correlated with coating weight gain and drug release was pH independent.
Hydroxypropyl methylcellulose barriers with erodible and/or gellable characteristics formed using press coating technology for tablet dosage forms have been described to achieve time-programmed release of drugs (6). Barrier formulation variables, such as grade of hydroxypropyl methylcellulose, water-soluble and water-insoluble excipients, significantly altered the lag time and the release rate from the center cores.
Special grades of hydroxypropyl methylcellulose, e.g., METHOLOSE® 60SH, 90SH (Shin-Etsu Ltd., Japan), and METHOCEL® F4M (Dow Chemical Company, USA), as a hydrophilic matrix material have been used to achieve bimodal drug release for several drugs, i.e., aspirin, ibuprofen, and adinazolam (7). Bimodal release is characterized by a rapid initial release, followed by a period of constant release, and finalized by a second rapid drug release.
Tablets or capsules coated with a hydrophobic wax-surfactant layer, made from an aqueous dispersion of carnauba wax, beeswax, polyoxyethylene sorbitan monooleate, and hydroxypropyl methylcellulose have been used for rapid drug release after a predetermined lag time. However, even though a two-hour lag time was achieved for the model drug theophylline at a higher coating level (60%), three hours were required for a complete release of theophylline after the lag time. (8)
A sustained-release drug delivery system is described in U.S. Pat. No. 4,871,549. When this system is placed into dissolution medium or the gastrointestinal tract, water influx and the volume expansion of the swelling agent cause the explosion of the water permeable membrane. The drug thus releases after a predetermined time period. The OROS® push-pull system (Alza Company) has been developed for pulsatile delivery of water-soluble and water-insoluble drugs (9,10), e.g. the OROS-CT® system and is based on the swelling properties of an osmotic core compartment which provides a pH-independent, time-controlled drug release.
The PULSINCAP® dosage form releases its drug content at either a predetermined time or at a specific site (e.g., colon) in the gastrointestinal tract (11). The drug formulation is contained within a water-insoluble capsule body and is sealed with a hydrogel plug. Upon oral administration, the capsule cap dissolves in the gastric juice and the hydrogel plug swells. At a controlled and predetermined time point, the swollen plug is ejected from the PULSINCAP® dosage form and the encapsulated drug is released. A pulsatile capsule system containing captopril with release after a nominal 5-hr period was found to perform reproducibly in dissolution and gamma scintigraphy studies. However, in the majority of subjects, no measurable amounts of the drug were observed in the blood, possibly due to instability of the drug in the distal intestine. (12)
ADDERALL® comprises a mixture of four amphetamine salts, dextroamphetamine sulfate, dextroamphetamine saccharate, amphetamine asparate monohydrate and amphetamine sulfate, which in combination, are indicated for treatment of Attention Deficity Hyperactivity Disorder in children from 3-10 years of age. One disadvantage of current treatment is that a tablet form is commonly used which many young children have difficulty in swallowing. Another disadvantage of current treatment is that two separate dose are administered, one in the morning and one approximately 4-6 hours later, commonly away from home under other than parental supervision. This current form of treatment, therefore, requires a second treatment which is time-consuming, inconvenient and may be problematic for those children having difficulty in swallowing table formulations.
SUMMARY OF THE INVENTION
Accordingly, in view of a need for successfully administering a multiple unit pulsed dose of amphetamine salts and mixtures thereof, the present invention provides an oral multiple unit pulsed dose delivery system for amphetamine salts and mixtures thereof. FIG. 1 illustrates the desired target plasma level profile of the pharmaceutical active contained within the delivery system.
In accordance with a preferred embodiment of the present invention, there is provided a pharmaceutical composition for delivering one or more pharmaceutically active amphetamine salts that includes:
    • (a) one or more pharmaceutically active amphetamine salts that are covered with an immediate release coating, and
    • (b) one or more pharmaceutically active amphetamine salts that are covered with an enteric release coating wherein (1) the enteric release coating has a defined minimum thickness and/or (2) there is a protective layer between the at least one pharmaceutically active amphetamine salt and the enteric release coating and/or (3) there is a protective layer over the enteric release coating.
In one embodiment, the immediate release and enteric release portions of the composition are present on the same core.
In another embodiment, the immediate release and enteric release components are present on different cores.
It is also contemplated that the composition may include a combination of the hereinabove referred to cores (one or more cores that include both components on the same core and one or more cores that include only one of the two components on the core).
The present invention provides a composition in which there is immediate release of drug and enteric release of drug wherein the enteric release is a pulsed release and wherein the drug includes one or more amphetamine salts and mixtures thereof.
The immediate release component releases the pharmaceutical agent in a pulsed dose upon oral administration of the delivery system.
The enteric release coating layer retards or delays the release of the pharmaceutical active or drug for a specified time period (“lag time”) until a predetermined time, at which time the release of the drug is rapid and complete, i.e., the entire dose is released within about 30-60 minutes under predetermined environmental conditions, i.e. a particular location within the gastrointestinal tract.
The delay or lag time will take into consideration factors such as transit times, food effects, inflammatory bowel disease, use of antacids or other medicaments which alter the pH of the GI tract.
In a preferred embodiment, the lag time period is only time-dependent, i.e., pH independent. The lag time is preferably within 4 to 6 hours after oral administration of the delivery system.
In one aspect, the present invention is directed to a composition that provides for enteric release of at least one pharmaceutically active amphetamine salt, including at least one pharmaceutically active amphetamine salt that is coated with an enteric coating wherein (1) the enteric release coating has a defined minimum thickness and/or (2) there is a protective layer between the at least one pharmaceutically active amphetamine salt and the enteric release coating and/or (3) there is a protective layer over the enteric release coating.
In attempting to provide for enteric release of an amphetamine salt, applicants found that use of an enteric release coating as generally practiced in the art did not provide effective enteric release.
Typical enteric coating levels did not meet the above requirements for the desired dosage profile of amphetamine salts. Using the typical amount of enteric coating (10-20μ) resulted in undesired premature leakage of the drug from the delivery system into the upper gastrointestinal tract and thus no drug delivery at the desired location in the gastrointestinal tract after the appropriate lag time. Thus this coating did not meet the requirements for the drug release profile to provide full beneficial therapeutic activity at the desired time.
Surprisingly, applicants found that using a thicker application of enteric coating on the formulation allowed for the second pulsed dose to be released only and completely at the appropriate time in the desired predetermined area of the gastrointestinal tract, i.e., in the intestine.
This was surprising because an increase in thickness of about 5-10μ of enteric coatings above a minimum thickness of about 10-20μ typically does not have a significant effect on release of drug from within such coatings. Enteric coatings typically are pH dependent and will only dissolve/disperse when exposed to the appropriate environment. Typically, application of a thicker coating (greater than 20μ) will only marginally increase the time for complete release at the appropriate environmental condition i.e., for a brief period of time (20 minutes). Using the typical coating, applicants could not achieve the desired result—rather, the coating leaked before the predetermined time in an inappropriate environment resulting in significant loss of the therapeutic agent.
Accordingly, in one aspect, the pulsed enteric release of the amphetamine salts is accomplished by employing a certain minimum thickness of the enteric coating.
In one embodiment of the invention, the pulsed dose delivery comprises a composition which comprises one or more pharmaceutically active amphetamine salts; an enteric coating over the one or more pharmaceutically active amphetamine salts, wherein the thickness of the enteric coating layer is at least 25μ; a further layer of one or more pharmaceutically active amphetamine salts over the enteric coating layer; and an immediate release layer coating. The thicker enteric coating surprisingly provides the required delayed immediate release of the pharmaceutically active amphetamine salt at the desired time in the desired area of the gastrointestinal tract. FIG. 2 illustrates a model of this delivery system.
In this aspect, the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed around which the enteric coating is applied. Alternatively, a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
It has further been discovered that a delayed immediate release drug delivery can also be accomplished by coating the drug first with a protective layer prior to applying the enteric coating.
Thus, in another embodiment, the pulsed enteric release is accomplished by employing a protective layer between the drug and the enteric coating. When using a protective coating, the enteric coating may be of an increased thickness or may be of lower thickness.
Thus, in another aspect, the object of the invention is met by providing a composition comprising one or more pharmaceutically active amphetamine salts; a protective layer coating over the one or more pharmaceutically active amphetamine salt layer(s), and an enteric coating layer over the protective coating layer; a further pharmaceutically active amphetamine salt layer and an immediate release layer coating. In a preferred embodiment of this aspect, the thickness of the enteric coating is at least 25μ, and the protective layer comprises an immediate release coating.
With respect to this embodiment of the invention, the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed, during the core seed manufacturing process, around which the protective coating is applied. Alternatively, a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
In another embodiment, the pulsed enteric release is accomplished by employing a protective layer over the enteric coating.
Accordingly, in this embodiment of the present invention, there is provided a pulsed dose release drug delivery system comprising one or more pharmaceutically active amphetamine salts; an enteric coating layer over the pharmaceutically active amphetamine salt layer(s); and a protective layer over the enteric coating; a second pharmaceutically active amphetamine salt layer; and an immediate release layer coating.
In one aspect of this embodiment, the protective layer is comprised of one or more components, which includes an immediate release layer and a modifying layer. The modifying layer is preferably comprised of a semi water-permeable polymer. Applicants have surprisingly found that a semi-permeable polymer coating used in combination with an immediate release layer coating provided a delayed pulsed release drug delivery profile when layered over the enteric coating.
Thus, in this embodiment, the protective layer comprises a semi-permeable polymer and an immediate release coating layer. In a preferred embodiment, the modifying layer comprises a first layer of a semi-permeable polymer which is adjacent to the enteric coating layer and a second coating layer over the semi-permeable polymer coating layer comprising an immediate release polymer coating layer.
In one aspect of this embodiment, a semi-permeable polymer, which may comprise a low water-permeable pH-insensitive polymer, is layered onto the outer surface of the enteric layer, in order to obtain prolonged delayed release time. This semi-permeable polymer coating controls the erosion of the pH-sensitive enteric polymer in an alkaline pH environment in which a pH-sensitive polymer will dissolve rapidly. Another pH-sensitive layer may be applied onto the surface of a low water-permeability layer to further delay the release time.
In a still further aspect of the invention, in addition to a protective layer, the composition comprises an acid which is incorporated into the pharmaceutical active layer or coated onto the surface of the active layer to reduce the pH value of the environment around the enteric polymer layer. The acid layer may also be applied on the outer layer of the pH-sensitive enteric polymer layer, followed by a layer of low water-permeability polymer. The release of the active thus may be delayed and the dissolution rate may be increased in an alkaline environment.
In a further embodiment, the protective coating may be used both over the drug and over the enteric coating.
With respect of this embodiment of the invention, the one or more pharmaceutically active amphetamine salts can be provided within or as a part of a core seed, during the core seed manufacturing process, around which the enteric coating is applied. Alternatively, a core seed can be coated with one or more layers of one or more pharmaceutically active amphetamine salts.
The drug delivery system of the present invention as described herein preferably comprises one or a number of beads or beadlets in a dosage form, either capsule, tablet, sachet or other method of orally administering the beads.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrates a multiple pulse drug delivery system target plasma profile of the drug delivery system of the present invention. The profile reflects an immediate-release component followed by a delayed-release component.
FIG. 2a graphically illustrates a pulsed dose delivery system.
FIGS. 2b and c graphically illustrate the drug release mechanism from the proposed delivery system.
FIG. 3 is a plot of the present drug released versus time from the drug-loaded pellets described in Example 1 which exemplifies the immediate release component of the present invention.
FIG. 4 is a plot of the percent drug released versus time from the coated pellets described in Example 2 which exemplifies the immediate release component and the delayed release components of the present invention.
FIG. 5 is a plot of the percent drug released versus time from the coated pellets described in Example 3 which exemplifies the immediate release component and the delayed release components of the present invention.
FIG. 6 illustrates the drug release profile of coated pellets described in Example 4 which exemplifies the immediate release component and the delayed release components of the present invention.
FIG. 7 is a plot of a profile of plasma amphetamine concentration after administration of a composite capsule containing the immediate release pellets and delayed release pellets from Examples 1 and 2, respectively.
FIG. 8 is a plot of a profile of plasma amphetamine concentration after administration of a composite capsule containing the immediate release pellets and delayed release pellets from Examples 1 and 3, respectively.
DETAILED DESCRIPTION OF THE INVENTION
The present invention comprises a core or starting seed, either prepared or commercially available product. The cores or starting seeds can be sugar spheres; spheres made from microcrystalline cellulose and any suitable drug crystals.
The materials that can be employed in making drug-containing pellets are any of those commonly used in pharmaceutics and should be selected on the basis of compatibility with the active drug and the physicochemical properties of the pellets. The additives except active drugs are chosen below as examples.
Binders such as cellulose derivatives such as methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer and the like.
Disintegration agents such as corn starch, pregelatinized starch, cross-linked carboxymethylcellulose (AC-DI-SOL®), sodium starch glycolate (EXPLOTAB®), cross-linked polyvinylpyrrolidone (PLASDONE XL®), and any disintegration agents used in tablet preparations.
Filling agents such as lactose, calcium carbonate, calcium phosphate, calcium sulfate, microcrystalline cellulose, dextran, starches, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
Surfactants such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, bile salts, glyceryl monostearate, PLURONIC® line (BASF), and the like.
Solubilizers such as citric acid, succinic acid, fumaric acid, malic acid, tartaric acid, maleic acid, glutaric acid sodium bicarbonate and sodium carbonate and the like.
Stabilizers such as any antioxidation agents, buffers, acids, and the like, can also be utilized.
Methods of manufacturing the core include
    • a. Extrusion-Spheronization—Drug(s) and other additives are granulated by addition of a binder solution. The wet mass is passed through an extruder equipped with a certain size screen. The extrudates are spheronized in a marumerizer. The resulting pellets are dried and sieved for further applications.
    • b. High-Shear Granulation—Drug(s) and other additives are dry-mixed and then the mixture is wetted by addition of a binder solution in a high shear-granulator/mixer. The granules are kneaded after wetting by the combined actions of mixing and milling. The resulting granules or pellets are dried and sieved for further applications.
    • c. Solution or Suspension Layering—A drug solution or dispersion with or without a binder is sprayed onto starting seeds with a certain particle size in a fluid bed processor or other suitable equipment. The drug thus is coated on the surface of the starting seeds. The drug-loaded pellets are dried for further applications.
For purposes of the present invention, the core particles have a diameter in the range of about 50-1500 microns; preferably 100-800 microns.
These particles can then be coated in a fluidized bed apparatus with an alternating sequence of coating layers.
The core may be coated directly with a layer or layers of at least one pharmaceutically active amphetamine salts and/or the pharmaceutically active amphetamine salt may be incorporated into the core material. Pharmaceutical active amphetamine salts contemplated to be within the scope of the present invention include amphetamine base, all chemical and chiral derivatives and salts thereof; methylphenidate, all chemical and chiral derivatives and salts thereof; phenylpropanolamine and its salts; and all other compounds indicated for the treatment of attention deficit hyperactivity disorder (ADHD).
A protective layer may be added on top of the pharmaceutical active containing layer and also may be provided between active layers. A separation or protective layer may be added onto the surface of the active-loaded core, and then the enteric layer is coated thereupon. Another active layer may also be added to the enteric layer to deliver an initial dose.
A protective coating layer may be applied immediately outside the core, either a drug-containing core or a drug-layered core, by conventional coating techniques such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. Suitable materials for the protective layer include cellulose derivatives such as hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, ethyl cellulose aqueous dispersions (AQUACOAT®, SURELEASE®), EUDRAGIT® RL 30D, OPADRY® and the like. The suggested coating levels are from 1 to 6%, preferably 2-4% (w/w).
The enteric coating layer is applied onto the cores with or without seal coating by conventional coating techniques, such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. All commercially available pH-sensitive polymers are included. The pharmaceutical active is not released in the acidic stomach environment of approximately below pH 4.5, but not limited to this value. The pharmaceutical active should become available when the pH-sensitive layer dissolves at the greater pH, after a certain delayed time; or after the unit passes through the stomach. The preferred delay time is in the range of two to six hours.
Enteric polymers include cellulose acetate phthalate, Cellulose acetate trimellitate, hydroxypropyl methylcellulose phtalate, polyvinyl acetate phtalate, carboxymethylethylcellulose, co-polymerized methacrylic acid/methacrylic acid methyl esters such as, for instance, materials known under the trade name EUDRAGIT® L12.5, L100, or EUDRAGIT® S12.5, S100 or similar compounds used to obtain enteric coatings. Aqueous collodial polymer dispersions or re-dispersions can be also applied, e.g. EUDRAGIT® L 30D-55, EUDRAGIT® L100-55, EUDRAGIT® S100, EUDRAGIT® preparation 4110D (Rohm Pharma); AQUATERIC®, AQUACOAT® CPD 30 (FMC); KOLLICOAT MAE® 30D and 30DP (BASF); EASTACRYL® 30D (Eastman Chemical).
The enteric polymers used in this invention can be modified by mixing with other known coating products that are not pH sensitive. Examples of such coating pro ducts include the neutral methacrylic acid esters with a small portion of trimethylammonioethyl methacrylate chloride, sold currently under the trade names EUDRAGIT® RS and EUDRAGIT® RL; a neutral ester dispersion without any functional groups, sold under the trade names EUDRAGIT® NE30D; and other pH independent coating products.
The modifying component of the protective layer used over the enteric coating can include a water penetration barrier layer (semipermeable polymer) which can be successively coated after the enteric coating to reduce the water penetration rate through the enteric coating layer and thus increase the lag time of the drug release. Sustained-release coatings commonly known to one skilled in the art can be used for this purpose by conventional coating techniques such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. For example, the following materials can be used, but not limited to: Cellulose acetate, Cellulose acetate butyrate, Cellulose acetate propionate, Ethyl cellulose, Fatty acids and their esters, Waxes, zein, and aqueous polymer dispersions such as EUDRAGIT® RS and SURELEASE®, cellulose acetate latex. The combination of above polymers and hydrophilic polymers such as Hydroxyethyl cellulose, Hydroxypropyl cellulose (KLUCEL®, Hercules Corp.), Hydroxypropyl methylcellulose (METHOCEL®, Dow Chemical Corp.), Polyvinylpyrrolidone can also be used.
An overcoating layer can further optionally be applied to the composition of the present invention. OPADRY®, OPADRY II® (Colorcon) and corresponding color and colorless grades from Colorcon can be used to protect the pellets from being tacky and provide colors to the product. The suggested levels of protective or color coating are from 1 to 6%, preferably 2-3% (w/w).
Many ingredients can be incorporated into the overcoating formula, for example to provide a quicker immediate release, such as plasticizers: acetyltriethyl citrate, triethyl citrate, acetyltributyl citrate; dibutylsebacate, triacetin, polyethylene glycols, propylene glycol and the others; lubricants: talc, colloidal silica dioxide, magnesium stearate, calcium stearate, titanium dioxide, magnesium silicate, and the like.
The composition, preferably in beadlet form, can be incorporated into hard gelatin capsules, either with additional excipients, or alone. Typical excipients to be added to a capsule formulation include, but are not limited to: filters such as microcrystalline cellulose, soy polysaccharides, calcium phosphate dihydrate, calcium sulfate, lactose, sucrose, sorbitol, or any other inert filler. In addition, there can be flow aids such as fumed silicon dioxide, silica gel, magnesium stearate, calcium stearate or any other material imparting flow to powders. A lubricant can further be added if necessary by using polyethylene glycol, leucine, glyceryl behenate, magnesium stearate or calcium stearate.
The composition may also be incorporated into a tablet, in particular by incorporation into a tablet matrix, which rapidly disperses the particles after ingestion. In order to incorporate these particles into such a tablet, a filler/binder must be added to a table that can accept the particles, but will not allow their destruction during the tableting process. Materials that are suitable for this purpose include, but are not limited to, microcrystalline cellulose (AVICEL®), soy polysaccharide (EMCOSOY®), pre-gelatinized starches (STARCH® 1500, NATIONAL® 1551), and polyethylene glycols (CARBOWAX®). The materials should be present in the range of 5-75% (w/w), with a preferred range of 25-50% (w/w).
In addition, disintegrants are added in order to dispense the beads once the tablet is ingested. Suitable disintegrants include, but are not limited to: cross-linked sodium carboxymethyl cellulose (AC-DI-SOL®), sodium starch glycolate (EXPLOTAB®, PRIMOJEL®), and cross-linked polyvinylpolypyrrolidone (Plasone-XL). These materials should be present in the rate of 3-15% (w/w), with a preferred range of 5-10% (w/w).
Lubricants are also added to assure proper tableting, and these can include, but are not limited to: magnesium stearate, calcium stearate, stearic acid, polyethylene glycol, leucine, glyceryl behanate, and hydrogenated vegetable oil. These lubricants should be present in amounts from 0.1-10% (w/w), with a preferred range of 0.3-3.0% (w/w).
Tablets are formed, for example, as follows. The particles are introduced into a blender along with AVICEL®, disintegrants and lubricant, mixed for a set number of minutes to provide a homogeneous blend which is then put in the hopper of a tablet press with which tablets are compressed. The compression force used is adequate to form a tablet; however, not sufficient to fracture the beads or coatings.
It will be appreciated that the multiple dosage form of the present invention can deliver rapid and complete dosages of pharmaceutically active amphetamine salts to achieve the desired levels of the drug in a recipient over the course of about 8 hours with a single oral administration.
In so doing, the levels of drug in blood plasma of the pharmaceutically active amphetamine salts will reach a peak fairly rapidly after about 2 hours, and after about 4 hours a second pulse dose is released, wherein a second fairly rapid additive increase of plasma drug levels occurs which slowly decreases over the course of the next 12 hours.
The following examples are presented to illustrate and do not limit the invention.
EXAMPLES Example 1 Immediate Release Formulation
The following formulation was used to layer the drug onto sugar spheres. Nonpareil seeds (30/35 mesh, Paulaur Corp., NJ), 6.8 kg were put into a FLM-15 fluid bed processor with a 9″ Wurster column and fluidized at 60° C. The suspension of mixed amphetamine salts (MAS) with 1% HPMC E5 Premium (Dow Chemical) as a binder was sprayed onto the seed under suitable conditions. Almost no agglomeration and no fines were observed with a yield of at least 98%. The drug-loaded cores were used to test enteric coatings and sustained release coatings.
TABLE 1
Ingredients Amount (%)
Nonpareil seed 88.00
mixed amphetamine salts 11.40
METHOCEL ® E5 Premium 0.60
Water *
*removed during processing
The drug release profile of the drug-loaded pellets of this example is shown in FIG. 3.
Example 2
The following formulation was used to coat the mixed amphetamine salts loaded (MASL) pellets from Example 1 with the EUDRAGIT® L 30D-55 (Rohm Pharma, Germany) coating dispersion. 2 kg of MASL pellets were loaded into a fluid bed processor with a reduced Wurster column equipped with a precision coater (MP 2/3, Niro Inc) (see Examples 3 and 4). The coating dispersion was prepared by dispersing Triethyl citrate, Talc and EUDRAGIT® L 30D-55 into water and mixing for at least 30 minutes. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidized MASL pellets. The spraying was continued until the targeted coating level was achieved (20 μ) . The coated pellets were dried at 30-35° C. for 5 minutes before stopping the process. The enteric coated PPA MASL pellets were tested at different pH buffers by a USP paddle method. The drug content was analyzed using HPLC. The results showed that the enteric coating delayed the drug release from the coated pellets until after exposure to pH 6 or higher. (Reference #AR98125-4)
TABLE 2
Ingredients Amount (%)
MASL pellets 40.0070.00
EUDRAGIT ® L 30D-55 24.88
Triethyl citrate 2.52
Talc 2.60
Water *
*removed during processing
The drug release profile of the coated pellets of this example is shown in FIG. 4.
Example 3
The following formulation was used to coat the MASL pellets from Example 1 with the EUDRAGIT® 4110D (Rohm Pharma, Germany) coating dispersion. MASL pellets (2 kg) were loaded in a fluid bed processor with a reduced Wurster column (GPGC-15, Glatt). The coating dispersion was prepared by dispersing Triethyl citrate, Talc and EUDRAGIT® 4110D into water and mixing for at least 30 minutes. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidized MASL pellets. The spraying was continued until the targeted coating level was achieved. The coated pellets were dried at 30-35° C. for 5 minutes before stopping the process. The enteric coated MASL pellets were tested using a USP paddle method at different pH buffers. The drug content was analyzed using HPLC. The enteric coating delayed the drug release for several hours from the coated pellets until the pH value reached 6.8 or higher, as shown below in Table 3. (Reference #AR98125-3)
TABLE 3
Ingredients Amount (%)
MASL pellets 70.00
EUDRAGIT ® 4110D 26.24
Triethyl citrate 0.76
Talc 3.00
Water *
*removed during processing
The drug release profile of coated pellets of this example is shown in FIG. 5.
Example 4
The following formulation was selected to coat the enteric coated MASL pellets. Coated MASL pellets from Example 2 or coated MASL pellets from Example 3 (2 kg of either) were loaded into a fluid bed processor with a reduced Wurster column (GPGC-15, Glatt). The coating dispersion was prepared by mixing SURELEASE® (Colorcon) and water for at least 15 minutes prior to spraying. Under suitable fluidization conditions, the coating dispersion was sprayed onto the fluidizated pellets. The spraying was continued until the targeted coating level was achieved. The coated pellets were coated with a thin layer of OPADRY® white (Colorcon) (2%) to prevent the tackiness of the coated pellets during storage. The coated pellets were then dried at 35-40° C. for 10 minutes before discharging from the bed. The drug dissolution from both coated pellets was performed using a USP paddle method at different pH buffers. The drug content was analyzed using HPLC. The 8% SURELEASE® coating slightly sustained the drug release from EUDRAGIT® L 30D-55 coated pellets at pH 7.5 buffer, while the SURELEASE® coating delayed the drug release up to 2 hours after the buffer switched from pH 1 to pH 7.5. (Reference ##AR98125-1)
TABLE 4
Ingredients Amount, (%)
Enteric coated MASL pellets 90.00
SURELEASE ® 8.00
OPADRY ® white 2.00
Water *
*removed during processing
The drug release profile of the coated pellets from this example is shown in FIG. 6.
Example 5
A pulsatile delivery system can be achieved by combining the immediate release pellets (Example 1) with delayed release pellets (Example 2 or Example 3). The immediate-release pellets equivalent to half the dose and the delayed-release pellets equivalent to half the dose are filled into a hard gelatin capsule to produce the oral pulsed dose delivery system. The delayed-release portion releases the amphetamine salts rapidly and completely, after a specified lag time. The capsule products containing immediate-release pellets and delayed-release pellets (Example 1 plus Example 2 and Example 1 plus Example 3) were tested in a crossover human study. FIGS. 7 and 8 show the typical profiles of plasma amphetamine concentration after administration of a composite capsule containing the immediate-release pellets and delayed-release pellets from Examples 1 and 2 (10 mg dose each pellet type) and a capsule containing the pellets from immediate-release pellets and delayed-release pellets from Examples 1 and 3 (10 mg dose each pellet type), respectively. The general plasma profiles are similar to the desired target plasma level profile shown in FIG. 1.
It is to be understood, however, that the scope of the present invention is not to be limited to the specific embodiments described above. The invention may be practiced other than as particularly described and still be within the scope of the accompanying claims.
CITED LITERATURE
  • 1. B. Lemmer, “Circadian Rhythms and Drug Delivery”, J. Controlled Release, 16, 63-74 (1991)
  • 2. B. Lemmer, “Why are so may Biological Systems Periodic?” in Pulsatile Drug Delivery: Current Applications and Future Trends, R Gurny, H E Junginger and N A Peppas, eds. (Wissenschaftliche Verlagsgesellschaft mbH Stuttgart, Germany 1993) pp.11-24
  • 3. X. Xu and P I Lee, “Programmable Drug Delivery from an Erodible Association Polymer System”, Pharm. Res. 10(8), 1144-1152 (1993)
  • 4. A. Gazzaniga, M E Sangalli, and F Giodano, “Oral Chonotropic Drug Delivery Systems: Achievement of Time and/or Site Specificity”, Eur J. Pharm. Biopharm., 40(4), 246-250 (1994)
  • 5. A Gazzaniga, C Busetti, L Moro, M E Sangalli and F Giordano, “Time Dependent Oral Delivery Systems for Colon Targeting”, S.T.P. Pharma Sciences 5(1), 83-88 (1996)
  • 6. U Conte, L Maggi, M L Torre, P Giunchedi and A Lamanna, “Press-coated Tablets for Time programmed Release of Drugs”, Biomaterials, 14(13), 1017-1023 (1993)
  • 7. A C Shah International Patent Application WO87/00044
  • 8. P S Walia, P Jo Mayer Stout and R Turton, “Preliminary Evaluation of an Aqueous Wax Emulsion for Controlled Release Coating”,Pharm Dev Tech, 3(1), 103-113 (1998)
  • 9. F Theeuwes, “OROS® Osmotic System Development”, Drug Dev Ind Pharm 9(7), 1331-1357 (1983)
  • 10. F Theeuwes, “Triggered Pulsed and Programmed Drug Delivery” in Novel Drug Delivery and its Therapeutic Application, L F Prescott and W S Nimmos, eds (Wiley, New York, 1989) pp.323-340
  • 11. M McNeil, A Rashid and H Stevens, International Patent App WO90/09168
  • 12. I R Wilding, S S Davis, M Bakhshaee, H N E Stevens, R A Sparrow and J Brennan, “Gastrointestinal Transit and Systemic Absorption of Captopril from a Pulsed Release Formulation”, Pharm Res 9(5), 654-657 (1992)

Claims (20)

1. A pharmaceutical formulation for delivery of a mixture of amphetamine base salts effective to treat ADHD in a human patient comprising:
an immediate release dosage form that provides immediate release upon oral administration to said patient;
a delayed enteric release dosage form that provides delayed release upon oral administration to said patient; and
a pharmaceutically acceptable carrier;
wherein said amphetamine base salts comprise dextroamphetamine sulfate, dextroamphetamine saccharate, amphetamine aspartate monohydrate and amphetamine sulfate;
wherein said pharmaceutical formulation is sufficient to maintain an effective level of amphetamine base salts in the patient over the course of at least 8 hours without further administration of amphetamine base salt, and the peak plasma concentration of amphetamine base salts reached after release of said delayed enteric release dosage form exceeds the peak plasma concentration previously reached after release of said immediate release dosage form; and
wherein said pharmaceutical formulation, when containing about a total dose of 20 mg, will produce in a human individual a plasma concentration versus time curve (ng/ml versus hours) having an area under the curve (AUC) of about 467 to about 714 ng hr/ml.
2. A formulation of claim 1 wherein said plasma concentration curve has a maximum concentration (Cmax) of about 22.5 to about 40 ng/ml for about a total dose of 20 mg.
3. A formulation of claim 2 wherein the time after said oral administration to reach said Cmax value is about 7 to about 10 hours.
4. A formulation of claim 1 wherein the time after said oral administration to reach maximum concentration of said plasma concentration curve is about 7 to about 10 hours.
5. A formulation of claim 2, 3 or 4 wherein said AUC is about 714 ng hr/ml.
6. A formulation of claim 3 wherein said AUC is about 714 ng hr/ml, the time after said oral administration to reach said Cmax value is about 7 hours and Cmax is about 40 ng/ml.
7. A formulation of claim 2 wherein Cmax is about 40 ng/ml.
8. A formulation of claim 3 or 4 wherein said time is about 7 hours.
9. A formulation of one of claims 1-4, 6 or 7 wherein said salts are contained in about equal amounts within each of said dosage forms.
10. A formulation of one of claims 1-4, 6 or 7 wherein said delayed enteric release dosage form comprises a coating of a thickness of at least greater than 20 μm which comprises dried about 30% (dry substance) aqueous dispersion of an anionic copolymer based on methacrylic acid and acrylic acid ethyl ester, said coating being soluble at a pH of about 5.5 upwards.
11. A formulation of claim 10 wherein said thickness is at least 25 μm.
12. A pharmaceutical formulation for delivery of a mixture of amphetamine base salts effective to treat ADHD in a human patient comprising:
an immediate release dosage form that provides immediate release upon oral administration to said patient;
a delayed enteric release dosage form that provides delayed release upon oral administration to said patient, wherein said enteric release dosage form comprises a coating of a thickness of at least greater than 20 μm which comprises dried aqueous dispersion of an anionic copolymer based on methacrylic acid and acrylic acid ethyl ester, said coating being soluble at a pH of about 5.5 upwards; and
a pharmaceutically acceptable carrier;
wherein said amphetamine base salts comprise dextroamphetamine sulfate, dextroamphetamine saccharate, amphetamine aspartate monohydrate and amphetamine sulfate;
wherein said pharmaceutical formulation is sufficient to maintain an effective level of amphetamine base salts in the patient over the course of at least 8 hours without further administration of amphetamine base salt, and the peak plasma concentration of amphetamine base salts reached after release of said delayed enteric release dosage form exceeds the peak plasma concentration of said salts previously reached after release of said immediate release dosage form.
13. A formulation of claim 12 wherein said thickness is at least 25 μm.
14. A formulation of claim 12, wherein said dried aqueous dispersion of an anionic copolymer is a dried about 30% (dry substance) aqueous dispersion of an anionic copolymer.
15. A formulation of claim 1 formulated for a total dose of 20 mg.
16. A formulation of claim 2 formulated for a total dose of 20 mg.
17. A formulation of claim 1 formulated for a total dose different from about 20 mg and having an AUC proportional to said 20 mg AUC.
18. A formulation of claim 2 formulated for a total dose different from about 20 mg and having a Cmax proportional to said 20 mg Cmax.
19. The pharmaceutical formulation of claim 1, wherein the delayed release is pH independent.
20. The pharmaceutical formulation of claim 1, which further comprises a protective coating layer.
US11/091,010 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system Expired - Lifetime USRE41148E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/091,010 USRE41148E1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/176,542 US6322819B1 (en) 1998-10-21 1998-10-21 Oral pulsed dose drug delivery system
US11/091,010 USRE41148E1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system
PCT/US1999/024554 WO2000023055A1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/807,462 Reissue US6605300B1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system

Publications (1)

Publication Number Publication Date
USRE41148E1 true USRE41148E1 (en) 2010-02-23

Family

ID=22644775

Family Applications (7)

Application Number Title Priority Date Filing Date
US09/176,542 Ceased US6322819B1 (en) 1998-10-21 1998-10-21 Oral pulsed dose drug delivery system
US09/176,542 Granted US20010055613A1 (en) 1998-10-21 1998-10-21 Oral pulsed dose drug delivery system
US09/807,462 Ceased US6605300B1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system
US11/091,010 Expired - Lifetime USRE41148E1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system
US10/172,705 Abandoned US20030124188A1 (en) 1998-10-21 2002-06-14 Oral pulsed dose drug delivery system
US10/758,417 Abandoned US20040219213A1 (en) 1998-10-21 2004-01-16 Oral pulsed dose drug delivery system
US11/091,011 Expired - Lifetime USRE42096E1 (en) 1998-10-21 2005-03-24 Oral pulsed dose drug delivery system

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/176,542 Ceased US6322819B1 (en) 1998-10-21 1998-10-21 Oral pulsed dose drug delivery system
US09/176,542 Granted US20010055613A1 (en) 1998-10-21 1998-10-21 Oral pulsed dose drug delivery system
US09/807,462 Ceased US6605300B1 (en) 1998-10-21 1999-10-20 Oral pulsed dose drug delivery system

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/172,705 Abandoned US20030124188A1 (en) 1998-10-21 2002-06-14 Oral pulsed dose drug delivery system
US10/758,417 Abandoned US20040219213A1 (en) 1998-10-21 2004-01-16 Oral pulsed dose drug delivery system
US11/091,011 Expired - Lifetime USRE42096E1 (en) 1998-10-21 2005-03-24 Oral pulsed dose drug delivery system

Country Status (10)

Country Link
US (7) US6322819B1 (en)
EP (2) EP1977736A1 (en)
JP (2) JP2002527468A (en)
AT (1) ATE427101T1 (en)
AU (1) AU1214500A (en)
CA (1) CA2348090C (en)
DE (1) DE69940673D1 (en)
ES (1) ES2323910T3 (en)
HK (1) HK1038701A1 (en)
WO (1) WO2000023055A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070264323A1 (en) * 2006-05-12 2007-11-15 Shire Llc Controlled dose drug delivery system
US20090019148A1 (en) * 2007-07-13 2009-01-15 Britton Zachary E Method and apparatus for internet traffic monitoring by third parties using monitoring implements
US20090157875A1 (en) * 2007-07-13 2009-06-18 Zachary Edward Britton Method and apparatus for asymmetric internet traffic monitoring by third parties using monitoring implements
US20100024032A1 (en) * 2008-07-24 2010-01-28 Zachary Edward Britton Method and apparatus for effecting an internet user's privacy directive
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US9545399B2 (en) 2012-08-15 2017-01-17 Tris Pharma, Inc. Methylphenidate extended release chewable tablet
US9675704B2 (en) 2006-03-16 2017-06-13 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US9974752B2 (en) 2014-10-31 2018-05-22 Purdue Pharma Methods and compositions particularly for treatment of attention deficit disorder
US10142086B2 (en) 2015-06-11 2018-11-27 At&T Intellectual Property I, L.P. Repeater and methods for use therewith
US10722473B2 (en) 2018-11-19 2020-07-28 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US11590228B1 (en) 2015-09-08 2023-02-28 Tris Pharma, Inc Extended release amphetamine compositions
US11590081B1 (en) 2017-09-24 2023-02-28 Tris Pharma, Inc Extended release amphetamine tablets

Families Citing this family (199)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6322819B1 (en) * 1998-10-21 2001-11-27 Shire Laboratories, Inc. Oral pulsed dose drug delivery system
JP4613275B2 (en) 1998-11-02 2011-01-12 エラン ファーマ インターナショナル,リミティド Multiparticulate modified release composition
US20060240105A1 (en) * 1998-11-02 2006-10-26 Elan Corporation, Plc Multiparticulate modified release composition
DE19855421C2 (en) * 1998-11-02 2001-09-20 Alcove Surfaces Gmbh Implant
US20090149479A1 (en) * 1998-11-02 2009-06-11 Elan Pharma International Limited Dosing regimen
US20030170181A1 (en) * 1999-04-06 2003-09-11 Midha Kamal K. Method for preventing abuse of methylphenidate
ATE279186T1 (en) * 1999-08-31 2004-10-15 Gruenenthal Gmbh SUSTAINED-RELEASE PHARMACEUTICAL FORM CONTAINING TRAMADOL ACCHARINATE
EP2269587A1 (en) 1999-10-29 2011-01-05 Euro-Celtique S.A. Controlled release hydrocodone formulations
US10179130B2 (en) 1999-10-29 2019-01-15 Purdue Pharma L.P. Controlled release hydrocodone formulations
AU1789301A (en) * 1999-11-22 2001-06-04 Alza Corporation Osmotic dosage form comprising first and second
DK1248594T3 (en) * 2000-01-19 2006-02-06 Mannkind Corp Multi-tip release formulation for drug delivery
US6544555B2 (en) 2000-02-24 2003-04-08 Advancis Pharmaceutical Corp. Antibiotic product, use and formulation thereof
US7674480B2 (en) * 2000-06-23 2010-03-09 Teva Pharmaceutical Industries Ltd. Rapidly expanding composition for gastric retention and controlled release of therapeutic agents, and dosage forms including the composition
US6500457B1 (en) * 2000-08-14 2002-12-31 Peirce Management, Llc Oral pharmaceutical dosage forms for pulsatile delivery of an antiarrhythmic agent
US20020068078A1 (en) 2000-10-13 2002-06-06 Rudnic Edward M. Antifungal product, use and formulation thereof
AU2738302A (en) 2000-10-30 2002-05-15 Euro Celtique Sa Controlled release hydrocodone formulations
CA2440641A1 (en) 2001-03-13 2002-09-19 Anand R. Baichwal Chronotherapeutic dosage forms containing glucocorticosteroid
EP2311440A1 (en) * 2001-04-05 2011-04-20 Collagenex Pharmaceuticals, Inc. Controlled delivery of tetracycline compounds and tetracycline derivatives
US20020187192A1 (en) * 2001-04-30 2002-12-12 Yatindra Joshi Pharmaceutical composition which reduces or eliminates drug abuse potential
AR034813A1 (en) * 2001-07-20 2004-03-17 Novartis Ag PHARMACEUTICAL COMPOSITIONS AND USE OF THE SAME
US6837696B2 (en) * 2001-09-28 2005-01-04 Mcneil-Ppc, Inc. Apparatus for manufacturing dosage forms
US20040146559A1 (en) * 2002-09-28 2004-07-29 Sowden Harry S. Dosage forms having an inner core and outer shell with different shapes
US20030228368A1 (en) * 2001-09-28 2003-12-11 David Wynn Edible solid composition and dosage form
JP2005508325A (en) 2001-09-28 2005-03-31 マクニール−ピーピーシー・インコーポレイテッド Dosage form having an inner core and an outer shell
US20040253312A1 (en) * 2001-09-28 2004-12-16 Sowden Harry S. Immediate release dosage form comprising shell having openings therein
US7122143B2 (en) 2001-09-28 2006-10-17 Mcneil-Ppc, Inc. Methods for manufacturing dosage forms
US7838026B2 (en) 2001-09-28 2010-11-23 Mcneil-Ppc, Inc. Burst-release polymer composition and dosage forms comprising the same
US7776314B2 (en) 2002-06-17 2010-08-17 Grunenthal Gmbh Abuse-proofed dosage system
PA8578501A1 (en) 2002-07-25 2005-02-04 Pharmacia Corp DOSAGE FORM ONCE A DAY OF PRAMIPEXOL
US6913768B2 (en) * 2002-09-24 2005-07-05 Shire Laboratories, Inc. Sustained release delivery of amphetamine salts
US20040062778A1 (en) * 2002-09-26 2004-04-01 Adi Shefer Surface dissolution and/or bulk erosion controlled release compositions and devices
US7807197B2 (en) 2002-09-28 2010-10-05 Mcneil-Ppc, Inc. Composite dosage forms having an inlaid portion
US20040191302A1 (en) 2003-03-28 2004-09-30 Davidson Robert S. Method and apparatus for minimizing heat, moisture, and shear damage to medicants and other compositions during incorporation of same with edible films
US8999372B2 (en) * 2002-11-14 2015-04-07 Cure Pharmaceutical Corporation Methods for modulating dissolution, bioavailability, bioequivalence and drug delivery profile of thin film drug delivery systems, controlled-release thin film dosage formats, and methods for their manufacture and use
US20040131662A1 (en) 2003-11-12 2004-07-08 Davidson Robert S. Method and apparatus for minimizing heat, moisture, and shear damage to medicants and other compositions during incorporation of same with edible films
US7988993B2 (en) 2002-12-09 2011-08-02 Andrx Pharmaceuticals, Inc. Oral controlled release dosage form
JP4865330B2 (en) 2002-12-13 2012-02-01 デュレクト コーポレーション Oral drug delivery system
EP1572164A2 (en) * 2002-12-18 2005-09-14 Pain Therapeutics, Inc. Oral dosage forms with therapeutically active agents in controlled release cores and immediate release gelatin capsule coats
US20040208931A1 (en) * 2002-12-30 2004-10-21 Friend David R Fast dissolving films for oral administration of drugs
AU2004210671A1 (en) * 2003-02-10 2004-08-26 Shire Canada Inc. Enantiomeric amphetamine compositions for the treatment of ADHD
US20040186180A1 (en) 2003-03-21 2004-09-23 Gelotte Cathy K. Non-steroidal anti-inflammatory drug dosing regimen
PL1615622T3 (en) * 2003-04-07 2012-12-31 Supernus Pharmaceuticals Inc Once daily formulations of doxycyclines
CA2529984C (en) 2003-06-26 2012-09-25 Isa Odidi Oral multi-functional pharmaceutical capsule preparations of proton pump inhibitors
CA2533178C (en) 2003-07-21 2014-03-11 Advancis Pharmaceutical Corporation Antibiotic product, use and formulation thereof
AU2004258953B2 (en) 2003-07-21 2011-02-10 Shionogi, Inc. Antibiotic product, use and formulation thereof
EP1648418A4 (en) 2003-07-21 2011-11-16 Middlebrook Pharmaceuticals Inc Antibiotic product, use and formulation thereof
DE102005005446A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Break-resistant dosage forms with sustained release
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
DE10361596A1 (en) 2003-12-24 2005-09-29 Grünenthal GmbH Process for producing an anti-abuse dosage form
JP2007502296A (en) 2003-08-11 2007-02-08 アドバンシス ファーマスーティカル コーポレイション Robust pellet
WO2005016278A2 (en) 2003-08-12 2005-02-24 Advancis Pharmaceuticals Corporation Antibiotic product, use and formulation thereof
WO2005023184A2 (en) 2003-08-29 2005-03-17 Advancis Pharmaceuticals Corporation Antibiotic product, use and formulation thereof
WO2005027877A1 (en) 2003-09-15 2005-03-31 Advancis Pharmaceutical Corporation Antibiotic product, use and formulation thereof
KR101164881B1 (en) * 2003-09-19 2012-07-23 썬 파마슈티컬 인더스트리스 리미티드 Oral drug delivery system
US10213387B2 (en) 2003-09-19 2019-02-26 Sun Pharma Advanced Research Company Ltd. Oral drug delivery system
BRPI0414311A (en) 2003-09-19 2008-03-04 Penwest Pharmaceutical Co controlled release dosage forms
US10226428B2 (en) 2003-09-19 2019-03-12 Sun Pharma Advanced Research Company Ltd. Oral drug delivery system
US20050069590A1 (en) * 2003-09-30 2005-03-31 Buehler Gail K. Stable suspensions for medicinal dosages
US20050074514A1 (en) * 2003-10-02 2005-04-07 Anderson Oliver B. Zero cycle molding systems, methods and apparatuses for manufacturing dosage forms
ATE493981T1 (en) * 2003-11-04 2011-01-15 Supernus Pharmaceuticals Inc ONCE DAILY DOSAGE FORMS OF TROSPIUM
WO2005044238A1 (en) * 2003-11-07 2005-05-19 Ranbaxy Laboratories Limited Modified release solid dosage form of amphetamine salts
DE10353186A1 (en) * 2003-11-13 2005-06-16 Röhm GmbH & Co. KG Multilayer dosage form containing a modulatory substance in relation to the release of active ingredient
US8067029B2 (en) * 2004-01-13 2011-11-29 Mcneil-Ppc, Inc. Rapidly disintegrating gelatinous coated tablets
US7879354B2 (en) 2004-01-13 2011-02-01 Mcneil-Ppc, Inc. Rapidly disintegrating gelatinous coated tablets
JP2005239696A (en) * 2004-01-30 2005-09-08 Daiichi Suntory Pharma Co Ltd Medicinal hard capsule preparation blended with inorganic substance
US20050196448A1 (en) * 2004-03-05 2005-09-08 Hai Yong Huang Polymeric compositions and dosage forms comprising the same
US20050196442A1 (en) * 2004-03-05 2005-09-08 Huang Hai Y. Polymeric compositions and dosage forms comprising the same
US20050196447A1 (en) * 2004-03-05 2005-09-08 Huang Hai Y. Polymeric compositions and dosage forms comprising the same
US20050196446A1 (en) * 2004-03-05 2005-09-08 Huang Hai Y. Polymeric compositions and dosage forms comprising the same
US7712288B2 (en) 2004-05-28 2010-05-11 Narayanan Ramasubramanian Unified ingestion package and process for patient compliance with prescribed medication regimen
TWI354569B (en) * 2004-05-28 2011-12-21 Bristol Myers Squibb Co Coated tablet formulation and method
US20050265955A1 (en) * 2004-05-28 2005-12-01 Mallinckrodt Inc. Sustained release preparations
DE102004032049A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
US8394409B2 (en) 2004-07-01 2013-03-12 Intellipharmaceutics Corp. Controlled extended drug release technology
JP2008505124A (en) 2004-07-02 2008-02-21 アドバンシス ファーマスーティカル コーポレイション Pulse delivery tablets
MX2007001850A (en) 2004-08-13 2007-03-28 Boehringer Ingelheim Int Extended release pellet formulation containing pramipexole or a pharmaceutically acceptable salt thereof, method for manufacturing the same and use thereof.
KR101406767B1 (en) 2004-08-13 2014-06-20 베링거 인겔하임 인터내셔날 게엠베하 Extended release tablet formulation containing pramipexole or a pharmaceutically acceptable salt thereof, method for manufacturing the same and use thereof
US10624858B2 (en) 2004-08-23 2020-04-21 Intellipharmaceutics Corp Controlled release composition using transition coating, and method of preparing same
US9149472B2 (en) * 2004-08-31 2015-10-06 Jack William Schultz Controlled release compositions for treatment of cognitive, emotional and mental ailments and disorders
US7350479B2 (en) * 2004-09-10 2008-04-01 United States Of America As Represented By The Secretary Of The Army System and method for controlling growth of aquatic plants utilizing bio-eroding means implanted in triploid grass carp
EP1811975A2 (en) * 2004-10-19 2007-08-01 The State of Oregon Acting by and Enteric coated compositions that release active ingredient(s) in gastric fluid and intestinal fluid
US20060087051A1 (en) * 2004-10-27 2006-04-27 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US20070190133A1 (en) * 2004-10-27 2007-08-16 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US20060088593A1 (en) * 2004-10-27 2006-04-27 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US20070281022A1 (en) * 2004-10-27 2007-12-06 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US8383159B2 (en) * 2004-10-27 2013-02-26 Mcneil-Ppc, Inc. Dosage forms having a microreliefed surface and methods and apparatus for their production
US20060088587A1 (en) * 2004-10-27 2006-04-27 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US20060088586A1 (en) * 2004-10-27 2006-04-27 Bunick Frank J Dosage forms having a microreliefed surface and methods and apparatus for their production
US20060121112A1 (en) * 2004-12-08 2006-06-08 Elan Corporation, Plc Topiramate pharmaceutical composition
GB0427455D0 (en) * 2004-12-15 2005-01-19 Jagotec Ag Dosage forms
US8481565B2 (en) 2004-12-27 2013-07-09 Eisai R&D Management Co., Ltd. Method for stabilizing anti-dementia drug
DE102005005449A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Process for producing an anti-abuse dosage form
ITBO20050123A1 (en) * 2005-03-07 2005-06-06 Alfa Wassermann Spa GASTRORESISTIC PHARMACEUTICAL FORMULATIONS CONTAINING RIFAXIMINA
US20060204575A1 (en) * 2005-03-11 2006-09-14 Hengsheng Feng Amphetamine formulations
JP2008534530A (en) * 2005-03-29 2008-08-28 エボニック レーム ゲゼルシャフト ミット ベシュレンクテル ハフツング Multiparticulate drug form containing pellets with a substance having a modulating effect on active ingredient release
CA2601339A1 (en) * 2005-03-29 2006-10-05 Roehm Gmbh Multiparticulate pharmaceutical form comprising pellets with a matrix which influences the delivery of a modulatory substance
EP1874274A2 (en) * 2005-04-06 2008-01-09 Mallinckrodt Inc. Matrix-based pulse release pharmaceutical formulation
AU2006235483B2 (en) * 2005-04-12 2010-11-25 Elan Pharma International Limited Controlled release compositions comprising a cephalosporin for the treatment of a bacterial infection
US8673352B2 (en) 2005-04-15 2014-03-18 Mcneil-Ppc, Inc. Modified release dosage form
WO2006118265A1 (en) * 2005-04-28 2006-11-09 Eisai R & D Management Co., Ltd. Composition containing antidementia agent
US20070077300A1 (en) 2005-09-30 2007-04-05 Wynn David W Oral compositions containing a salivation inducing agent
US8778924B2 (en) 2006-12-04 2014-07-15 Shionogi Inc. Modified release amoxicillin products
US8357394B2 (en) 2005-12-08 2013-01-22 Shionogi Inc. Compositions and methods for improved efficacy of penicillin-type antibiotics
US10064828B1 (en) * 2005-12-23 2018-09-04 Intellipharmaceutics Corp. Pulsed extended-pulsed and extended-pulsed pulsed drug delivery systems
WO2007079082A2 (en) * 2005-12-30 2007-07-12 Advancis Pharmaceutical Corporation Gastric release pulse system for drug delivery
TW200808380A (en) * 2006-02-21 2008-02-16 Shinetsu Chemical Co Enteric-coated preparation for site-specific delivery of drug to site within the small intestine
US9561188B2 (en) 2006-04-03 2017-02-07 Intellipharmaceutics Corporation Controlled release delivery device comprising an organosol coat
RS58162B1 (en) 2006-04-04 2019-03-29 Kg Acquisition Llc Oral dosage forms including an antiplatelet agent and an acid inhibitor
US8518926B2 (en) 2006-04-10 2013-08-27 Knopp Neurosciences, Inc. Compositions and methods of using (R)-pramipexole
WO2007121188A2 (en) * 2006-04-10 2007-10-25 Knopp Neurosciences, Inc. Compositions and methods of using r(+) pramipexole
US20070238942A1 (en) * 2006-04-10 2007-10-11 Esophamet Corp. Apparatus and method for detecting gastroesophageal reflux disease (gerd)
US8299052B2 (en) 2006-05-05 2012-10-30 Shionogi Inc. Pharmaceutical compositions and methods for improved bacterial eradication
US10960077B2 (en) 2006-05-12 2021-03-30 Intellipharmaceutics Corp. Abuse and alcohol resistant drug composition
CA2651890C (en) 2006-05-12 2015-01-27 Shire Llc Controlled dose drug delivery system
EP2465500A1 (en) 2006-05-16 2012-06-20 Knopp Neurosciences, Inc. Therapeutically effective amounts of R(+) and S(-) pramipexole for use in the treatment of parkinson's disease
PL2026803T3 (en) 2006-05-16 2012-05-31 Knopp Neurosciences Inc Compositions of r(+) and s(-) pramipexole and methods of using the same
PE20080907A1 (en) * 2006-08-31 2008-08-22 Novartis Ag EXTENDED RELEASE GASTRO-RETENTIVE ORAL DRUG DELIVERY SYSTEM FOR VALSARTAN
ITMI20061692A1 (en) 2006-09-05 2008-03-06 Alfa Wassermann Spa USE OF POLYOLS TO OBTAIN STYLE POLYMORPHIC FORMS OF RIFAXIMINA
JP5389656B2 (en) 2006-10-20 2014-01-15 マクニール−ピーピーシー・インコーポレーテツド Acetaminophen / ibuprofen combinations and methods for their use
US8524695B2 (en) 2006-12-14 2013-09-03 Knopp Neurosciences, Inc. Modified release formulations of (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and methods of using the same
US20100104621A1 (en) * 2007-02-21 2010-04-29 Connected Health Systems, Llc Treating adhd and other diseases involving inflammation
JP2010521496A (en) 2007-03-14 2010-06-24 ノップ ニューロサイエンシーズ、インク. Synthesis of chiral purified substituted benzothiazolediamine
US20080268047A1 (en) * 2007-04-24 2008-10-30 Xavier University Of Louisiana Controlled Release Multiple Layer Coatings
US20080317677A1 (en) * 2007-06-22 2008-12-25 Szymczak Christopher E Laser Marked Dosage Forms
US20080317678A1 (en) * 2007-06-22 2008-12-25 Szymczak Christopher E Laser Marked Dosage Forms
EP2044932A1 (en) * 2007-10-04 2009-04-08 Laboratorios del Dr. Esteve S.A. Mechanical protective layer for solid dosage forms
EP2044929A1 (en) * 2007-10-04 2009-04-08 Laboratorios del Dr. Esteve S.A. Oral fast distintegrating tablets
CN101842085B (en) * 2007-10-31 2013-01-30 麦克内尔-Ppc股份有限公司 Orally disintegrated dosage form
WO2009088414A2 (en) 2007-12-06 2009-07-16 Durect Corporation Oral pharmaceutical dosage forms
BRPI0821735A2 (en) * 2007-12-21 2015-06-16 Mcneil Ppc Inc Tablet Manufacturing
MX2010008861A (en) * 2008-02-15 2011-02-22 Sun Pharma Advanced Res Co Ltd Oral controlled release tablet with reduced burst effect.
CN102149369B (en) 2008-04-18 2016-08-10 因泰克制药有限公司 Carbidopa/lipodopa gastroretentive drug delivery
US8282957B2 (en) * 2008-06-26 2012-10-09 Mcneil-Ppc, Inc. Coated particles containing pharmaceutically active agents
WO2010022140A1 (en) 2008-08-19 2010-02-25 Knopp Neurosciences, Inc. Compositions and methods of using (r)-pramipexole
US20100260844A1 (en) 2008-11-03 2010-10-14 Scicinski Jan J Oral pharmaceutical dosage forms
WO2010061696A1 (en) 2008-11-25 2010-06-03 日産自動車株式会社 Conductive member and solid state polymer fuel cell using same
WO2010064100A1 (en) * 2008-12-04 2010-06-10 Intec Pharma Ltd. Baclofen gastroretentive drug delivery system
WO2010148409A1 (en) * 2009-06-19 2010-12-23 Knopp Neurosciences, Inc. Compositions and methods for treating amyotrophic lateral sclerosis
RU2555531C2 (en) 2009-07-22 2015-07-10 Грюненталь Гмбх Misuse protected dosage form for oxidation sensitive opioids
KR101738369B1 (en) 2009-07-22 2017-05-22 그뤼넨탈 게엠베하 Hot-melt extruded controlled release dosage form
US8858210B2 (en) 2009-09-24 2014-10-14 Mcneil-Ppc, Inc. Manufacture of variable density dosage forms utilizing radiofrequency energy
US9610224B2 (en) 2009-09-24 2017-04-04 Johnson & Johnson Consumer Inc. Manufacture of tablet in a die utilizing powder blend containing water-containing material
US8313768B2 (en) 2009-09-24 2012-11-20 Mcneil-Ppc, Inc. Manufacture of tablet having immediate release region and sustained release region
GB201002612D0 (en) * 2010-02-16 2010-03-31 Jagotec Ag Improvements in or relating to organic compounds
JP5968300B2 (en) 2010-03-24 2016-08-10 ジャズ、ファーマシューティカルズ、インコーポレイテッドJazz Pharmaceuticals Inc. Controlled release dosage forms for high dose, water soluble and hygroscopic drug substrates
US10463633B2 (en) * 2010-04-23 2019-11-05 Kempharm, Inc. Therapeutic formulation for reduced drug side effects
SG186301A1 (en) * 2010-06-16 2013-02-28 Takeda Pharmaceuticals Usa Inc Novel modified release dosage forms of xanthine oxidoreductase inhibitor or xanthine oxidase inhibitors
EP2611426B1 (en) 2010-09-02 2014-06-25 Grünenthal GmbH Tamper resistant dosage form comprising inorganic salt
US9119809B2 (en) 2011-03-23 2015-09-01 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US11241391B2 (en) 2011-03-23 2022-02-08 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US8927010B2 (en) 2011-03-23 2015-01-06 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US10905652B2 (en) 2011-03-23 2021-02-02 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US9603809B2 (en) 2011-03-23 2017-03-28 Ironshore Pharmaceuticals & Development, Inc. Methods of treatment of attention deficit hyperactivity disorder
DK2688557T3 (en) 2011-03-23 2017-11-27 Ironshore Pharmaceuticals & Dev Inc PROCEDURES AND COMPOSITIONS FOR TREATMENT OF DIFFICULTY OF ATTENTION
US9498447B2 (en) 2011-03-23 2016-11-22 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US9283214B2 (en) 2011-03-23 2016-03-15 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US8916588B2 (en) 2011-03-23 2014-12-23 Ironshore Pharmaceuticals & Development, Inc. Methods for treatment of attention deficit hyperactivity disorder
US10292937B2 (en) 2011-03-23 2019-05-21 Ironshore Pharmaceuticals & Development, Inc. Methods of treatment of attention deficit hyperactivity disorder
WO2013003622A1 (en) 2011-06-28 2013-01-03 Neos Therapeutics, Lp Dosage forms for oral administration and methods of treatment using the same
AR087359A1 (en) 2011-07-29 2014-03-19 Gruenenthal Gmbh TEST ALTERATION TABLET PROVIDING IMMEDIATE RELEASE OF THE PHARMACO
EA201400172A1 (en) 2011-07-29 2014-06-30 Грюненталь Гмбх SUSTAINABLE TO DESTRUCTION TABLET THAT PROVIDES IMMEDIATE RELEASE OF MEDICINES
WO2013059676A1 (en) * 2011-10-21 2013-04-25 Subhash Desai Compositions for reduction of side effects
CA2758556A1 (en) * 2011-11-17 2013-05-17 Pharmascience Inc. Pharmaceutical composition of amphetamine mixed salts
US9512096B2 (en) 2011-12-22 2016-12-06 Knopp Biosciences, LLP Synthesis of amine substituted 4,5,6,7-tetrahydrobenzothiazole compounds
MX362357B (en) 2012-04-18 2019-01-14 Gruenenthal Gmbh Tamper resistant and dose-dumping resistant pharmaceutical dosage form.
US9511028B2 (en) 2012-05-01 2016-12-06 Johnson & Johnson Consumer Inc. Orally disintegrating tablet
US9233491B2 (en) 2012-05-01 2016-01-12 Johnson & Johnson Consumer Inc. Machine for production of solid dosage forms
US9445971B2 (en) 2012-05-01 2016-09-20 Johnson & Johnson Consumer Inc. Method of manufacturing solid dosage form
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
US9662313B2 (en) 2013-02-28 2017-05-30 Knopp Biosciences Llc Compositions and methods for treating amyotrophic lateral sclerosis in responders
US9572885B2 (en) 2013-03-15 2017-02-21 Durect Corporation Compositions with a rheological modifier to reduce dissolution variability
JP6445537B2 (en) 2013-05-29 2018-12-26 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Tamper-resistant dosage forms containing one or more particles
EA032465B1 (en) 2013-07-12 2019-05-31 Грюненталь Гмбх Tamper-resistant oral pharmaceutical dosage form containing ethylene-vinyl acetate polymer and process for the production thereof
US9468630B2 (en) 2013-07-12 2016-10-18 Knopp Biosciences Llc Compositions and methods for treating conditions related to increased eosinophils
US10828284B2 (en) 2013-07-12 2020-11-10 Knopp Biosciences Llc Compositions and methods for treating conditions related to elevated levels of eosinophils and/or basophils
WO2015023786A1 (en) 2013-08-13 2015-02-19 Knopp Biosciences Llc Compositions and methods for treating plasma cell disorders and b-cell prolymphocytic disorders
AU2014306597B2 (en) 2013-08-13 2018-05-17 Knopp Biosciences Llc Compositions and methods for treating chronic urticaria
AU2014356581C1 (en) 2013-11-26 2020-05-28 Grunenthal Gmbh Preparation of a powdery pharmaceutical composition by means of cryo-milling
AU2015204763A1 (en) 2014-01-10 2016-07-21 Johnson & Johnson Consumer Inc. Process for making tablet using radiofrequency and lossy coated particles
AU2015261060A1 (en) 2014-05-12 2016-11-03 Grunenthal Gmbh Tamper resistant immediate release capsule formulation comprising Tapentadol
CA2949422A1 (en) 2014-05-26 2015-12-03 Grunenthal Gmbh Multiparticles safeguarded against ethanolic dose-dumping
US10398662B1 (en) 2015-02-18 2019-09-03 Jazz Pharma Ireland Limited GHB formulation and method for its manufacture
EP3261625A4 (en) * 2015-02-27 2018-10-24 Cingulate Therapeutics LLC Tripulse release stimulant formulations
EP3285745A1 (en) 2015-04-24 2018-02-28 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
JP2018526414A (en) 2015-09-10 2018-09-13 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Protection against oral overdose with abuse-inhibiting immediate release formulations
US20170296476A1 (en) * 2016-04-15 2017-10-19 Grünenthal GmbH Modified release abuse deterrent dosage forms
US11602512B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
UY37341A (en) 2016-07-22 2017-11-30 Flamel Ireland Ltd FORMULATIONS OF GAMMA-MODIFIED RELEASE HYDROXIBUTIRATE WITH IMPROVED PHARMACOCINETICS
US11504347B1 (en) 2016-07-22 2022-11-22 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602513B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
EP3372225A1 (en) * 2017-03-09 2018-09-12 Develco Pharma Schweiz AG Novel dosage form
US20180263936A1 (en) 2017-03-17 2018-09-20 Jazz Pharmaceuticals Ireland Limited Gamma-hydroxybutyrate compositions and their use for the treatment of disorders
US10493026B2 (en) 2017-03-20 2019-12-03 Johnson & Johnson Consumer Inc. Process for making tablet using radiofrequency and lossy coated particles
BR112019024708A2 (en) * 2017-06-14 2020-06-09 SpecGx LLC delayed-release pharmaceutical compositions
WO2019073028A1 (en) 2017-10-13 2019-04-18 Grünenthal GmbH Modified release abuse deterrent dosage forms
KR20210094513A (en) 2018-11-19 2021-07-29 재즈 파마슈티칼즈 아일랜드 리미티드 Alcohol-Resistant Drug Formulations
JP2022522270A (en) 2019-03-01 2022-04-15 フラメル アイルランド リミテッド Gamma-hydroxybutyrate composition with improved pharmacokinetics in dietary intake
US11779557B1 (en) 2022-02-07 2023-10-10 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11583510B1 (en) 2022-02-07 2023-02-21 Flamel Ireland Limited Methods of administering gamma hydroxybutyrate formulations after a high-fat meal

Citations (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2099402A (en) 1934-01-17 1937-11-16 Pratt Food Company Pill or tablet
US2738303A (en) 1952-07-18 1956-03-13 Smith Kline French Lab Sympathomimetic preparation
US3048526A (en) 1958-08-04 1962-08-07 Wander Company Medicinal tablet
US3365365A (en) 1965-08-09 1968-01-23 Hoffmann La Roche Repeat action pharmaceutical compositions in the form of discrete beadlets
US3979349A (en) 1973-05-07 1976-09-07 Rohm Gmbh Dispersions of water-soluble polymers and methods for making and using the same
US4049791A (en) 1974-06-21 1977-09-20 Delco Chemical Company, Inc. Prolonged acting appetite suppressant and anti-obesity compositions containing amphetamine adipate, dextroamphetamine adipate, amphetamine sulfate and dextroamphetamine sulfate as the active agents
JPS5982311A (en) 1982-11-04 1984-05-12 Shionogi & Co Ltd Sustained release preparation of cephalexin
WO1987000044A1 (en) * 1985-07-02 1987-01-15 The Upjohn Company Therapeutic formulations with bimodal release characteristics
US4723958A (en) * 1986-05-23 1988-02-09 Merck & Co., Inc. Pulsatile drug delivery system
US4728512A (en) 1985-05-06 1988-03-01 American Home Products Corporation Formulations providing three distinct releases
US4765989A (en) * 1983-05-11 1988-08-23 Alza Corporation Osmotic device for administering certain drugs
US4794001A (en) 1986-03-04 1988-12-27 American Home Products Corporation Formulations providing three distinct releases
US4871549A (en) * 1985-07-19 1989-10-03 Fujisawa Pharmaceutical Co., Ltd. Time-controlled explosion systems and processes for preparing the same
US4891230A (en) * 1983-12-22 1990-01-02 Elan Corporation Plc Diltiazem formulation
US4894240A (en) * 1983-12-22 1990-01-16 Elan Corporation Plc Controlled absorption diltiazem formulation for once-daily administration
US4902516A (en) * 1987-04-10 1990-02-20 Chemie Holding Aktiengesellschaft Binder-free granules with delayed release of the active compound
US4917899A (en) * 1983-12-22 1990-04-17 Elan Corporation Plc Controlled absorption diltiazem formulation
WO1990009168A1 (en) * 1989-02-16 1990-08-23 National Research Development Corporation Dispensing device
US5002776A (en) * 1983-12-22 1991-03-26 Elan Corporation, Plc Controlled absorption diltiazem formulations
US5011692A (en) * 1985-12-28 1991-04-30 Sumitomo Pharmaceuticals Company, Limited Sustained pulsewise release pharmaceutical preparation
US5011694A (en) * 1988-08-11 1991-04-30 Rohm Gmbh Pharmaceutical dosage unit forms with delayed release
JPH03148215A (en) 1989-11-01 1991-06-25 Nippon Shinyaku Co Ltd Laminated formulation
US5051262A (en) * 1979-12-07 1991-09-24 Elan Corp., P.L.C. Processes for the preparation of delayed action and programmed release pharmaceutical forms and medicaments obtained thereby
US5093200A (en) * 1985-10-23 1992-03-03 Eisai Co., Ltd. Multilayer sustained release granule
US5137733A (en) 1990-06-28 1992-08-11 Tanabe Seiyaku Co., Ltd. Controlled release pharmaceutical preparation
US5202159A (en) 1990-12-27 1993-04-13 Standard Chemical & Pharmaceutical Corp., Ltd. Preparation method of microdispersed tablet formulation of spray-dried sodium diclofenac enteric-coated microcapsules
US5226902A (en) * 1991-07-30 1993-07-13 University Of Utah Pulsatile drug delivery device using stimuli sensitive hydrogel
US5229131A (en) * 1990-02-05 1993-07-20 University Of Michigan Pulsatile drug delivery system
US5260068A (en) 1992-05-04 1993-11-09 Anda Sr Pharmaceuticals Inc. Multiparticulate pulsatile drug delivery system
US5260069A (en) 1992-11-27 1993-11-09 Anda Sr Pharmaceuticals Inc. Pulsatile particles drug delivery system
US5275819A (en) * 1989-02-06 1994-01-04 Amer Particle Technologies Inc. Drug loaded pollen grains with an outer coating for pulsed delivery
US5312388A (en) * 1991-01-30 1994-05-17 Wong Patrick S L Osmotic device for delayed delivery of agent
US5364620A (en) * 1983-12-22 1994-11-15 Elan Corporation, Plc Controlled absorption diltiazem formulation for once daily administration
US5378474A (en) 1989-01-06 1995-01-03 F. H. Faulding & Co. Limited Sustained release pharmaceutical composition
EP0640337A2 (en) 1993-08-25 1995-03-01 Ss Pharmaceutical Co., Ltd. Controlled release-initiation and controlled release-rate pharmaceutical composition
US5395628A (en) * 1989-12-28 1995-03-07 Tanabe Seiyaku Co., Ltd. Controlled release succinic acid microcapsules coated with aqueous acrylics
US5407686A (en) * 1991-11-27 1995-04-18 Sidmak Laboratories, Inc. Sustained release composition for oral administration of active ingredient
US5411745A (en) 1994-05-25 1995-05-02 Euro-Celtique, S.A. Powder-layered morphine sulfate formulations
US5422121A (en) 1990-11-14 1995-06-06 Rohm Gmbh Oral dosage unit form
US5474786A (en) * 1994-03-23 1995-12-12 Ortho Pharmaceutical Corporation Multilayered controlled release pharmaceutical dosage form
US5541170A (en) 1981-07-31 1996-07-30 Tillotts Pharma Ag Orally administrable pharmaceutical compositions
WO1997003673A1 (en) 1995-07-14 1997-02-06 Medeva Europe Limited Sustained-release formulation of d-threo-methylphenidate
US5618559A (en) 1994-01-27 1997-04-08 G.D. Searle & Co. Method of making modified-release metronidazole compositions
JPH09249557A (en) 1996-03-14 1997-09-22 Shionogi & Co Ltd Sustained release preparation of slightly water-soluble medicine
JPH09267035A (en) 1996-01-30 1997-10-14 Kanegafuchi Chem Ind Co Ltd Production of nuclear particle
JPH1081634A (en) 1996-07-18 1998-03-31 Taisho Pharmaceut Co Ltd Base controlled in dissolution time
US5733575A (en) 1994-10-07 1998-03-31 Bpsi Holdings, Inc. Enteric film coating compositions, method of coating therewith, and coated forms
WO1998014168A2 (en) 1996-09-30 1998-04-09 Alza Corporation Dosage form providing a sustained and ascending drug release
US5800836A (en) * 1992-08-05 1998-09-01 F. H. Faulding & Co. Limited Pelletized pharmaceutical composition
US5824341A (en) * 1994-08-11 1998-10-20 Pharma Pass Composition providing selective release of an active ingredient
US5824343A (en) * 1989-08-28 1998-10-20 Pharmaceutical Delivery Systems Bioerodible polymers, compositions and therapeutic method
US5824342A (en) * 1994-07-01 1998-10-20 Fuisz Technologies Ltd. Flash flow formed solloid delivery systems
US5837284A (en) * 1995-12-04 1998-11-17 Mehta; Atul M. Delivery of multiple doses of medications
US5840329A (en) * 1997-05-15 1998-11-24 Bioadvances Llc Pulsatile drug delivery system
US5885616A (en) * 1997-08-18 1999-03-23 Impax Pharmaceuticals, Inc. Sustained release drug delivery system suitable for oral administration
US5885998A (en) * 1995-01-06 1999-03-23 Bencherif; Merouane Methods for prevention and treatment of attention deficit disorder
US5891474A (en) * 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
WO2000025752A1 (en) 1998-11-02 2000-05-11 Church, Marla, J. Multiparticulate modified release composition
WO2000035450A1 (en) 1998-12-17 2000-06-22 Euro-Celtique S.A. Controlled/modified release oral methylphenidate formulations
US6322819B1 (en) 1998-10-21 2001-11-27 Shire Laboratories, Inc. Oral pulsed dose drug delivery system
US20040059002A1 (en) 2002-09-24 2004-03-25 Shire Laboratories, Inc. Sustained release delivery of amphetamine salts
US6749867B2 (en) 2000-11-29 2004-06-15 Joseph R. Robinson Delivery system for omeprazole and its salts
US6764696B2 (en) 1998-04-29 2004-07-20 Cima Labs Inc. Effervescent drug delivery system for oral administration

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2881113A (en) * 1957-01-29 1959-04-07 Ortho Pharma Corp Therapeutically active compositions containing amphetamines
US2993836A (en) * 1958-02-20 1961-07-25 Dow Chemical Co Sustained release tablets
US3066075A (en) * 1960-07-25 1962-11-27 G W Carnrick Co Compositions comprising amphetamine and carboxymethyl cellulose in chemically combined form
NL297631A (en) * 1963-06-03
US5232705A (en) * 1990-08-31 1993-08-03 Alza Corporation Dosage form for time-varying patterns of drug delivery
US5156850A (en) * 1990-08-31 1992-10-20 Alza Corporation Dosage form for time-varying patterns of drug delivery
DE69331839T2 (en) * 1992-01-29 2002-12-12 Takeda Chemical Industries Ltd Fast-dissolving tablet and its manufacture
US5328697A (en) * 1992-02-10 1994-07-12 Mallinckrodt Veterinary, Inc. Compositions and processes for the sustained release of drugs
US5308348A (en) * 1992-02-18 1994-05-03 Alza Corporation Delivery devices with pulsatile effect
US5322697A (en) * 1992-05-28 1994-06-21 Meyer James H Composition and method for inducing satiety
WO1997007165A1 (en) * 1995-08-16 1997-02-27 Osi Specialties, Inc. Stable silane compositions on silica carrier
US5908850A (en) * 1995-12-04 1999-06-01 Celgene Corporation Method of treating attention deficit disorders with d-threo methylphenidate
US5922736A (en) * 1995-12-04 1999-07-13 Celegene Corporation Chronic, bolus administration of D-threo methylphenidate
US5773031A (en) * 1996-02-27 1998-06-30 L. Perrigo Company Acetaminophen sustained-release formulation
US5846568A (en) * 1996-09-19 1998-12-08 Xyrofin Oy Directly compressible lactitol and method
HRP970493A2 (en) * 1996-09-23 1998-08-31 Wienman E. Phlips Oral delayed immediate release medical formulation and method for preparing the same
US5945123A (en) * 1998-04-02 1999-08-31 K-V Pharmaceutical Company Maximizing effectiveness of substances used to improve health and well being
JP2002541093A (en) * 1999-04-06 2002-12-03 ファーマクエスト・リミテッド Pharmaceutical dosage form for pulsed delivery of d-threo-methylphenidate and a second CNS stimulant
DE60036874T2 (en) * 1999-09-02 2008-06-26 Nostrum Pharmaceuticals Inc. PELLET FORMULATION WITH CONTROLLED RELEASE

Patent Citations (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2099402A (en) 1934-01-17 1937-11-16 Pratt Food Company Pill or tablet
US2738303A (en) 1952-07-18 1956-03-13 Smith Kline French Lab Sympathomimetic preparation
US3048526A (en) 1958-08-04 1962-08-07 Wander Company Medicinal tablet
US3365365A (en) 1965-08-09 1968-01-23 Hoffmann La Roche Repeat action pharmaceutical compositions in the form of discrete beadlets
US3979349A (en) 1973-05-07 1976-09-07 Rohm Gmbh Dispersions of water-soluble polymers and methods for making and using the same
US4049791A (en) 1974-06-21 1977-09-20 Delco Chemical Company, Inc. Prolonged acting appetite suppressant and anti-obesity compositions containing amphetamine adipate, dextroamphetamine adipate, amphetamine sulfate and dextroamphetamine sulfate as the active agents
US5051262A (en) * 1979-12-07 1991-09-24 Elan Corp., P.L.C. Processes for the preparation of delayed action and programmed release pharmaceutical forms and medicaments obtained thereby
US5541170A (en) 1981-07-31 1996-07-30 Tillotts Pharma Ag Orally administrable pharmaceutical compositions
JPS5982311A (en) 1982-11-04 1984-05-12 Shionogi & Co Ltd Sustained release preparation of cephalexin
US4765989A (en) * 1983-05-11 1988-08-23 Alza Corporation Osmotic device for administering certain drugs
US5002776A (en) * 1983-12-22 1991-03-26 Elan Corporation, Plc Controlled absorption diltiazem formulations
US5616345A (en) * 1983-12-22 1997-04-01 Elan Corporation Plc Controlled absorption diltiazen formulation for once-daily administration
US5364620A (en) * 1983-12-22 1994-11-15 Elan Corporation, Plc Controlled absorption diltiazem formulation for once daily administration
US4891230A (en) * 1983-12-22 1990-01-02 Elan Corporation Plc Diltiazem formulation
US4894240A (en) * 1983-12-22 1990-01-16 Elan Corporation Plc Controlled absorption diltiazem formulation for once-daily administration
US4917899A (en) * 1983-12-22 1990-04-17 Elan Corporation Plc Controlled absorption diltiazem formulation
US4728512A (en) 1985-05-06 1988-03-01 American Home Products Corporation Formulations providing three distinct releases
WO1987000044A1 (en) * 1985-07-02 1987-01-15 The Upjohn Company Therapeutic formulations with bimodal release characteristics
US4871549A (en) * 1985-07-19 1989-10-03 Fujisawa Pharmaceutical Co., Ltd. Time-controlled explosion systems and processes for preparing the same
US5093200A (en) * 1985-10-23 1992-03-03 Eisai Co., Ltd. Multilayer sustained release granule
US5011692A (en) * 1985-12-28 1991-04-30 Sumitomo Pharmaceuticals Company, Limited Sustained pulsewise release pharmaceutical preparation
US4794001A (en) 1986-03-04 1988-12-27 American Home Products Corporation Formulations providing three distinct releases
US4723958A (en) * 1986-05-23 1988-02-09 Merck & Co., Inc. Pulsatile drug delivery system
US4902516A (en) * 1987-04-10 1990-02-20 Chemie Holding Aktiengesellschaft Binder-free granules with delayed release of the active compound
US5011694A (en) * 1988-08-11 1991-04-30 Rohm Gmbh Pharmaceutical dosage unit forms with delayed release
US5378474A (en) 1989-01-06 1995-01-03 F. H. Faulding & Co. Limited Sustained release pharmaceutical composition
US5275819A (en) * 1989-02-06 1994-01-04 Amer Particle Technologies Inc. Drug loaded pollen grains with an outer coating for pulsed delivery
WO1990009168A1 (en) * 1989-02-16 1990-08-23 National Research Development Corporation Dispensing device
US5824343A (en) * 1989-08-28 1998-10-20 Pharmaceutical Delivery Systems Bioerodible polymers, compositions and therapeutic method
JPH03148215A (en) 1989-11-01 1991-06-25 Nippon Shinyaku Co Ltd Laminated formulation
US5395628A (en) * 1989-12-28 1995-03-07 Tanabe Seiyaku Co., Ltd. Controlled release succinic acid microcapsules coated with aqueous acrylics
US5229131A (en) * 1990-02-05 1993-07-20 University Of Michigan Pulsatile drug delivery system
US5137733A (en) 1990-06-28 1992-08-11 Tanabe Seiyaku Co., Ltd. Controlled release pharmaceutical preparation
US5422121A (en) 1990-11-14 1995-06-06 Rohm Gmbh Oral dosage unit form
US5202159A (en) 1990-12-27 1993-04-13 Standard Chemical & Pharmaceutical Corp., Ltd. Preparation method of microdispersed tablet formulation of spray-dried sodium diclofenac enteric-coated microcapsules
US5312388A (en) * 1991-01-30 1994-05-17 Wong Patrick S L Osmotic device for delayed delivery of agent
US5226902A (en) * 1991-07-30 1993-07-13 University Of Utah Pulsatile drug delivery device using stimuli sensitive hydrogel
US5407686A (en) * 1991-11-27 1995-04-18 Sidmak Laboratories, Inc. Sustained release composition for oral administration of active ingredient
US5260068A (en) 1992-05-04 1993-11-09 Anda Sr Pharmaceuticals Inc. Multiparticulate pulsatile drug delivery system
US5800836A (en) * 1992-08-05 1998-09-01 F. H. Faulding & Co. Limited Pelletized pharmaceutical composition
US5260069A (en) 1992-11-27 1993-11-09 Anda Sr Pharmaceuticals Inc. Pulsatile particles drug delivery system
EP0640337A2 (en) 1993-08-25 1995-03-01 Ss Pharmaceutical Co., Ltd. Controlled release-initiation and controlled release-rate pharmaceutical composition
US5496561A (en) 1993-08-25 1996-03-05 Ss Pharmaceutical Co., Ltd. Controlled release-initiation and controlled release-rate pharmaceutical composition
JPH0761922A (en) 1993-08-25 1995-03-07 Ss Pharmaceut Co Ltd Release start controlling type pharmaceutical preparation
US5618559A (en) 1994-01-27 1997-04-08 G.D. Searle & Co. Method of making modified-release metronidazole compositions
US5474786A (en) * 1994-03-23 1995-12-12 Ortho Pharmaceutical Corporation Multilayered controlled release pharmaceutical dosage form
US5411745A (en) 1994-05-25 1995-05-02 Euro-Celtique, S.A. Powder-layered morphine sulfate formulations
US5824342A (en) * 1994-07-01 1998-10-20 Fuisz Technologies Ltd. Flash flow formed solloid delivery systems
US5824341A (en) * 1994-08-11 1998-10-20 Pharma Pass Composition providing selective release of an active ingredient
US5733575A (en) 1994-10-07 1998-03-31 Bpsi Holdings, Inc. Enteric film coating compositions, method of coating therewith, and coated forms
US5885998A (en) * 1995-01-06 1999-03-23 Bencherif; Merouane Methods for prevention and treatment of attention deficit disorder
WO1997003673A1 (en) 1995-07-14 1997-02-06 Medeva Europe Limited Sustained-release formulation of d-threo-methylphenidate
US5837284A (en) * 1995-12-04 1998-11-17 Mehta; Atul M. Delivery of multiple doses of medications
JPH09267035A (en) 1996-01-30 1997-10-14 Kanegafuchi Chem Ind Co Ltd Production of nuclear particle
JPH09249557A (en) 1996-03-14 1997-09-22 Shionogi & Co Ltd Sustained release preparation of slightly water-soluble medicine
JPH1081634A (en) 1996-07-18 1998-03-31 Taisho Pharmaceut Co Ltd Base controlled in dissolution time
WO1998014168A2 (en) 1996-09-30 1998-04-09 Alza Corporation Dosage form providing a sustained and ascending drug release
US5891474A (en) * 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5840329A (en) * 1997-05-15 1998-11-24 Bioadvances Llc Pulsatile drug delivery system
WO1999003471A1 (en) 1997-07-14 1999-01-28 Mehta, Atul, M. Improved delivery of multiple doses of medications
US5885616A (en) * 1997-08-18 1999-03-23 Impax Pharmaceuticals, Inc. Sustained release drug delivery system suitable for oral administration
US6764696B2 (en) 1998-04-29 2004-07-20 Cima Labs Inc. Effervescent drug delivery system for oral administration
US6322819B1 (en) 1998-10-21 2001-11-27 Shire Laboratories, Inc. Oral pulsed dose drug delivery system
US6605300B1 (en) 1998-10-21 2003-08-12 Shire Laboratories, Inc. Oral pulsed dose drug delivery system
WO2000025752A1 (en) 1998-11-02 2000-05-11 Church, Marla, J. Multiparticulate modified release composition
WO2000035450A1 (en) 1998-12-17 2000-06-22 Euro-Celtique S.A. Controlled/modified release oral methylphenidate formulations
US6749867B2 (en) 2000-11-29 2004-06-15 Joseph R. Robinson Delivery system for omeprazole and its salts
US20040059002A1 (en) 2002-09-24 2004-03-25 Shire Laboratories, Inc. Sustained release delivery of amphetamine salts

Non-Patent Citations (217)

* Cited by examiner, † Cited by third party
Title
Actavis Elizabeth LLC's Supplemental Responses to Plaintiff Shire LLC's Interrogatory Nos. 5 and 6 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Nov. 14, 2007.
Adderall XR Package Inset, Sep. (2004).
Agyilirah GA and Bauker SB, Polymers for Enteric Coating Applications, Polymers for Controlled Drug Delivery (Peter J. Tarcha ed. 1991) 39-66.
Amendment dated Sep. 24, 2007 in U.S. Appl. No. 11/091,011.
American Chemical Society, Polymer Preprints, pp. 633-634, vol. 34, No. 1, Mar. 1993.
Angrist et al., Early Pharmacokinetics and Clinical Effects of Oral D-Amphetamine in Normal Subjects, Biol. Psychiatry 1987, 22: 1357-1368.
Ansel et al., Rate Controlled Dosage Forms and Drug Delivery Systems, Pharmaceutical Dosage Forms and Drug Delivery Systems, 6th Ed. (1995), 213-222.
Answer and Counterclaims in Shire Laboratories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Jul. 24, 2006.
Answer and Counterclaims in Shire Laboratories, Inc. and Shire, LLC v. Andrx Pharmaceuticals, LLC, et al., in the United States District Court for the Southern District of Florida, Case No. 07-22201-CIV-Cooke/Brown, Aug. 31, 2007.
Answering Expert Report of Dr. Alexander Klibanov, Apr. 25, 2005.
Answering Expert Report of Robert Langer, Apr. 25, 2005.
Barr Laboratores' Memorandum In Support of its Motion to Amend its Pleadings and exhibits thereto.
Barr Laboratories' Answer. Affirmative Defenses and Counterclaim in Case No. 03-CV-1219(VM)(DFE) (S D N Y ) 2003. *
Barr Laboratories' Inc.'s Objections and Responses to Shire laboratories Inc.'s Second Set of Interrogatories (Nos. 8-11).
Barr Laboratories' Objections and Responses to Plaintiff Shire Laboratories Inc.'s Fifth Set of Interrogatories (No. 17).
Barr Laboratories' Objections and Responses to Plaintiff Shire Laboratories Inc.s' Fourth Set of Interrogatories (Nos. 15-16).
Barr Laboratories' Supplemental Objections and Responses to Plaintiff Shire Laboratories Inc.'s Third Set of Interrogatories (Nos. 12-14 Redacted).
Barr's Paragraph IV Certification against Parent U S Patent 6,322,819 on Jan. 14, 2003. *
Bauer, et al., Cellulose Acetate Phthalate (CAP) and Trimellitate (CAT), Coated Pharmaceutical Dosage Forms (1998), 102-104.
Bodmeier et al., The Influence of Buffer Species and Strength on Dittiazem HCl Release from Beads Coated with the Aqueous Cationic Polmer Dispersions, Eudraglt RS, RL 30D, Pharmaceutical Research vol. 13, No. 1, 1996, 52-56.
Brauer et al., Acute Tolerance to Subjective but not Cardiovascular Effects of d-Amphetamine in Normal, Healthy Men, Journal of Clinical Psychopharmacology, 1996; 16(1):72-76.
Brown et al., Behavior and Motor Activity Response in Hyperactive Children and Plasma Amphetamine Levels Following a Sustained Release Preparation, Journal of the American Academy of Child Psychiatry, 19:225-239, 1980.
Brown et al., Plasma Levels of d-Amphetamine in Hyperactive Children, Psychopharmacology 62, 133-140, 1979.
Burns et al., A Study of Enteric-coated Liquid-filled Hard Gelatin Capsules with Biphasic Release Characteristics, International Journal of Pharmaceutics 110 (1994) 291-296.
Burnside Deposition Transcript, Feb. 2, 2005.
Burnside Deposition Transcript, Feb. 3, 2005.
C. Lin et al., Bioavailability of d-pseudoephedrine and Azatadine from a Repeat Action Tablet Formulation, J Int Med Res (1982), 122-125.
C. Lin et al., Compartive Bioavailability of d-pseudoephedrine from a Conventional d-pseudoephedrine Sulfate Tablet and from a Repeat Action Tablet, J Int Med Res (1982) 10, 126-128.
Chan, Materials Used for Effective Sustained-Release Products, Proceedings of the International Symposium held on Jan. 29-31, 1987 (The Bombay College of Pharmacy 1988), 69-84.
Chan, New Polymers for Controlled Release Products, Controlled Release Dosage Forms Proceedings of the International Symposium held on Jan. 29-31, 1987 (The Bombay College of Pharmacy 1988) 59-111.
Chang Deposition Transcript, Jan. 20, 2005.
Chang Deposition Transcript, Sep. 8, 2004.
Chang et al., Preparation and Evaluation of Shellac Pseudolatex as an Aqueous Enteric Coating Systems for Pellets, International Journal of Pharmaceuticals, 60 (1990) 171-173, 1990.
Charles S.L. Chiao and Joseph R. Robinson, Sustained-Release Drug Delivery Systems, Remington: The Science and Practice of Pharmacy, Tenth Edition (1995) 1660-1675.
Chaumeil et al., Enrobages gastro-resistants a l'acetophtalate de cellulose, Annales Pharmaceutiques Francaises, 1973, No. 5, pp. 375-384.
Cody et al., Amphetamine Enantiomer Excretion Profile Following Administration of Adderall, Journal of Analytical Toxicology, vol. 2, Oct. 2003, 485-492.
Colony Pharmaceuticals, Inc.'s Amended Responses to Plaintiff Shire LLC's Interrogatories (Nos. 1-7) in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Jun. 5, 2007.
Colony Pharmaceuticals, Inc.'s Responses to Plaintiff Shire LLC's Interrogatories (Nos. 1-7) in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, May 29, 2007.
Colony Pharmaceuticals, Inc.'s Supplemental Responses to Plaintiff Shire LLC's Interrogatory Nos. 2-4 and 7 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Sep. 5, 2007.
Complaint for Declaratory Judgment, Impax Laboratories, Inc. v. Shire International Laboratories, Inc. (Civ. Action No. 05772) and Exhibits attached thereto.
Complaint in Shire Laboratories v. Teva Pharmaceutical Industries Ltd., and exhibits thereto, Case No. 2:06-cv-00952-SD, Mar. 2, 2006.
Conte, et al., Biomaterials, vol. 14, No. 13, pp. 1017-1023 (1993), Press-coated tablets for time-programmed release of drugs. *
Corrected Expert Report of Vladimir P. Torchilin, Ph.D. D.Sc. Regarding the Invalidity of U.S. Patent No. 6,605,300 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Dec. 21, 2007.
Couch Deposition Transcript, Sep. 14, 2004.
Court Docket for Shire Laboratories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Jan. 8, 2007.
Daynes, Treatment of Noctural Enuresis with Enteric-Coated Amphetamine, The Practitioner, No. 1037, vol. 173, Nov. 1954.
Defendant Sandoz, Inc.'s Answers and Objections to Plaintiff Shire LLC's Interrogatories (Nos. 1-9), in the United States District Court for the District of Colorado, Case No. 07-CV-00197-EWN-CBS, Jun. 18, 2007.
Defendant Sandoz, Inc.'s Answers and Objections to Plaintiff Shire LLC's Second Set of Interrogatories (Nos. 10-19), in the United States District Court for the District of Colorado, Case No. 07-CV-00197-EWN-CBS, Nov. 20, 2007.
Defendant Sandoz, Inc.'s Answers and Objections to Plaintiff Shire LLC's Third Set of Interrogatories (Nos. 20-25) and Supplement to Answers to Interrogatories 8 and 9, in the United States District Court for the District of Colorado, Case No. 07-CV-00197-EWN-CBS, Dec. 10, 2007.
Defendants' Response to Plaintiff Shire's First Set of Interrogatories (1-12) in Shire Labatories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Sep. 20, 2006.
Defendants' Responses to Plaintiff Shire's Second Set of Interrogatories (No. 13) in Shire Laboratories v. Teva Pharmaceuticals Industries Ltd., Case No. 2:06-cv-00952-SD, Nov. 8, 2006.
Defendants' Responses to Plaintiff's First Set of Request for the Production of Documents and Things (1-70) in Shire Laboratories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Oct. 4, 2006.
Defendants' Responses to Plaintiff's Second Set of Requests for the Production of Documents and Things (71-80) in Shire Laboratories v. Teva Pharmaceuticals Industries Ltd., Case No. 2:06-cv-00952-SD, Nov. 8, 2006.
Deposition transcript of Honorable Gerald J. Mossinghoff and exhibits thereto.
Edward Stempel, Prolonged Drug Action, HUSA's Pharmaceutical Dispensing, Sixth Edition, 1966, 464, 481-485.
Exh. 10, Excerpts from the Deposition Transcript of Xiaodi Guo, dated Jan. 24, 2005.
Exh. 11, Excerpts from the Deposition Transcript of Xiaodi Guo, dated Jul. 26, 2004.
Exh. 12, Excerpts from the Deposition Transcript of Edward Rudnic, dated Jul. 28, 2004.
Exh. 13, Excerpts from the Deposition Transcript of Richard Rong-Kun Chang, dated Jan. 20, 2005.
Exh. 14, Impax Laboratories Answer And Affirmative Defenses Shire Laboratories, Inc. v. Impax Laboratories, Inc., Civil Action No. 03-CV-01164-GMS.
Exh. 15, Barr Laboratories' Amended Answer, Affirmative Defenses, And Counterclaims, Shire Laboratories, Inc. v. Barr Laboratories, Inc., Civil Action No. 03-CV-6632-PKC.
Exh. 16, Barr Laboratories' Amended Answer, Affirmative Defenses And Counterclaims, Shire Laboratories, Inc. v. Barr Laboratories, Inc., Civil Action No. 03-CV-1219-PKC.
Exh. 17, Reply to Barr Laboratories Inc.'s Amended Answer, Affirmative Defenses And Counterclaims, Shire Laboratories, Inc. v. Barr Laboratories, Inc., Civil Action No. 03-CV-6632-PKC.
Exh. 18, Civil Docket For Case #: 1:03-cv-01164-GMS, Shire Laboratories, Inc. v. Impax Laboratories, Inc., Civil Action No. 03-CV-01164-GMS.
Exh. 19, Civil Docket For Case #: 1:05-cv-00020-GMS, Shire Laboratories, Inc. v. Impax Laboratories, Inc., Civil Action No. 05-20-GMS.
Exh. 20, Civil Docket For Case #: 1:03-cv-06632-VM-DFE, Shire Laboratories, Inc. v. Barr Laboratories, Inc., Civil Action No. 03-CV-6632-PKC.
Exh. 21, Civil Docket For Case #: 1:03-cv-01219-PKC-DFE, Shire Laboratories, Inc. v. Barr Laboratories, Inc., Civil Action No. 03-CV-1219-PKC.
Exh. 25, Barr Laboratories, Inc.'s '819 Notification Pursuant to § 505(j)(B)(ii) of the Federal Food, Drug and Cosmetic Act (21 U.S.C. § 355(j)(2)(B)(ii) and 21 C.F.R. § 314.95.
Exh. 26, Barr Laboratories, Inc.'s '300 Notification Pursuant to § 505(j)(2)(B)(ii) of the Federal Food, Drug and Cosmetic Act (21 U.S.C. § 355(j)(2)(B)(ii) and 21 C.F.R. § 314.95).
Exh. 27, Order Construing The Terms Of U.S. Patent Nos. 6,322,819 and 6,605,300, Shire Laboratories, Inc. v. Impax Laboratories, Inc., Civil Action No. 03-CV-01164-GMS.
Exh. 3, Excerpts from the Deposition Transcript of Richard Chang, dated Sep. 8, 2004.
Exh. 4, Excerpts from the Deposition Transcript of Richard A. Couch, dated Sep. 14, 2004.
Exh. 5, Excerpts from the Deposition Transcript of Kimberly Fiske, dated Sep. 17, 2004.
Exh. 6, Excerpts from the Deposition Transcript of Charlotte M. McGuiness, dated Aug. 6, 2004.
Exh. 7, Excerpts from the Deposition Transcript of Beth Burnside, dated Feb. 2, 2005.
Exh. 8, Excerpts from the Deposition Transcript of Donald John Treacy, Jr., dated Aug. 31, 2004.
Exh. 9, Excerpts from the Deposition Transcript of Beth Burnside, dated Feb. 3, 2005.
Expert Report of Arthur J. Steiner in Shire LLC v. Colony Pharmaceuticals, Inc., in the United States District Court for the District of Maryland, Case No. 1:07-cv-00718, Dec. 20, 2007.
Expert Report of Dr. Joseph R. Robinson and exhibits thereto, Feb. 28, 2005.
Expert Report of Harry G. Brittain, Ph.D. FRSC in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB and exhibits thereto, Dec. 19, 2007.
Expert Report of the Honorable Gerald J. Mossinghoff and exhibits thereto, Mar. 16, 2005.
Expert Report of Vladimir P. Torchilin, Ph.D., D.Sc. Regarding the Invalidity of U.S. patent No. 6,605,300 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB and exhibits thereto, Dec. 20, 2007.
Fiske Deposition Transcript, Sep. 17, 2004.
Freedom of Information Request Results for-Dexadrine (SmithKline Beecham): May 20, 1976 Disclosable Approval Information.
Freedom of Information Request Results for—Dexadrine (SmithKline Beecham): May 20, 1976 Disclosable Approval Information.
Fukumori, Coating of Multiparticulates Using Polymeric Dispersions, Multiparticulate Oral Drug Delivery (Swarbrick and Selassie eds. 1994), 79-110.
Garnett et al., Pharmacokinetic Evaluation of Twice-Daily Extended-Release Carbamazepine (CBZ) and Four-Times- Daily Immediate-Release CBZ in Patients with Epilepsy, Epilepsia 39(3):274-279, 1998.
Gazzaniga, et al., Eur. J. Pharm. Biopharm., vol. 40, No. 4, pp. 246-250 (1994), Oral Chronotopic Drug Delivery System: Achievement of Time and/or Site Specificity. *
Gazzaniga, et al., S.T.P. Pharma Sciences, vol. 5, No. 1, gs. 83-88 (1995), Time dependent oral delivery for colon targeting. *
Ghebre-Sellassie et al., "Evaluation of acrylic-based modified-release film coatings," International Journal of Pharmaceutics, 1987;37:211-218.
Glatt, The World of the Fluid Bed, Fluid Bed Systemsm, 1-19.
Goodhart et al., An Evaluation of Aqueous Film-forming Dispersions for Controlled Release, Pharmaceutical Technology, Apr. 1984.
Greenhill et al., A Pharmacokinetic/Pharmacodynamic Study Comparing a Single Morning Dose of Adderall to Twice-Daily Dosing in Children with ADHD. J. Am. Acad. Adolesc. Psychiatry, 42:10, Oct. 2003.
Guidance for Industry: Extended Release Oral Dosage Forms: Development, Evaluation, and Application of In Vitro/In Vivo Correlations (1997).
Guidance for Industry: Food-Effect Bioavailability and Fed Bioequivalence Studies (2002).
Guidance for Industry: SUPAC-MR: Modified Release Solid Oral Dosage Forms (1997).
Guo Deposition Transcript, Jan. 24, 2005.
Guo Deposition Transcript, Jul. 26, 2004.
Hall HS and Pondell RE, Controlled Release Technologies: Methods, Theory, and Applications, pp. 133-154 (Agis F. Kydonieus ed. 1980).
Handbook of Pharmaceutical Excipients: Ethylcellulose, Polymethacrylates, (4th ed. (2003), 237-240, 462-468.
Handbook of Pharmaceutical Excipients: Plymethacrylates, (2nd ed. 1994), 361-366.
Hans-Martin Klein & Rolf W. Gunther, Double Contrast Small Bowel Follow-Through with an Acid-Resistant Effervescent Agent, Investigative Radiology vol. 28, Jul. 1993.
Harrington Deposition Transcript, Jul. 27, 2005.
Harris et al., Aqueous Polymeric Coating for Modified-Release Pellets, Acqueous Polymeric Coating for Pharmaceutical Dosage Forms (McGinity ed., 1989).
Holt, Bioequivalence Studies of Ketoprofen: Product formulation, Pharmacokinetics, Deconvolution, and In Vitro - In Vivo correlations, Thesis submitted to Oregon State University, Aug. (1997).
Husson et al., Influence of Size Polydispersity on Drug Release from Coated Pellets, International Journal of Pharmaceutics, 86 (1992) 113-121, 1992.
Impax Laboratories, Inc.'s First Amended Answer and Affirmative Defenses.
Impax Laboratories, Inc.'s First Supplemental Responses to Shire Laboratories Inc.'s First Set of Interrogatories (Nos. 11-12).
Impax Laboratories, Inc.'s Memorandum in Support of the Motion to Amend its Answer dated Feb. 25, 2005 and exhibits thereto.
Impax Laboratories, Inc.'s Reply Memorandum in Support of the Motion to Amend its Answer dated Mar. 18, 2005 and exhibits thereto.
Ishibashi et al., Design and Evaluatin of a New Capsule-type Dosage Form for Colon-Targeted Delivery of Drugs, International Journal of Pharmaceutics 168, (1998) 31-40, 1998.
J. Sjogren, Controlled release oral formulation technology, Rate Control in Drug Therapy, (1985) 38-47.
Jarowski, The Pharmaceutical Pilot Plant, Pharmaceutical Dosage Forms: Tablets, vol. 3, 2nd Ed. (1990), 303-367.
Judgement and Order of Permanent Injunction in Shire LLC v. Teva Pharmaceutical in the United States District Court for the Eastern District of Pennsylvania, Civil Action No. 06-952-SD, Mar. 6, 2008.
Judgment and Order of Permanaent Injunction in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Apr. 14, 2008.
Judgment and Order of Permanent in Shire Laboratories, Inc. and Shire, LLC v. Andrx Pharmaceuticals, LLC, et al., In the United States District Court for the Southern District of Florida, Miami Division, Case No. 07-22201-CIV-Cooke/Brown, Nov. 19, 2007.
Judgment and Order of Permanent Injunction in Shire Laboratories, Inc. v. Andrx Pharmaceuticals, LLC in the United States District Court for the Southern District of Florida, Miami Division, Case No. 07-22201-Civ-Cooke/Brown, Nov. 19, 2007.
Judgment and Order of Permanent Injunction in Shire LLC v. Teva Pharmaceutical Industries Ltd. in the United States District Court for the Eastern District of Pennsylvania, Civil Action No. 06-952-SD, Mar. 6, 2008.
Kao et al., Lag Time Method to Delay Drug Release to Various Sites in the Gastrointestinal Tract, Journal of Controlled Release 44(1997) 263-270.
Kennerly S. Patrick & John S. Markowitz, Pharmacology of Methylphenidate, Amphetamine Enantiomers and Permoline in Attention-Deficit Hyperactivity Disorder, Human Psychopharmacology, vol. 12, 527-546 (1997).
Kiriyama et al., The Bioavailability of Oral Dosage Forms of a New HIV-1 Protease Inhibitor, KNI-272, in Beagle Dogs, Biopharmaceutics & Drug Disposition, vol. 17 125-134 (1996).
Klaus Lehmann, Coating of Multiparticulates Using Polymeric Solutions, Multiparticulate Oral Drug Delivery (Swarbrick and Sellassie ed., 1994).
Krowczynski & Brozyna, Extended-Release Dosage Forms, pp. 123-131 (1987).
Leon Lachman, Herbert A. Lieberman, Joseph L. Kanig, The Theory and Practice of Industrial Pharmacy, Second Edition (1976) 371-373.
Leopold & Eikeler, Eudragit E as Coating Material for the pH-Controlled Drug Release in the Topical Treatment of Inflammatory Bowel Disease (IBD), Journal of Drug Targeting, 1998, vol. 6, No. 2, pp. 85-94.
Lin & Cheng, In-vitro Dissolution Behaviour of Spansule-type Micropellets Prepared by Pan Coating Method, Pharm. Ind. 51 No. 5 (1989).
Liu et al., Comparative Release of Phenylprepanolamine HCl from Long-Acting Appetite Suppressant Product: Acutrim vs. Dexatrim, Drug Development and Industrial Pharmacy, 10(10), 1639-1661 (1984).
Marcotte et al., Kinetics of Protein Diffusion from a Poly(D,L-Lactide) Reservoir System, Journal of Pharmaceutical Sciences, vol. 79, No. 5, May 1990.
Mathir et al., In vitro characterization of a controlled-release chlorpheniramine maleate delivery system prepared by the air-suspension technique, J. Microencapsulaton, vol. 14, No. 6, 743-751 (1997).
McGough et al., Pharmacokinetics of SL1381 (Adderall XR), an Extended-Release Formulation of Adderall, Journal of the American Academy of Child & Adolescent Psychiatry, vol. 42, No. 6, Jun. 2003.
McGraw-Hill Dictionary of Scientific and Technical Terms, 5th Ed. (1994), 97, 972.
McGuiness Deposition Transcript, Aug. 6, 2004.
Mehta et al., Evaluation of Fluid-bed Processes for Enteric Coating Systems, Pharmaceutical Technology, Apr. 1986.
Memorandum of Law in Support of Defendants' Motion for Certification for Immediate Appeal Pursuant to 28 U.S.C. § 1292(b) in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Jan. 17, 2008.
Memorandum of Law in Support of Defendants' Motion for Clarification or, in the Alternative for Reconsideration in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Jan. 16, 2008.
Modern Pharmaceutics, Banker, et al., eds., Marcel Dekker, Inc., New York, p. 350 (1996). *
Moller, Dissolution Testing of Delayed Release Preparations, Proceedings of the International Symposium held on Jan. 29-31, 1987 (The Bombay College of Pharmacy 1988), 85-111.
Neville et al., Disintegration of Dextran Sulfate Tablet Products: Effect of Physicochemical Properties, Drug Development and Industrial Pharmacy, New York, NY, vol. 18, No. 19, Jan. 1, 1992 (Jan. 1, 1992), pp. 2069-2079, XP009092848, ISSN: 0363-9045.
Office Action dated Dec. 6, 2007 in U.S. Appl. No. 11/091,011.
Office Action dated Jun. 22, 2007 in Japanese patent Application No. 2000-576830.
Office Action dated Jun. 22, 2007 in U.S. Appl. No. 11/091,011.
Office Action issued Jul. 24, 2008 in U.S. Appl. No. 11/091,011.
Office Action mailed Mar. 2, 2005 in European Patent Aplication No. 99 970594.0-2123.
Opening Expert Report of Dr. Michael Mayersohn and exhibits thereto, Mar. 12, 2005.
Opening Expert Report of Dr. Walter Chambliss and exhibits thereto, Mar. 15, 2005.
Order and Memorandum Denying Colony's Motion for Partial Summary Judgment of Noninfringement of the '819 and '300 Patents in Shire LLC v. Colony Pharmaceuticals, Inc., in the United States District Court for the District of Maryland, Case No. CCB-07-718, Jan. 2, 2008.
Ozturk et al., "Kinetics of Release from Enteric-Coated Tablets," Pharmaceutical Research, 1988;5:550-565.
Patrick et al., Pharmacology of Methylphenidate, Amphetamine Enantiomers and pemoline in Attention-Deficit Hyperactivity Disorder, Human Psychopharmacology, vol. 12, pp. 527-546 (1997).
PDR Drug Information for Ritalin LA Capsules, Apr. (2004).
Pelham et al., A Comparison of Morning-Only and Morning/Late Afternoon Adderall to Morning-Only, Twice-Daily, and Three Times-Daily Methylphenidate in Children with Attention-Deficit/Hyperactivity Disorder, Pediatrics, vol. 104, No. 6, Dec. 1999.
Petition Under Section 8 and exhibits thereto, submitted to the Canadian Patent Office on Dec. 4, 2006.
Physicians' Desk Reference: Adderall, 51st Ed. (1997).
Physicians' Desk Reference: Adderall, 56th Ed. (2002).
Physicians' Desk Reference: Dexedrine 56th Ed. (2002).
Physicians' Desk Reference: Ritalin, 56th Ed. (2002).
Plaintiff Shire LLC's Responses to Interrogatories Nos. 1-13 in Shire LLC v. Colony Parmaceuticals, Inc., in the United States District Court for the District of Maryland, Case No. 1:07-cv-00718-CCB, Jun. 6, 2007.
Plaintiff Shire LLC's Supplemental Responses to Interrogatory Nos. 1-5, 8, 9, & 12 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB, Aug. 22, 2007.
Plaintiff's Response to Defendants' First Set of Interrogatories in Shire Labatories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Oct. 11, 2006.
Plaintiff's Response to Defendants' First Set of Production Requests in Shire Laboratories v. Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Oct. 11, 2006.
Plaintiffs Shire Laboratories, Inc.'s and Shire LLC's Reply to Defendant Andrx Pharmaceuticals, LLC's Counterclaims in Shire Laboratories, Inc. and Shire, LLC v.Andrx Pharmaceuticals, LLC, et al., in the United States District Court for the Southern District of Florida, Miami Division, Case No. 07-22201-CIV-Cooke/Brown, Sep. 24, 2007.
Porter and Bruno Coating of Pharmaceutical Solid-Dosage Forms, 77-160.
Pozzi, et al., J. Controlled Release, vol. 31, pp. 99-108 (1994), The Time Clock System: a new oral dosage form for fast and complete release of drug after a predetermined lag time. *
Prescribing Information: Dexedrine, brand of dextroamphetamine sulfate (2001).
R. Bianchini & C. Vecchio, Oral Controlled Release Optimization of Pellets Prepared by Extrusion-Spheronization Processing, IL Farmaco 44(6), 645-654, 1989.
Rambali, et al., Using experimental design to optimize the process parameters in fluidized bed granulation on a semi-full scale, International Journal of Pharmaceutics 220 (2001) 149-160.
Remington: The Science and Practice of Pharmacy, Basic Pharmacokinetics, 16th Ed. (1980), 693.
Remington: The Science and Practice of Pharmacy, Elutriation, 20th Ed. (2000), 690.
Remington's Pharmaceutical Sciences, Fifteenth Edition (1975) 1624-1625.
Remington's Pharmaceutical Sciences, RPS XIV, 1700-1714.
Reply to Counterclaims in Shire Laboratories v Teva Pharmaceutical Industries Ltd., Case No. 2:06-cv-00952-SD, Aug. 16, 2006.
Rong-Kun Chang and Joseph R. Robinson, Sustained Drug Release from Tablets and Particles Through Coating, Pharmaceutical Dosage Forms: Tablets (Marcel Dekker, Inc. 1990), 199-302.
Rong-Kun Chang et al., Formulation Approaches for Oral Pulsatile Drug Delivery, American Pharmaceutical Review.
Rong-Kun Chang, A Comparison of Rheological and Enteric Properties among Organic Solutions, Ammonium Salt Aqueous Solutions, and Latex Systems of Some Enteric Polymers, Pharmaceutical Technology, Oct. 1990.
Rosen et al., Absorption and Excretion of Radioactively Tagged Dextroamphetamine Sulfate from a Sustained-Release Preparation, Jama, vol. 194, No. 11, Dec. 13, 1965. 145-147.
Rosen, et al., Absorption and Excretion of Radioactively Tagged Dextroamphetamine Sulfate From a Sustained-Release Preparation, Journal of the American Medical Association, Dec. 13, 1965, vol. 194, No. 11, 1203-1205.
Rudnic Deposition Transcript, Jul. 28, 2004.
Schaffer Deposition Transcript, Aug. 17, 2005.
Scheiffele, et al., Studies Comparing Kollicoat MAE 30 D with Commercial Cellulose Derivatives for Enteric Coating on Caffeine Cores, Drug Development and Industrial Pharmacy, 24(9), 807-818 (1998), 807-818.
Second Amended Complaint for Patent Infringement and Declaratory Relief in Shire Laboratories, Inc. and Shire, LLC v. Andrx Pharmaceuticals, LLC, et al., in the United States District Court for the Southern District of Florida, Miami Division, Case No. 07-22201-CIV-Cooke/Brown, Nov. 15, 2007.
Serajuddin, et al., Selection of Solid Dosage Form Composition through Drug-Excipient Compatibility Testing, Journal of Pharmaceutical Sciences vol. 88, No. 7, Jul. 1999, 696-704.
Shargel;. Pharmacokinetics of Oral Absorption, Applied Biopharmaceutics & Pharmacokinetics. 5th Ed. (2005), 164-166.
Sheen, et al., Aqueous Film Coating Studies of Sustained Release Nicotinic Acid Pellets: An In-Vitro Evaluation, Drug Development and Industrial Pharmacy, 18(8), 851-860 (1992).
Shire Laboratories Inc.'s Opposition to Barr Laboratories' Motion to Amend Its Answers and Counterclaims, Sep. 15, 2004.
Slattum, et al., Comparison of Methods for the Assessment of Central Nervous System Stimulant Response after Dextroamphetamine Adminstration to Healthy Male Volunteers, J. Clin Pharmacol 1996; 36: 1039-1050.
Snire Laboratory Inc's Complaint against Barr Laboratories based on Parent U.S. patent 6,322,815 in U S District Court for the Southern District of New York (Case No. 03-CV-1219(VM)(DFE)) 2003. *
Snire Laboratory Inc's Complaint against Barr Laboratories based on Parent U.S. patent 6,322,815 in U S District Court for the Southern District of New York (Case No. 03-CV-1219(VM)(DFE)) 2003. *
Sprowls' American Pharmacy: An Introduction to Pharmaceutical Techniques and Dosage Forms, 7th Ed. (1974), 387-388.
Sriamornsak, et al., Development of sustained release theophylline pellets coated with calcium pectinate, Journal of Controlled Release 47 (1997) 221-232.
Stevens, et al., Controlled, Multidose, Pharmacokinetic Evaluation of Two Extended-Release Carbamazepine Formulations (Carbatrol and Tegretol-XR), Journal of Pharmaceutical Sciences vol. 87, No. 12, Dec. 1998, 1531-1534.
Supplemental Expert Report of Harry G. Brittain, PhD, FRSC in Shire LLC v. Colony Pharaceuticals, Inc., in the United States District Court of the District of Maryland, Case No. 1:07-cv-00718, Feb. 15, 2008.
Supplemental Expert Report of Vladimir P. Torchilin, Ph.D., D.Sc. Regarding the Invalidity of U.S. Patent No. 6,605,300 in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Civil Action No. 1:07-cv-00718-CCB and exhibits thereto, Feb. 15, 2008.
Teva Notice letter: Feb. 21, 2005.
Teva Notice letter: Jun. 1, 2005.
The Merck Index: Amphetamine, 12th Ed., 620.
The Merck Index: Amphetamine, 13th Ed. (2001), 97, 1089.
The United States Pharmacopeia 23, National Formulary 18 (1995) pp. 1791-1799.
The United States Pharmacopeia 26, National Formulary 21 (2003) pp. 2157-2165.
The United States Pharmacopeia 27, National Formulary 22 (2004) pp. 2302-2312.
Transcript of Beth A. Burnside Deposition in Shire LLC v. Colony Pharmaceuticals, in the United States District Court for the District of Maryland, Case No. 1:07-cv-00718-CCB, Nov. 9, 2007.
Transcript of Beth A. Burnside Deposition in Shire LLC vs. Sandoz Inc. in the United States District Court for the District of Colorado, Case No. 07-CV-00197-EWN-CBS, Nov. 30, 2007.
Transcript of Kimberly Fiske Farrand Deposition in Shire, LLC v. Sandoz, Inc. in Shire, LLC v. Sandoz, Inc. in the United States District Court for the District of Colorado, Dec. 4, 2007.
Transcript of Richard A. Couch 30(b)(6) Deposition in Shire LLC vs. Sandoz Inc. in the United States District Court for the District of Colorado, Dec. 14, 2007.
Transcript of Richard A. Couch Deposition in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Case No. 1:07-cv-00718-CCB, Nov. 15, 2007.
Transcript of Richard Rong-Kun Chang in Shire LLC v. Colony Pharmaceuticals, Inc. in the United States District Court for the District of Maryland, Case No. 1:07-cv-00718-CCB, Nov. 20, 2007.
Treacy Deposition Transcript, Aug. 31, 2004.
Treatise on Controlled Drug Delivery, pp. 285-299 (Agis Kydonieus ed. 1992).
Tulloch, et al., SL1381 (Adderall XR), a Two-component, Extended-Release Formulation of Mixed Amphetamine Salts: Bioavailability of Three Test formulatons and Comparison of Fasted, Fed, and Sprinkled Administration, PHARMACOTHERAPY vol. 22, No. 11. (2002), 1405-1415.
U.S. Appl. No. 11/091,011, filed Apr. 7, 2009 Non-Final Office Action.
US 6,034,101, 3/2000, Gupta et al. (withdrawn)
Vasilevska, et al., Preparation and Dissolution Characteristics of Controlled Release Diltiazem Pellets, Drug Development and Industrial Pharmacy, 18(15), 1649-1661 (1992).
Walia, et al., Pharm. Dev. Tech., vol. 3, No. 1, pp. 103-113 (1998), Preliminary Evaluation of an Aqueous Wax Emulsion for Controlled-Release Coating. *
Watano, et al., Evaluation of Aqueous Enteric Coated Granules prepared by Moisture Control Method in Tumbling Fluidized Bed Process, Chem. Pharm. Bull. 42(3) 663-667 (1994).
Wesdyk, et al., Factors affecting differences in film thickness of beads coated in fluidized bed units, International Journal of Pharmaceutics, 93 101-109, (1993).
Wigal, et al., Evaluation of Infividual Subjects in the Analog Clasroom Setting; II. Effects of Dose of Amphetamine (Adderall), Psychopharmacology Bulletin, vol. 34, No. 4, pp. 833-838, 1998.
Wilding et al., Pharmaceutical Research, vol. 9, No. 5, pp. 654-657 (1992), Gastrointestinal Transit and Systemic Absorption of Captopil from Pulsed-Release Formulation. *
Wouessidjewe, Aqueous polymethacrylate Dispersions as Coating Materials for Sustained and Enteric Release Systems, S.T.P. Pharma Sciences 7(6) 469-475 (1997).
Xu, et al., Pharmaceutical Research, vol. 10, No. 8, pp. 1144-1152 (1993), Programmable Drug Delivery from an Erodible Association Polymer System. *

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9675704B2 (en) 2006-03-16 2017-06-13 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US10086087B2 (en) 2006-03-16 2018-10-02 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US9675703B2 (en) 2006-03-16 2017-06-13 Tris Pharma, Inc Modified release formulations containing drug - ion exchange resin complexes
US20070264323A1 (en) * 2006-05-12 2007-11-15 Shire Llc Controlled dose drug delivery system
US8846100B2 (en) 2006-05-12 2014-09-30 Shire Llc Controlled dose drug delivery system
US9173857B2 (en) 2006-05-12 2015-11-03 Shire Llc Controlled dose drug delivery system
US20090019148A1 (en) * 2007-07-13 2009-01-15 Britton Zachary E Method and apparatus for internet traffic monitoring by third parties using monitoring implements
US20090157875A1 (en) * 2007-07-13 2009-06-18 Zachary Edward Britton Method and apparatus for asymmetric internet traffic monitoring by third parties using monitoring implements
US20100024032A1 (en) * 2008-07-24 2010-01-28 Zachary Edward Britton Method and apparatus for effecting an internet user's privacy directive
US9009838B2 (en) 2008-07-24 2015-04-14 Front Porch, Inc. Method and apparatus for effecting an internet user's privacy directive
US9545399B2 (en) 2012-08-15 2017-01-17 Tris Pharma, Inc. Methylphenidate extended release chewable tablet
US11103494B2 (en) 2012-08-15 2021-08-31 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US11633389B2 (en) 2012-08-15 2023-04-25 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US9844544B2 (en) 2012-08-15 2017-12-19 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US9844545B2 (en) 2012-08-15 2017-12-19 Tris Pharma, Inc. Methylphenidate extended release chewable tablet
US11103495B2 (en) 2012-08-15 2021-08-31 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US10857143B2 (en) 2012-08-15 2020-12-08 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US10507203B2 (en) 2012-08-15 2019-12-17 Tris Pharma, Inc Methylphenidate extended release chewable tablet
US10363224B2 (en) 2013-03-13 2019-07-30 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules
US8889190B2 (en) 2013-03-13 2014-11-18 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
US10172878B2 (en) 2013-03-15 2019-01-08 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules
US9555005B2 (en) 2013-03-15 2017-01-31 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US10568841B2 (en) 2014-10-31 2020-02-25 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10688060B2 (en) 2014-10-31 2020-06-23 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10507186B2 (en) 2014-10-31 2019-12-17 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US11896722B2 (en) 2014-10-31 2024-02-13 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10512613B2 (en) 2014-10-31 2019-12-24 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10512612B2 (en) 2014-10-31 2019-12-24 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10292939B2 (en) 2014-10-31 2019-05-21 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10500162B2 (en) 2014-10-31 2019-12-10 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10292938B2 (en) 2014-10-31 2019-05-21 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10111839B2 (en) 2014-10-31 2018-10-30 Purdue Pharma Methods and compositions particularly for treatment of attention deficit disorder
US9974752B2 (en) 2014-10-31 2018-05-22 Purdue Pharma Methods and compositions particularly for treatment of attention deficit disorder
US10449159B2 (en) 2014-10-31 2019-10-22 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder
US10142086B2 (en) 2015-06-11 2018-11-27 At&T Intellectual Property I, L.P. Repeater and methods for use therewith
US11590228B1 (en) 2015-09-08 2023-02-28 Tris Pharma, Inc Extended release amphetamine compositions
US11590081B1 (en) 2017-09-24 2023-02-28 Tris Pharma, Inc Extended release amphetamine tablets
US10722473B2 (en) 2018-11-19 2020-07-28 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder

Also Published As

Publication number Publication date
US6322819B1 (en) 2001-11-27
US20030124188A1 (en) 2003-07-03
CA2348090A1 (en) 2000-04-27
EP1123087A4 (en) 2004-02-11
EP1123087A1 (en) 2001-08-16
HK1038701A1 (en) 2002-03-28
US20040219213A1 (en) 2004-11-04
AU1214500A (en) 2000-05-08
CA2348090C (en) 2004-04-13
DE69940673D1 (en) 2009-05-14
US20010055613A1 (en) 2001-12-27
JP2008303223A (en) 2008-12-18
JP2002527468A (en) 2002-08-27
ES2323910T3 (en) 2009-07-27
USRE42096E1 (en) 2011-02-01
US6605300B1 (en) 2003-08-12
WO2000023055A9 (en) 2000-08-31
WO2000023055A1 (en) 2000-04-27
EP1977736A1 (en) 2008-10-08
EP1123087B1 (en) 2009-04-01
ATE427101T1 (en) 2009-04-15

Similar Documents

Publication Publication Date Title
USRE41148E1 (en) Oral pulsed dose drug delivery system
JP6815358B2 (en) Timed pulse emission system
US20180344669A1 (en) Controlled dose drug delivery system
US8007827B2 (en) Pharmaceutical dosage forms having immediate release and/or controlled release properties
CA2651890C (en) Controlled dose drug delivery system
US20100080846A1 (en) Dipyridamole and acetylsalicylic acid formulations and process for preparing same
AU2006236052B2 (en) Oral pulsed dose drug delivery system
AU2004200325B2 (en) Oral Pulsed Dose Drug Delivery System
AU2013273835A1 (en) Timed, pulsatile release systems

Legal Events

Date Code Title Description
AS Assignment

Owner name: SHIRE LABORATORIES, INC.,MARYLAND

Free format text: RELEASE OF SECURITY INTEREST UNDER THE AMENDED AND RESTATED CREDIT AGREEMENT;ASSIGNOR:DLJ CAPITAL FUNDING, INC. AS ADMINISTRATIVE AGENT;REEL/FRAME:016914/0056

Effective date: 20040910

Owner name: SHIRE LABORATORIES, INC., MARYLAND

Free format text: RELEASE OF SECURITY INTEREST UNDER THE AMENDED AND RESTATED CREDIT AGREEMENT;ASSIGNOR:DLJ CAPITAL FUNDING, INC. AS ADMINISTRATIVE AGENT;REEL/FRAME:016914/0056

Effective date: 20040910

AS Assignment

Owner name: SHIRE LLC,KENTUCKY

Free format text: MERGER;ASSIGNOR:SHIRE LABORATORIES, INC.;REEL/FRAME:018648/0711

Effective date: 20061215

Owner name: SHIRE LLC, KENTUCKY

Free format text: MERGER;ASSIGNOR:SHIRE LABORATORIES, INC.;REEL/FRAME:018648/0711

Effective date: 20061215

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12

IPR Aia trial proceeding filed before the patent and appeal board: inter partes review

Free format text: TRIAL NO: IPR2017-00011

Opponent name: MYLAN PHARMACEUTICALS INC.,MYLAN INC. ANDMYLAN N.V

Effective date: 20161004