WO1998010762A2 - Controlled release dosage form of r-(z)-alpha-methoxyimino-alpha-(1-azabicyclo2.2oct-c-yl)acetonitrile monohydrochloride - Google Patents

Controlled release dosage form of r-(z)-alpha-methoxyimino-alpha-(1-azabicyclo2.2oct-c-yl)acetonitrile monohydrochloride Download PDF

Info

Publication number
WO1998010762A2
WO1998010762A2 PCT/GB1997/002418 GB9702418W WO9810762A2 WO 1998010762 A2 WO1998010762 A2 WO 1998010762A2 GB 9702418 W GB9702418 W GB 9702418W WO 9810762 A2 WO9810762 A2 WO 9810762A2
Authority
WO
WIPO (PCT)
Prior art keywords
dosage form
coated
beads
compound
polymer
Prior art date
Application number
PCT/GB1997/002418
Other languages
French (fr)
Other versions
WO1998010762A3 (en
Inventor
Susan Marie Milosovich
William Muldoon
James Albert Napper
Laurence Rousseau
Joseph Sauer
Original Assignee
Smithkline Beecham Plc
Smithkline Beecham Corporation
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
Priority to JP10513352A priority Critical patent/JP2001500150A/en
Application filed by Smithkline Beecham Plc, Smithkline Beecham Corporation filed Critical Smithkline Beecham Plc
Priority to CZ99832A priority patent/CZ83299A3/en
Priority to AU41288/97A priority patent/AU724086B2/en
Priority to PL97332074A priority patent/PL332074A1/en
Priority to HU9904401A priority patent/HUP9904401A3/en
Priority to NZ334268A priority patent/NZ334268A/en
Priority to IL12878197A priority patent/IL128781A0/en
Priority to CA002265661A priority patent/CA2265661A1/en
Priority to EP97939064A priority patent/EP0929301A2/en
Priority to BR9711734A priority patent/BR9711734A/en
Publication of WO1998010762A2 publication Critical patent/WO1998010762A2/en
Publication of WO1998010762A3 publication Critical patent/WO1998010762A3/en
Priority to NO991194A priority patent/NO991194L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1617Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, 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
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • 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
    • 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

  • the present invention relates to a novel formulation, and to its use in the treatment and/or prophylaxis of certain disorders.
  • [R-(Z)]- ⁇ -(methoxyimino)- ⁇ -(l-azabicyclo [2.2.2]oct-3-yl)acetonitrile monohydrochloride (compound X) and methods for its preparation are disclosed in EP-A-0392803, WO95/31456 and WO93/17018.
  • the compound enhances acetylcholine function via an action at muscarinic receptors within the central nervous system, and is therefore of potential use in the treatment and/or prophylaxis of dementia in mammals.
  • WO96/12486 discloses the use of compound X in the manufacture of a medicament for enhancing amyloid precursor protein processing along a non- amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease.
  • the present invention provides a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof.
  • controlled release any formulation technique wherein release of the active substance from the dosage form is modified to occur at a slower rate than that from an immediate release product, such as a conventional swallow tablet or capsule.
  • Controlled release includes delayed release wherein release of the active substance from the dosage form is modified to occur at a later time than that from a conventional immediate release product.
  • the subsequent release of active substance from a delayed release formulation may also be controlled to occur at a slower rate.
  • Controlled Drug Delivery Fundamentals and Applications, 2nd Edition. Eds. J.R. Robinson, V.H.L. Lee. Marcel Dekker Inc. New York 1987.
  • Such controlled release formulations are preferably formulated in a manner such that release of compound X is effected throughout the gastro-intestinal tract, and takes place predominantly over the first eight to twelve hours following ingestion.
  • Preferred formulations include wax matrices, swellable and/or gellable polymer or hydrogel matrices, tablets coated with release controlling polymers or waxes, and pellets, granules or beads comprising matrices or coated with release controlling polymers or waxes and then formulated as capsules, compressed tablets or suspensions.
  • Suitable waxes for matrix formation or release controlling coating include non- ionic beeswax derivatives such as Gelucire 62/05, 50/02 or 50/13 (Gattefosse), glyceryl behenate, other fatty acid mono-, di- or tri-esters of glycerol such as Precirol ATO5
  • microcrystalline wax hydrogenated castor oil or hydrogenated vegetable oil
  • long-chain aliphatic alcohols such as stearyl alcohol and carnuba wax.
  • Suitable materials for the formation of hydrogel matrices or swellable and/or gellable polymer matrices may be selected from alkyl celluloses, hydroxyalkylcelluloses, polyvinyl alcohol, polymethacrylatcs, polymethylmethacrylates, methacrylate/divinylbenzene copolymers, carboxymethylamide, polyoxyalkylenc glycols, polyvinyl pyrrolidone and carboxymethyl cellulose.
  • the swellable polymeric material in particular may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropylmethylcellulose, carboxymethylamide, potassium methacrylate/divinylbenzene copolymer, polymethylmethacrylate, crosslinked polyvinyipyrrolidone and high molecular weight polyvinyl alcohol.
  • the gellable polymeric material in particular may be selected from methylcellulose, carboxymethylcellulose, low-molecular weight hydroxypropylmethylcellulose, low-molecular weight polyvinylalcohols, polyoxyethyleneglycols and non-cross-linked polyvinyipyrrolidone.
  • the swellable and gellable polymeric material in particular may be selected from medium-viscosity hydroxypropylmethylcellulose and medium-viscosity polyvinylalcohols.
  • Release controlling polymers include hydrogel polymers such as those listed above, hydrophobic polymers and enteric, or pH dependent, polymers.
  • Suitable materials for the formation of hydrophobic release controlling polymer coatings include alkyl celluloses, which may be used in the form of latex suspensions such as Surelease (Colorcon) or Aquacoat (FMC), and methacrylic acid derivatives, which may be used in the form of latex suspensions such as Eudragit RS, RL and NE (Rohm).
  • Suitable materials for the formation of enteric or pH dependent polymer coatings include methacrylic acid derivatives, which may be used in the form of latex suspensions such as Eudragit L and S (Rohm).
  • Seal coats film layers used to separate the various functional layers of the formulation or to provide a final layer to the outside of the formulation, contain suitable materials for film forming such as alkylcelluloses, which may be used in the form of latex suspensions such as Surelease (Colorcon) or Aquacoat (FMC), and hydroxyalkycelluloses such as hydroxypropylmethylcellulose (for example Opadry (Colorcon)).
  • alkylcelluloses which may be used in the form of latex suspensions such as Surelease (Colorcon) or Aquacoat (FMC), and hydroxyalkycelluloses such as hydroxypropylmethylcellulose (for example Opadry (Colorcon)).
  • the formulation may also include plasticisers such as triethyl citrate, dibutyl sebacate or medium chain triglycerides in the release controlling polymer layer.
  • plasticisers such as triethyl citrate, dibutyl sebacate or medium chain triglycerides in the release controlling polymer layer.
  • Pellet-forming materials include suitable grades of microcrystalline cellulose such as Avicel PHlOl (FMC). Granules may be formed from any of the commonly used pharmaceutical fillers or diluents such as lactose, lactose monohydrate, mannitol, microcrystalline cellulose, dicalcium phosphate or search.
  • FMC Avicel PHlOl
  • Beads may be formed by layering or spraying on non-pareil seeds.
  • Suitable ingredients in controlled-release dosage forms include polyethylene glycol and propylene glycol and these, as well as the pharmaceutical fillers, may be used to modify the release rate by inclusion in matrices, pellets, granules or beads.
  • the formulation may also include hydrophobic excipients that retard the release from the formulation such as ethylcellulose, talc, colloidal silicon dioxide or glyceryl monostearate and/or one or more binders such as hydroxypropylmethylcellulose, microcrystalline cellulose or polyvinyipyrrolidone.
  • hydrophobic excipients that retard the release from the formulation such as ethylcellulose, talc, colloidal silicon dioxide or glyceryl monostearate and/or one or more binders such as hydroxypropylmethylcellulose, microcrystalline cellulose or polyvinyipyrrolidone.
  • wetting agents such as sodium lauryl sulphate, lubricants such as magnesium stearate and glidants such as colloidal silica may also be included.
  • a particularly preferred formulation comprises drug-layered beads coated with a release controlling polymer either alone or in combination with drug-layered beads not coated with a release controlling polymer (immediate release beads).
  • appropriate size non-pareil sugar beads may be layered with a solution or dispersion containing the active substance, inert excipients, and/or retardants such as ethylcellulose, talc, colloidal silicon dioxide or glyceryl monostearate and/or one or more binders such as hydroxypropylmethylcellulose or polyvinyipyrrolidone.
  • the layering of the active substance may be accomplished at a predetermined rate and temperature using either a coating pan or a fluid bed drier.
  • the layered beads may be seal coated with a suitable film forming polymer such as hydroxypropylmethylcellulose (e.g. Opadry) or Eudragit® L30D-55 (a methacrylic acid copolymer) and then may be coated with one or more suitable release controlling polymers preferably selected from from alkyl celluloses, hydroxyalkylcelluloses, sodium carboxymethyl cellulose and methacrylic acid derivatives, such as ethylcellulose, Eudragit® RS, Eudragit® RL or Methocel E4M, to produce beads that release compound X over an eight to twelve hour period and/or release compound X in one or more pulses. Seal coated beads may be used for an immediate release dose.
  • a suitable film forming polymer such as hydroxypropylmethylcellulose (e.g. Opadry) or Eudragit® L30D-55 (a methacrylic acid copolymer)
  • suitable release controlling polymers preferably selected from from alkyl celluloses, hydroxyalkylcelluloses
  • the controlled release or a mixture of controlled release and immediate release beads may then be filled into an appropriate size capsule or compressed with inert excipients into tablets of appropriate physical parameters such as shape, size, hardness and disintegration.
  • the polymer(s), release controlling plus any seal coat polymer(s), preferably make up 10 to 30% by weight of the total dosage form.
  • Plasticizer is normally present and may make up at least 2% by weight.
  • Binder(s) and retardant(s) typically make up to 3-10% by weight.
  • the polymer matrix is preferably a hydrogel polymer selected from alkyl celluloses such as methylcellulose, hydroxyalkylcelluloses such as hydroxypropylcellulose and hydroxypropylmethylcellulose, polyvinyl alcohol, polymethacrylates, cross-linked polyvinyipyrrolidone and sodium carboxymethyl cellulose.
  • the polymers typically make up 10 to 50% by weight of the tablet.
  • the matrix tablet can be sealed with a hydrophobic release controlling polymer coating such as ethylcellulose (Surelease (Colorcon)) to retard the hydration of the hydrogel matrix in the tablet.
  • the hydrophobic coating polymer typically make up 4 to 10% by weight of the tablet.
  • a particular aspect of the invention provides a system for the controlled release of an active substance which is compound X, its parent free base or any other pharmaceutically acceptable salt thereof, comprising (a) a deposit-core comprising an effective amount of the active substance and having defined geometric form, and (b) a support-platform applied to said deposit-core, wherein said deposit-core contains at least the active substance, and at least one member selected from the group consisting of (1) a polymeric material which swells on contact with water or aqueous liquids and a gellable polymeric material wherein the ratio of the said swellable polymeric material to said gellable polymeric material is in the range 1 :9 to 9:1 , and (2) a single polymeric material having both swelling and gelling properties, and wherein the support-platform is an elastic support, applied to said deposit-core so
  • the swellable polymeric material in (1) may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropyl-methylcellulose, carboxy-methyl starch, potassium methacrylate/divinylbenzene copolymer, crosslinked polyvinyipyrrolidone and polyvinyl alcohol.
  • the gellable polymeric material in (1) may be selected from methylcellulose and non-cross-linked polyvinyipyrrolidone.
  • the support-platform may comprise; polymers such as polyhydroxypropylmethy] cellulose, polyvinyl alcohol, polyacrylate, polymethacrylate, polyhydroxpropyl cellulose and polysodium carboxymethylcellulose; plasticizers such as polyoxyethylene glycols, castor oil, hydrogenated cator oil, ethyl phthalate, butyl phthalate, natural glycerides, synthetic glycerides and semisynthetic glycerides; binders such as polyvinyipyrrolidone, methylcellulose, ethyl cellulose gum arabic and alginic acid; hydrophilic agents such as mannitol, lactose, starch and colloidal silica; and/or hydrophobic agents such as hydrogenated castor oil, magnesium stearate, a fatty substance, wax, natural glycerides and synthetic glycerides.
  • plasticizers such as polyoxyethylene glycols, castor oil, hydrogenated cator oil, ethy
  • the polymer(s) typically make up 30 to 90% by weight of the support-platform, for example about 35 to 40%.
  • Plasticizer may make up at least 2% by weight of the support-platform, for example about 15 to 20%.
  • Binder(s), hydrophilic agent(s) and hydrophobic agent(s) typically total up to about 50%) by weight of the support-platform, for example about 40 to 50%.
  • a further aspect of the invention provides a system for the controlled-rate release of compound X, consisting of: a) a deposit-core comprising effective amounts of compound X and having defined geometric form, b) a support-platform applied to said deposit-core wherein said deposit-core contains, mixed with the active substance, at least one member selected from the group consisting of a (a) 5-80%) by weight of the total weight of deposit-core of a polymeric material having a high degree of swelling on contact with water or aqueous liquids and 90-10% by weight of the total weight of the deposit core of a gellable polymeric material, and (b) a single polymeric material having both swelling and gelling properties, and other adjuvants able to provide the mixture with suitable characteristics for compression and for intake of water, and wherein said support-platform consists of a polymeric material insoluble in
  • the swellable polymeric material in (a) may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropyl-methylcellulose, carboxy-methylamide, potassium methacrylate/divinylbenzene copolymer, polymethylmethacrylate, crosslinked polyvinyipyrrolidone and high molecular weight polyvinyl alcohol.
  • the gellable polymeric material in (a) may be selected from methylcellulose, carboxymethylcellulose, low-molecular weight hydroxypropylmethylcellulose, low-molecular weight polyvinylalcohols, polyoxyethyleneglycols and non-cross-linked polyvinyipyrrolidone.
  • the swellable and gellable polymeric material in (b) may be selected from medium- viscosity hydroxypropylmethylcellulose and medium-viscosity polyvinylalcohols.
  • the support platform may comprise insoluble polymeric material selected from acrylates, cellulose, ethylcellulose, cellulose acetate-propionate, polyethylene, methacrylates, acrylic acid copolymers and high-molecular weight polyvinylalcohols.
  • Such formulation may be prepared as generally described in US 4,839,177.
  • WO 94/06416 discloses a yet further alternative controlled release formulations suitable for use in the present invention.
  • a yet further aspect of the invention provides a system for the controlled-rate release of compound X, consisting of a pharmaceutical compressed tablet capable of releasing compound X at different rates, consisting of three layers, wherein
  • a first layer contains compound X with immediate or controlled release formulation, composed of rapidly swelling and/or soluble and/or erodible polymeric substances by contact with aqueous fluids, and adjuvants;
  • a second layer contains compound X, either equal to or different from those of the first layer, with slow release formulation, composed of swelling and/or gellable and/or erodible polymeric substances by contact with aqueous fluids, and adjuvants;
  • a low-permeability barrier-type layer coating said second layer or, alternatively, placed between the first and second layer, consisting of polymeric substances, adjuvants, plasticizing agents and, if necessary, compound X.
  • the polymeric substances of the first layer may be selected from cross-linked polyvinyipyrrolidone, low- and medium-molecular-weight hydroxypropyl cellulose and hydroxypropyl methylcellulose, cross-linked sodium carboxymethylcellulose, carboxymethyl starch, potassium methacrylate-divinylbenzene copolymer, polyvinyl alcohols, starches, starch derivatives, microcrystalline cellulose and cellulose derivatives, ⁇ -cyclodextrin and dextrin derivatives.
  • the polymeric substances of the second layer may be selected from the group consisting of hydroxypropyl methylcellulose having molecular weight from 1 ,000 to 4,000,000, hydroxypropyl cellulose having molecular weight from 2,000 to 2,000,000, carboxyvinyl polymers, polyvinyl alcohols, glucans, scleroglucans, mannans, xanthans, alginic acid and derivatives thereof, carboxymethylcellulose and derivatives thereof, poly(methyl vinyl ethers/maleic anhydride), ethylcellulose, methylcellulose, and cellulose derivatives.
  • the adjuvants of the first and second layers may be selected from the group consisting of starch, pregelled starch, calcium phosphate, mannitol, lactose, saccharose, glucose, sorbitol, microcrystalline cellulose, gelatin, polyvinyipyrrolidone, methylcellulose, starch solution, ethylcellulose, arabic gum, tragacanth gum, magnesium stearate, stearic acid, colloidal silica, glyceryl monostearate, hydrogenated castor oil, waxes, and mono-, bi-, and trisubstituted glycerides.
  • the polymeric substances of the barrier type layer may be selected from the group consisting of hydroxypropyl methylcellulose having molecular weight from 1,000 to 4,000,000, hydroxypropyl cellulose having molecular weight from 2,000 to 2,000,000, carboxyvinyl polymers, polyvinyl alcohols, glucans, scleroglucans, mannans, xanthans, carboxymethylcellulose, ethylcellulose, and methylcellulose.
  • the adjuvants of the barrier-type layer may be selected from the group consisting of glyceryl monostearate, semisynthetic glycerides, glyceryl palmitostearate, glyceryl behenate, polyvinyipyrrolidone, gelatine, ethylcellulose, methylcellulose, sodium carboxymethylcellulose, magnesium stearate, stearic acid, sodium stearate, talc, sodium benzoate, boric acid, and colloidal silica.
  • the plasticizing agents of the barrier-type layer may be selected from the group consisting of hydrogenated castor oil, fatty acids, substituted triglycerides and glycerides, polyoxyethylene glycols and derivatives thereof having molecular weight from 400 to 60,000.
  • Such formulation may be prepared as generally described in WO 94/06416.
  • the dosage form preferably contains compound X itself.
  • Compound X has active doses around 5-125 microgramme ( ⁇ g) (calculated as free base). It has been found through administration to human patients that efficacy as a cognition enhancer may be obtained at daily doses below O.Olmg/kg more particularly 0.003mg/kg and below, for example 0.000 l-0.003mg/kg, such as 0.00035-0.003mg/kg, 0.0007-0.003mg/kg, 0.000 l-0.0007mg/kg or 0.00035-0.002mg/kg.
  • Suitable unit doses to achieve such daily doses are 5, 12.5, 25, 50 or 75 ⁇ g, administered twice daily or 50 ⁇ g or lOO ⁇ g, once daily. Such unit doses are calculated on the basis of 50-70kg individuals and as free base.
  • the in vitro release profile of the dosage form i.e. the amount of compound X released over time will be selected so that it will provide an area under the in vivo plasma profile curve that is similar to that obtained following conventional oral administration of a fast release tablet, 5 to 75 ⁇ g (calculated as free base) compound X twice a day.
  • the dosage form of the invention may be used in the treatment and or prophylaxis of dementia, including Alzheimer's disease, in mammals, and for enhancing amyloid precursor protein processing along a non-amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease. These disorders are herein after referred to as "the Disorders”.
  • the present invention provides a method of treating "the Disorders" by administering an effective amount of a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof, to a sufferer in need thereof.
  • the present invention further provides the use of a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating "the Disorders".
  • the present invention also provides a pharmaceutical composition for use in the treatment of "the disorders" which comprises a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof.
  • pellets are produced by extrusion/spheronization, using water as a granulation liquid and an appropriate size fraction is obtained by screening. Pellets are then coated in a fluid bed coater (bottom spray) with 2-10% (w/w) of an aqueous Surelease dispersion (15% solids in dispersion).
  • Pellets are produced by extrusion/spheronization using water and sodium laurylsulphate as a granulation liquid, and an appropriate size fraction is obtained by screening. Pellets may additionally be coated in a fluid bed coater (bottom spray) with aqueous polymer dispersions to further reduce release rates and obtain the desired release profiles.
  • Example 5 Hydrophilicity
  • Tablets may be prepared by the following procedure: 1. Blend the starch and HPC in a high shear mixer 2. Dissolve the drug into a small quantity of water and spray into blend while mixing
  • Tablets may be prepared by the following procedure:
  • Tablets may be prepared as described in US5433123
  • Example 9 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used:
  • Seal coating solution A solution of Opadry® Clear (YS- 1-9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Polymer Coating A polymer coating dispersion containing ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at an 10% to 25% weight gain, in particular 10, 12, 15, 17, 22 and 25%.
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh non- pareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads.
  • a portion of the immediate release beads were polymer coated to a weight gain of 10% to 25% with the Surelease® coating dispersion.
  • the final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
  • Seal coating A seal coating dispersion containing Eudragit® L30D-55 of the following composition was made and used for seal coating the drug layered beads at an 4% weight gain.
  • Purified water q.s. Total 100 Polymer Coating A polymer coating dispersion containing ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at an 10% to 25% weight gain.
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Eudragit® L30D-55 seal coating dispersion to a weight gain of 4% to produce the immediate release beads.
  • a portion of the immediate release beads were polymer coated to a weight gain of 10% tp 25% with the Surelease® coating dispersion.
  • the final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
  • Seal coating solution A solution of Opadry® Clear (YS-1-9025A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Polymer Coating A polymer coating dispersion containing Ethylcellulose
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads.
  • a portion of the immediate release beads were polymer coated to a weight gain of 10% with the Aquacoat® coating dispersion.
  • the final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
  • Seal coating solution A solution of Opadry® Clear (YS-1 -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Polymer Coating A polymer coating dispersion containing Eudragit® RS or RS/RL of the following composition was made and used for polymer coating the seal coated beads at an 10% weight gain.
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads.
  • a portion of the immediate release beads were polymer coated to a weight gain of 10% with the Eudragit® RS or RS/RL coating dispersion.
  • the final polymer coated beads can be produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
  • Example 13 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used.
  • a medicated layer solution of the following composition was used:
  • Seal coating solution A solution of Opadry® Clear (YS- 1-7006) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Example 14 (Ethylcellulose coated beads with a retardant) 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used.
  • a medicated layer solution of the following composition was used: Component % w/w Function
  • Seal coating solution A solution of Opadry® Clear (YS- 1-9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Polymer Coating A polymer coating dispersion containing Ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at 10% weight gain.
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads.
  • a portion of the immediate release beads were polymer coated to a weight gain of 10% with the Surelease® coating dispersion.
  • the final polymer coated beads can be produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
  • Table 5 Release Profile of Ethylcellulose Coated Beads, with Retardant, of Compound X in Water
  • Seal coating solution A solution of Opadry® Clear (YS-l -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads.
  • the drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads.
  • a portion of the immediate release beads were enteric coated to a weight gain of 20% with the Eudragit® enteric coating dispersion.
  • the final enteric coated beads were produced by seal coating the enteric coated beads to a weight gain of 2% with the Opadry Clear seal coating solution.
  • Example 16 (matrix tablet) Ingredient mg/tablet Function
  • Seal coating solution A solution of Opadry® Clear (YS-l -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
  • Polymer Coating A polymer coating dispersion containing Ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at 10% weight gain.

Abstract

A controlled release formulation of an acetonitrile compound and its use in the treatment and/or prophylaxis of certain disorders.

Description

CONTROLLED RELEASE DOSAGE FORM OF [R-(Z)]-ALPHA-(METHOXYIMTNO)-ALPHA-( 1 - AZABICYCLO[2.2.2]OCT-3-YL)ACETONlTRILE MONOHYDROCHLORroE
The present invention relates to a novel formulation, and to its use in the treatment and/or prophylaxis of certain disorders. [R-(Z)]-α-(methoxyimino)-α-(l-azabicyclo [2.2.2]oct-3-yl)acetonitrile monohydrochloride (compound X) and methods for its preparation are disclosed in EP-A-0392803, WO95/31456 and WO93/17018. The compound enhances acetylcholine function via an action at muscarinic receptors within the central nervous system, and is therefore of potential use in the treatment and/or prophylaxis of dementia in mammals. WO96/12486 discloses the use of compound X in the manufacture of a medicament for enhancing amyloid precursor protein processing along a non- amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease.
Fast-release swallow tablet and oral solution formulations of compound X both result in rapid absorption of the compound into the circulation, and require twice a day dosing for optimal efficacy.
It has now been surprisingly found that it is possible to formulate compound X, which has very high water solubility and is active at extremely low doses, in such a way that release is controlled to take place over a period of hours. Such a formulation would require dosing only once a day: this is likely to improve compliance in a patient population characterised by poor memory; it may also reduce side-effects in case of accidental overdosing.
Accordingly, the present invention provides a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof.
By controlled release is meant any formulation technique wherein release of the active substance from the dosage form is modified to occur at a slower rate than that from an immediate release product, such as a conventional swallow tablet or capsule.
Controlled release includes delayed release wherein release of the active substance from the dosage form is modified to occur at a later time than that from a conventional immediate release product. The subsequent release of active substance from a delayed release formulation may also be controlled to occur at a slower rate.
Examples of controlled release formulations which are suitable for incorporating compound X are described in: Sustained Release Medications, Chemical Technology Review No. 177. Ed. J.C.
Johnson. Noyes Data Corporation 1980.
Controlled Drug Delivery, Fundamentals and Applications, 2nd Edition. Eds. J.R. Robinson, V.H.L. Lee. Marcel Dekker Inc. New York 1987. Such controlled release formulations are preferably formulated in a manner such that release of compound X is effected throughout the gastro-intestinal tract, and takes place predominantly over the first eight to twelve hours following ingestion.
Preferred formulations include wax matrices, swellable and/or gellable polymer or hydrogel matrices, tablets coated with release controlling polymers or waxes, and pellets, granules or beads comprising matrices or coated with release controlling polymers or waxes and then formulated as capsules, compressed tablets or suspensions.
Suitable waxes for matrix formation or release controlling coating include non- ionic beeswax derivatives such as Gelucire 62/05, 50/02 or 50/13 (Gattefosse), glyceryl behenate, other fatty acid mono-, di- or tri-esters of glycerol such as Precirol ATO5
(Gattefosse), microcrystalline wax, hydrogenated castor oil or hydrogenated vegetable oil, long-chain aliphatic alcohols such as stearyl alcohol and carnuba wax.
Suitable materials for the formation of hydrogel matrices or swellable and/or gellable polymer matrices may be selected from alkyl celluloses, hydroxyalkylcelluloses, polyvinyl alcohol, polymethacrylatcs, polymethylmethacrylates, methacrylate/divinylbenzene copolymers, carboxymethylamide, polyoxyalkylenc glycols, polyvinyl pyrrolidone and carboxymethyl cellulose. The swellable polymeric material in particular may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropylmethylcellulose, carboxymethylamide, potassium methacrylate/divinylbenzene copolymer, polymethylmethacrylate, crosslinked polyvinyipyrrolidone and high molecular weight polyvinyl alcohol. The gellable polymeric material in particular may be selected from methylcellulose, carboxymethylcellulose, low-molecular weight hydroxypropylmethylcellulose, low-molecular weight polyvinylalcohols, polyoxyethyleneglycols and non-cross-linked polyvinyipyrrolidone. The swellable and gellable polymeric material in particular may be selected from medium-viscosity hydroxypropylmethylcellulose and medium-viscosity polyvinylalcohols.
Release controlling polymers include hydrogel polymers such as those listed above, hydrophobic polymers and enteric, or pH dependent, polymers. Suitable materials for the formation of hydrophobic release controlling polymer coatings include alkyl celluloses, which may be used in the form of latex suspensions such as Surelease (Colorcon) or Aquacoat (FMC), and methacrylic acid derivatives, which may be used in the form of latex suspensions such as Eudragit RS, RL and NE (Rohm). Suitable materials for the formation of enteric or pH dependent polymer coatings include methacrylic acid derivatives, which may be used in the form of latex suspensions such as Eudragit L and S (Rohm). Seal coats, film layers used to separate the various functional layers of the formulation or to provide a final layer to the outside of the formulation, contain suitable materials for film forming such as alkylcelluloses, which may be used in the form of latex suspensions such as Surelease (Colorcon) or Aquacoat (FMC), and hydroxyalkycelluloses such as hydroxypropylmethylcellulose (for example Opadry (Colorcon)).
The formulation may also include plasticisers such as triethyl citrate, dibutyl sebacate or medium chain triglycerides in the release controlling polymer layer.
Pellet-forming materials include suitable grades of microcrystalline cellulose such as Avicel PHlOl (FMC). Granules may be formed from any of the commonly used pharmaceutical fillers or diluents such as lactose, lactose monohydrate, mannitol, microcrystalline cellulose, dicalcium phosphate or search.
Beads may be formed by layering or spraying on non-pareil seeds.
Other suitable ingredients in controlled-release dosage forms include polyethylene glycol and propylene glycol and these, as well as the pharmaceutical fillers, may be used to modify the release rate by inclusion in matrices, pellets, granules or beads.
The formulation may also include hydrophobic excipients that retard the release from the formulation such as ethylcellulose, talc, colloidal silicon dioxide or glyceryl monostearate and/or one or more binders such as hydroxypropylmethylcellulose, microcrystalline cellulose or polyvinyipyrrolidone.
Wetting agents such as sodium lauryl sulphate, lubricants such as magnesium stearate and glidants such as colloidal silica may also be included.
A particularly preferred formulation comprises drug-layered beads coated with a release controlling polymer either alone or in combination with drug-layered beads not coated with a release controlling polymer (immediate release beads). In the drug layering process onto non-pareil beads, appropriate size non-pareil sugar beads may be layered with a solution or dispersion containing the active substance, inert excipients, and/or retardants such as ethylcellulose, talc, colloidal silicon dioxide or glyceryl monostearate and/or one or more binders such as hydroxypropylmethylcellulose or polyvinyipyrrolidone. The layering of the active substance may be accomplished at a predetermined rate and temperature using either a coating pan or a fluid bed drier. The layered beads may be seal coated with a suitable film forming polymer such as hydroxypropylmethylcellulose (e.g. Opadry) or Eudragit® L30D-55 (a methacrylic acid copolymer) and then may be coated with one or more suitable release controlling polymers preferably selected from from alkyl celluloses, hydroxyalkylcelluloses, sodium carboxymethyl cellulose and methacrylic acid derivatives, such as ethylcellulose, Eudragit® RS, Eudragit® RL or Methocel E4M, to produce beads that release compound X over an eight to twelve hour period and/or release compound X in one or more pulses. Seal coated beads may be used for an immediate release dose. The controlled release or a mixture of controlled release and immediate release beads may then be filled into an appropriate size capsule or compressed with inert excipients into tablets of appropriate physical parameters such as shape, size, hardness and disintegration. The polymer(s), release controlling plus any seal coat polymer(s), preferably make up 10 to 30% by weight of the total dosage form. Plasticizer is normally present and may make up at least 2% by weight. Binder(s) and retardant(s) typically make up to 3-10% by weight.
Another particularly preferred formulation comprises a swellable and/or gellable polymer matrix tablet. The polymer matrix is preferably a hydrogel polymer selected from alkyl celluloses such as methylcellulose, hydroxyalkylcelluloses such as hydroxypropylcellulose and hydroxypropylmethylcellulose, polyvinyl alcohol, polymethacrylates, cross-linked polyvinyipyrrolidone and sodium carboxymethyl cellulose. The polymers typically make up 10 to 50% by weight of the tablet. The matrix tablet can be sealed with a hydrophobic release controlling polymer coating such as ethylcellulose (Surelease (Colorcon)) to retard the hydration of the hydrogel matrix in the tablet. The hydrophobic coating polymer typically make up 4 to 10% by weight of the tablet.
Such matrix tablet formulations can be prepared by either direct compression or wet granulation processes. Coating may be accomplished using a coating pan. Other preferred formulations are described in US Patent No. 5,422,123. Thus, a particular aspect of the invention provides a system for the controlled release of an active substance which is compound X, its parent free base or any other pharmaceutically acceptable salt thereof, comprising (a) a deposit-core comprising an effective amount of the active substance and having defined geometric form, and (b) a support-platform applied to said deposit-core, wherein said deposit-core contains at least the active substance, and at least one member selected from the group consisting of (1) a polymeric material which swells on contact with water or aqueous liquids and a gellable polymeric material wherein the ratio of the said swellable polymeric material to said gellable polymeric material is in the range 1 :9 to 9:1 , and (2) a single polymeric material having both swelling and gelling properties, and wherein the support-platform is an elastic support, applied to said deposit-core so that it partially covers the surface of the deposit-core and follows changes due to hydration of the deposit-core and is slowly soluble and/or slowly gellable in aqueous fluids. The swellable polymeric material in (1) may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropyl-methylcellulose, carboxy-methyl starch, potassium methacrylate/divinylbenzene copolymer, crosslinked polyvinyipyrrolidone and polyvinyl alcohol. The gellable polymeric material in (1) may be selected from methylcellulose and non-cross-linked polyvinyipyrrolidone.
The support-platform may comprise; polymers such as polyhydroxypropylmethy] cellulose, polyvinyl alcohol, polyacrylate, polymethacrylate, polyhydroxpropyl cellulose and polysodium carboxymethylcellulose; plasticizers such as polyoxyethylene glycols, castor oil, hydrogenated cator oil, ethyl phthalate, butyl phthalate, natural glycerides, synthetic glycerides and semisynthetic glycerides; binders such as polyvinyipyrrolidone, methylcellulose, ethyl cellulose gum arabic and alginic acid; hydrophilic agents such as mannitol, lactose, starch and colloidal silica; and/or hydrophobic agents such as hydrogenated castor oil, magnesium stearate, a fatty substance, wax, natural glycerides and synthetic glycerides. The polymer(s) typically make up 30 to 90% by weight of the support-platform, for example about 35 to 40%. Plasticizer may make up at least 2% by weight of the support-platform, for example about 15 to 20%. Binder(s), hydrophilic agent(s) and hydrophobic agent(s) typically total up to about 50%) by weight of the support-platform, for example about 40 to 50%.
Such formulation may be prepared as generally described in US 5,422,123. US-A-4 839 177 discloses a further alternative controlled release formulations suitable for use in the present invention. Thus a further aspect of the invention provides a system for the controlled-rate release of compound X, consisting of: a) a deposit-core comprising effective amounts of compound X and having defined geometric form, b) a support-platform applied to said deposit-core wherein said deposit-core contains, mixed with the active substance, at least one member selected from the group consisting of a (a) 5-80%) by weight of the total weight of deposit-core of a polymeric material having a high degree of swelling on contact with water or aqueous liquids and 90-10% by weight of the total weight of the deposit core of a gellable polymeric material, and (b) a single polymeric material having both swelling and gelling properties, and other adjuvants able to provide the mixture with suitable characteristics for compression and for intake of water, and wherein said support-platform consists of a polymeric material insoluble in aqueous liquids and partially coating said deposit core.
The swellable polymeric material in (a) may be selected from crosslinked sodium carboxymethylcellulose, crosslinked hydroxypropylcellulose, high molecular weight polyhydroxypropyl-methylcellulose, carboxy-methylamide, potassium methacrylate/divinylbenzene copolymer, polymethylmethacrylate, crosslinked polyvinyipyrrolidone and high molecular weight polyvinyl alcohol. The gellable polymeric material in (a) may be selected from methylcellulose, carboxymethylcellulose, low-molecular weight hydroxypropylmethylcellulose, low-molecular weight polyvinylalcohols, polyoxyethyleneglycols and non-cross-linked polyvinyipyrrolidone. The swellable and gellable polymeric material in (b) may be selected from medium- viscosity hydroxypropylmethylcellulose and medium-viscosity polyvinylalcohols. The support platform may comprise insoluble polymeric material selected from acrylates, cellulose, ethylcellulose, cellulose acetate-propionate, polyethylene, methacrylates, acrylic acid copolymers and high-molecular weight polyvinylalcohols.
Such formulation may be prepared as generally described in US 4,839,177.
WO 94/06416 discloses a yet further alternative controlled release formulations suitable for use in the present invention.
Thus a yet further aspect of the invention provides a system for the controlled-rate release of compound X, consisting of a pharmaceutical compressed tablet capable of releasing compound X at different rates, consisting of three layers, wherein
- a first layer contains compound X with immediate or controlled release formulation, composed of rapidly swelling and/or soluble and/or erodible polymeric substances by contact with aqueous fluids, and adjuvants;
- a second layer contains compound X, either equal to or different from those of the first layer, with slow release formulation, composed of swelling and/or gellable and/or erodible polymeric substances by contact with aqueous fluids, and adjuvants; - a low-permeability barrier-type layer coating said second layer or, alternatively, placed between the first and second layer, consisting of polymeric substances, adjuvants, plasticizing agents and, if necessary, compound X.
The polymeric substances of the first layer may be selected from cross-linked polyvinyipyrrolidone, low- and medium-molecular-weight hydroxypropyl cellulose and hydroxypropyl methylcellulose, cross-linked sodium carboxymethylcellulose, carboxymethyl starch, potassium methacrylate-divinylbenzene copolymer, polyvinyl alcohols, starches, starch derivatives, microcrystalline cellulose and cellulose derivatives, β-cyclodextrin and dextrin derivatives.
The polymeric substances of the second layer may be selected from the group consisting of hydroxypropyl methylcellulose having molecular weight from 1 ,000 to 4,000,000, hydroxypropyl cellulose having molecular weight from 2,000 to 2,000,000, carboxyvinyl polymers, polyvinyl alcohols, glucans, scleroglucans, mannans, xanthans, alginic acid and derivatives thereof, carboxymethylcellulose and derivatives thereof, poly(methyl vinyl ethers/maleic anhydride), ethylcellulose, methylcellulose, and cellulose derivatives.
The adjuvants of the first and second layers may be selected from the group consisting of starch, pregelled starch, calcium phosphate, mannitol, lactose, saccharose, glucose, sorbitol, microcrystalline cellulose, gelatin, polyvinyipyrrolidone, methylcellulose, starch solution, ethylcellulose, arabic gum, tragacanth gum, magnesium stearate, stearic acid, colloidal silica, glyceryl monostearate, hydrogenated castor oil, waxes, and mono-, bi-, and trisubstituted glycerides.
The polymeric substances of the barrier type layer may be selected from the group consisting of hydroxypropyl methylcellulose having molecular weight from 1,000 to 4,000,000, hydroxypropyl cellulose having molecular weight from 2,000 to 2,000,000, carboxyvinyl polymers, polyvinyl alcohols, glucans, scleroglucans, mannans, xanthans, carboxymethylcellulose, ethylcellulose, and methylcellulose.
The adjuvants of the barrier-type layer may be selected from the group consisting of glyceryl monostearate, semisynthetic glycerides, glyceryl palmitostearate, glyceryl behenate, polyvinyipyrrolidone, gelatine, ethylcellulose, methylcellulose, sodium carboxymethylcellulose, magnesium stearate, stearic acid, sodium stearate, talc, sodium benzoate, boric acid, and colloidal silica.
The plasticizing agents of the barrier-type layer may be selected from the group consisting of hydrogenated castor oil, fatty acids, substituted triglycerides and glycerides, polyoxyethylene glycols and derivatives thereof having molecular weight from 400 to 60,000.
Such formulation may be prepared as generally described in WO 94/06416.
The dosage form preferably contains compound X itself. Compound X has active doses around 5-125 microgramme (μg) (calculated as free base). It has been found through administration to human patients that efficacy as a cognition enhancer may be obtained at daily doses below O.Olmg/kg more particularly 0.003mg/kg and below, for example 0.000 l-0.003mg/kg, such as 0.00035-0.003mg/kg, 0.0007-0.003mg/kg, 0.000 l-0.0007mg/kg or 0.00035-0.002mg/kg. Suitable unit doses to achieve such daily doses are 5, 12.5, 25, 50 or 75μg, administered twice daily or 50μg or lOOμg, once daily. Such unit doses are calculated on the basis of 50-70kg individuals and as free base.
Suitably, the in vitro release profile of the dosage form i.e. the amount of compound X released over time will be selected so that it will provide an area under the in vivo plasma profile curve that is similar to that obtained following conventional oral administration of a fast release tablet, 5 to 75 μg (calculated as free base) compound X twice a day. Preferably 25-70% is released over 4hours and 70-100% is released over 8 hours.
The dosage form of the invention may be used in the treatment and or prophylaxis of dementia, including Alzheimer's disease, in mammals, and for enhancing amyloid precursor protein processing along a non-amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease. These disorders are herein after referred to as "the Disorders".
The present invention provides a method of treating "the Disorders" by administering an effective amount of a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof, to a sufferer in need thereof.
The present invention further provides the use of a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating "the Disorders".
The present invention also provides a pharmaceutical composition for use in the treatment of "the disorders" which comprises a controlled release oral dosage form containing compound X, its parent free base or any other pharmaceutically acceptable salt thereof.
The following examples illustrate the present invention.
Examples
In the following examples, the weight shown is the weight of free base; compound X is the hydrochloride salt, (pfb = pure free base). Mesh sizes are US standard.
Example 1 (Wax matrix)
Compound X 0.005-0.1 mg pfb
Gelucire 62/05 (Gattefosse) 190 mg Propylene glycol 10 mg
Example 2 (Film coated pellets)
Component mg/capsule (500 mg) Function
Compound X 0.005-0.1 mg pfb Active
Lactose 300 Hydrophilic diluent
Avicel PH lOl (FMC) 200 Inert pellet matrix
Film coat: w/w of pellet cores
Surelease (Colorcon) 2 - 10% Release controlling polymer coat Silicone antifoam Antifoaming agent Example 3 (Film coated pellets)
Component mg/capsule (500 mg) Function
Compound X 0.005-0.1 mg pfb Active Lactose 400 Hydrophilic diluent Avicel PH 101 100 Inert pellet matrix
Film coat: w/w of pellet cores Aquacoat (FMC) 2 - 10% Release controlling polymer coat Silicone antifoam Antifoaming agent Di-butylsebacate 20 - 30% (of polymer Plasticizer weight)
In Examples 2 and 3, pellets are produced by extrusion/spheronization, using water as a granulation liquid and an appropriate size fraction is obtained by screening. Pellets are then coated in a fluid bed coater (bottom spray) with 2-10% (w/w) of an aqueous Surelease dispersion (15% solids in dispersion).
Desired release profiles are obtained by mixing uncoated (= immediate release pellets) and coated pellets of suitable coating levels (= sustained release pellets), that are then filled into hard gelatine capsules.
Example 4 (matrix pellets)
Component mg/capsule (500 mg) Function
Compound X 0.005-0.1 mg pfb Active Glyceryl behenate 200 Hydrophobic matrix Avicel PH 101 300 Inert pellet matrix Sodium lauryl sulphate 0.1 Wetting agent
Pellets are produced by extrusion/spheronization using water and sodium laurylsulphate as a granulation liquid, and an appropriate size fraction is obtained by screening. Pellets may additionally be coated in a fluid bed coater (bottom spray) with aqueous polymer dispersions to further reduce release rates and obtain the desired release profiles. Example 5 (Hydrogel matrix)
Excipient % w/w mg /tablet mg/tablet
Compound X 0.003-0.07pfb 0.005pfb O.lpfb
Hydroxypropylcellulose 25 37.5 37.5
Purified water -
Starch to 100 109.5 108.5
Magnesium stearate 2 3.0 3.0
Total 100 150 150
Tablets may be prepared by the following procedure: 1. Blend the starch and HPC in a high shear mixer 2. Dissolve the drug into a small quantity of water and spray into blend while mixing
3. Wash spray mechanism with small volume of water into blend while mixing
4. Granulate mix with sufficient water to achieve a medium to heavy granule
5. Partially dry granule
6. Screen through a suitable mill 7. Complete drying of milled granule
8. Lubricate with Mg stearate
9. Compress into tablets with a target weight of 150mg
Example 6 (Wax matrix)
Excipient % w/w mg /tablet mg/tablet
Compound X 0.003 to 0.07pf 0.005pfb O.lpfb
Lactose Anhydrous to 100 to 150 to 150
Gelucire 62/05 18 27.0 27.0
Magnesium stearate 2 3.0 3.0
Total 100 150 150
Tablets may be prepared by the following procedure:
1. Preblend the drug with a small quantity of lactose
2. Sandwich the drug preblend with the remaining lactose and the required % of Gelucire 62/05 in a preheated pelletiser. 3. Pelletise until the required pellet size has been achieved
4. Remove the pellets and allow them to cool
5. Screen pellets as necessary
6. Lubricate pellets
7. Compress or encapsulate pellets Example 7 (Controlled release bilaycr tablet)
Active Layer
Component mg/tablet Function
Compound X 0.005-0.1 mg pfb Active Hydroxypropylmethylcellulose 68.5 Hydrogel matrix former
Mannitol 20 Soluble filler
Ethyl cellulose (applied in ethanolic solution) 7.5 Binder
Magnesium stearate 2 Lubricant
Colloidol silica 2 Glidant
Support platform Component mg/tablet Function
Hydroxypropylmethylcellulose 39.75 Hydrogel matrix former
Hydrogenated castor oil 6.5 Insoluble filler
Ethylcellulose (applied in ethanolic solution) 2.5 Binder
Yellow iron oxide pigment 0.5 Pigment
Magnesium stearate 0.5 Lubricant
Colloidal silica 0.25 Glidant
Tablets may be prepared as described in US5433123
Example 8 (Wax matrix)
Component % w/w Function
Compound X 0.02pfb Active
Gelucire 50/02 91.5 Wax matrix
Gelucire 50/13 5 Wax matrix
Propylene glycol 1.98 Solvent
Colloidal silica 1.5 Hydrophobic excipient
Sodium dihydrogi sn citrate 0-1.5 Stabilizer
Process:
The Gelucire waxes were melted together at around 60 degrees C. Compound X was dissolved in propylene glycol, and blended into the waxes. The colloidal silica was then also blended in, and the mixture filled into size 3 hard gelatin capsule shells. Table 1.: Release Profile of wax-filled capsules of Compound X in water (0% citrate)
Time (hr) % Released
1 13
3 29
5 53
8 73
Example 9 (Ethylcellulose coated beads) 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used:
Component % w/w Function
Compound X 0.003-0.05pfb Active
Opadry® Clear 3 Binder
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS- 1-9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A polymer coating dispersion containing ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at an 10% to 25% weight gain, in particular 10, 12, 15, 17, 22 and 25%.
Component % w/w Function
Surelease® 60 (25% as solids) Release controlling polymer coat with plasticiser
Purified water q.s.
Total 100
Drug layered beads were produced by layering the drug solution onto 25-30 mesh non- pareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads. A portion of the immediate release beads were polymer coated to a weight gain of 10% to 25% with the Surelease® coating dispersion. The final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
Table 2. Release Profile Range of Ethylcellulose coated beads, 10-25% by weight of
Compound X in Water
Time (hr) % Released
1 0.8-36
2 5-57 4 13-75 8 18-91
Example 10 (Ethylcellulose coated beads)
200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used:
Component % w/w Function
Compound X 0.003-0.05pfb Active
Opadry® Clear 3 Binder
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating: A seal coating dispersion containing Eudragit® L30D-55 of the following composition was made and used for seal coating the drug layered beads at an 4% weight gain.
Component % w/w Function
Eudragit® L30D-55 45 (30% as solids) Polymeric seal coat
Triethyl citrate 2.02 Plasticizer
Talc 3.10 Anti-tack
Purified water q.s. Total 100 Polymer Coating: A polymer coating dispersion containing ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at an 10% to 25% weight gain.
Component % w/w Function
Surelease® 60 (25%) as solids) Release controlling polymer coat with plasticiser Purified water q.s.
Total 100
Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Eudragit® L30D-55 seal coating dispersion to a weight gain of 4% to produce the immediate release beads. A portion of the immediate release beads were polymer coated to a weight gain of 10% tp 25% with the Surelease® coating dispersion. The final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
Table 3 : Release Profile of Eudragit® L30D Seal Coated/Ethylcellulose
Coated Beads of Compound X in Water
Time (hr) % Released, 10% Surelease
0.5 1.5
1 5
2 20 4 39 6 49 8 56
Example 11 (Ethylcellulose coated beads)
200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used: Component % w/w Function
Compound X 0.003-0.05pfb Active
Opadry® Clear 3 Binder
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS-1-9025A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A polymer coating dispersion containing Ethylcellulose
(Aquacoat®) of the following composition was made and used for polymer coating the seal coated beads at a 10% weight gain.
Component % w/w Function
Aquacoat® 50 (30% as solids) Release controlling polymer coat
Triethyl Citrate 2.02 Plasticizer
Purified water q.s. Total 100
Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads. A portion of the immediate release beads were polymer coated to a weight gain of 10% with the Aquacoat® coating dispersion. The final polymer coated beads were produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
Example 12 (Eudragit coated beads)
200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used: Component % w/w Function
Compound X O.003-0.05pfb Active
Opadry® Clear 3 Binder
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS-1 -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A polymer coating dispersion containing Eudragit® RS or RS/RL of the following composition was made and used for polymer coating the seal coated beads at an 10% weight gain.
Component % w/w Function
Eudragit® RS 30D 45 (30% as solids) Release controlling polymer coat Triethyl citrate 2.02 Plasticizer Talc 3.10 Anti-tack
Purified water q.s. Total 100
or
Component % w/w Function
Eudragit® RS 30D 36 (30% as solids) Release controlling polymer coat
Eudragit® RL 30D 9 (30%) as solids) Release controlling polymer coat
Triethyl citrate 2.02 Plasticizer
Talc 3.10 Anti-tack
Purified water q.s.
Total 100
Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads. A portion of the immediate release beads were polymer coated to a weight gain of 10% with the Eudragit® RS or RS/RL coating dispersion. The final polymer coated beads can be produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution.
Table 4: Release Profile of Eudragit® RS/RL coated beads of Compound X in water
Time (hr) % Released
0.5 0.2
1 0.3
2 0.4 4 1.9 6 13 8 20
Example 13 (Methocel coated beads) 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used:
Component % w/w Function
Compound X 0.003-0.05pfb Active
Methocel E4M 15 Release controlling polymer coat
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS- 1-7006) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Example 14 (Ethylcellulose coated beads with a retardant) 200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used: Component % w/w Function
Compound X 0.003-0. 05pfb Active
Opadry® Clear 1.5 Binder
Surelease® 1.5 Retardant
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS- 1-9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A polymer coating dispersion containing Ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at 10% weight gain.
Component % w/w Function
Surelease® 60 (25% as solids) Release controlling polymer coat with plasticiser Purified water q.s.
Total 100
Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads. A portion of the immediate release beads were polymer coated to a weight gain of 10% with the Surelease® coating dispersion. The final polymer coated beads can be produced by seal coating the polymer coated beads to a weight gain of 2% with the Opadry® Clear seal coating solution. Table 5. Release Profile of Ethylcellulose Coated Beads, with Retardant, of Compound X in Water
ne (hr) % Released
Without Retardant With Retardant
0.5 12 8
1 37 22
2 57 35
4 73 48
6 85 53
8 58
Example 15 (Enteric coated beads)
200 mg of non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size may be used. A medicated layer solution of the following composition was used:
Component % w/w Function
Compound X 0.003-0.05pfb Active
Opadry® Clear 3 Binder
Sodium dihydrogen citrate 1.5 Stabilizer
Purified water q.s.
Total 100
Seal coating solution: A solution of Opadry® Clear (YS-l -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A nolymer coating dispersion containing Eudragit® L30D-55 of the following composition was made and used for polymer coating the seal coated beads at an 20% weight gain.
Component % w/w Function Eudragit L30D-55 45.00 (30% as solids) Enteric (pH dependent) polymer Triethyl citrate 2.02 Plasticizer Talc 3.10 Anti-tack
Purified water q.s. Total 100 Drug layered beads were produced by layering the drug solution onto 25-30 mesh nonpareil beads using a Niro STREA-1 fluid bed dryer so as to layer 100 micrograms of the drug as the free base onto 200 mg of the non-pareil beads. The drug layered beads were seal coated with Opadry® Clear seal coating solution to a weight gain of 3% to produce the immediate release beads. A portion of the immediate release beads were enteric coated to a weight gain of 20% with the Eudragit® enteric coating dispersion. The final enteric coated beads were produced by seal coating the enteric coated beads to a weight gain of 2% with the Opadry Clear seal coating solution.
Example 16 (matrix tablet) Ingredient mg/tablet Function
Compound X 0.005-0.1 pfb Active
Hydroxpropyl Methcellulose E4M CR 75.0 Hydrogel matrix
Sodium Dihydrogen Citrate 3.00 Stabilizer
Lactose, Fast Flo 70.38 Hydrophilic diluent
Magnesium Stearate 1.50 Lubicant
Opadry® White 2.25 Seal coat polymer
Seal coating solution: A solution of Opadry® Clear (YS-l -9025 A) in purified water at 10% solids concentrations was made by dissolving 100 grams of Opadry® Clear into 900 grams of purified water.
Polymer Coating: A polymer coating dispersion containing Ethylcellulose (Surelease®) of the following composition was made and used for polymer coating the seal coated beads at 10% weight gain.
Component % w/w Function
Surelease® 60 (25%> as solids) Release controlling polymer coat with plasticiser
Purified water q.s. Total 100
700 grams of core tablets were coated using a Vector LDCS pan to a 3% weight gain with the Opadry® Clear seal coating solution. The seal coated tablets were then polymer coated to 4% weight gain using the Surelease®coating dispersion. Table 6. Release Profile for a Matrix Tablet of Compound X in water
Time (hr) % Dissolved 1 8 2 30 4 58 8 96
Example 17 (Controlled release bilayer tablet)
Active Layer
Component mg/tablet Function
Compound X 0.005-0 .1 mg pfb Active
Methocel K4M 15.00 Hydrogel polymer
Lactose monohydrate 62.0 Hydrophilic filler
Polyvinyipyrrolidone 3.0 Binder
Magnesium stearate 1.0 Hydrophobic lubricant
Syloid 244 1.0 Hydrophilic glidant
Support platform
Component mg/tablet Function
Compritol 888 15.0 Plasticizer
Lactose monohydrate 29.0 Hydrophilic filler
Polyvinyipyrrolidone 4.0 Binder
Magnesium stearate 1.5 Hydrophobic lubricant
Methocel E5 29.4 Hydrogel polymer
Iron oxide 0.1 Colourant

Claims

1. A controlled release oral dosage form containing [R-(Z) ]-α-(methoxyimino)-α-( 1 - azabicyclo [2.2.2]oct-3-yl)acetonitrile monohydrochloride (compound X), its parent free base or any other pharmaceutically acceptable salt thereof.
2. A dosage form according to claim 1 which provides an in vitro release profile selected to provide an area under the in vivo plasma profile curve that is similar to that obtained following conventional oral administration of a fast release tablet 5 to 75 μg (calculated as free base) compound X twice a day.
3 A dosage form according to claim 1 or 2 which provides an in vitro release profile of 25-70% over 4 hours and 70- 100% over 8 hours.
4. A dosage form according to any of claims 1 to 3 selected from wax matrices, swellable and/or gellable matrices, tablets coated with release controlling polymers or waxes, and pellets, granules or beads comprising matrices or coated with release controlling polymers or waxes and then formulated as capsules, compressed tablets or suspensions.
5. A dosage form according to any preceding claim comprising a swellable and/or gellable matrix selected from alkyl celluloses, hydroxyalkylcelluloses, polyvinyl alcohol, polymethacrylates, polymethylmethacrylates, methacrylate/divinylbenzene copolymers, carboxymethylamide, polyoxyalkylene glycols, polyvinyl pyrrolidonc and carboxymethyl cellulose.
6. A dosage form according to claim 5 wherein the matrix is selected from alkyl celluloses, hydroxyalkylcelluloses, polyvinyl alcohol, polymethacrylates, cross-linked polyvinyipyrrolidone and sodium carboxymethyl cellulose.
7. A dosage form according to claim 5 or 6 comprising a hydrogel matrix tablet coated with a hydrophobic release controlling polymer coating selected from alkyl celluloses and methacrylic acid derivatives.
8. A dosage form according to claim 7 wherein the polymer matrix comprises 10- 50% and the hydrophobic release controlling polymer comprises 4-10% by weight of the tablet.
9. A dosage form according to claim 7 or 8 comprising a tablet of the following composition (mg/tablet):
Compound X 0.005-0.1 pfb
Hydroxpropyl Methcellulose E4M CR 75.0
Sodium Dihydrogen Citrate 0-3.00 Lactose, Fast Flo 70.38-73.38
Magnesium Stearate 1.50
Opadry® White 2.25 seal coated with a solution of Opadry® Clear (YS-l -7006) in purified water at 10% solids concentrations and polymer coated with a 60% w/w (25% as solids) dispersion containing Ethylcellulose (Surelease®) at 10%> weight gain, formed into core tablets, coated with the Opadry® Clear seal coating solution and polymer coated to 4% weight gain using 60% w/w (25% as solids) dispersion containing Ethylcellulose (Surelease®). -
10. A dosage form according to any of claims 1 to 4 which comprises drug-layered beads coated with a release controlling polymer either alone or in combination with drug- layered beads not coated with a release controlling polymer (immediate release beads) and optionally, inert excipients and/or retardants and/or one or more binders.
1 1. A dosage form according to claim 10 wherein the layered beads are seal coated with a film-forming polymer.
12. A dosage form according to claim 10 or 1 1 wherein the release controlling polymer coating is selected from from alkyl celluloses, hydroxyalkylcelluloses, sodium carboxymethyl cellulose and methacrylic acid derivatives.
13. A dosage form according to any of claims 10 to 12 wherein the polymer(s) make up 10 to 30% by weight of the total dosage form.
14. A dosage form according to claim 10 in capsule form comprising non-pareil sugar beads of 16-20, 20-25 or 25-30 mesh size, coated to a drug loading of 1 OOmicrogrammes (calculated as free base) per 200mg beads, with a medicated aqueous layer solution of the following composition (%w/w):
Compound X 0.003-0.06pfb
Opadry® Clear 3
Sodium dihydride citrate 0-1.5 seal coated with a solution of Opadry® Clear (YS-l -7006) in purified water at 10% solids concentrations to a weight gain of 3%, and a portion of the beads further polymer coated to a weight gain of 10-25% with a 60%w/w (25% as solids) dispersion containing ethylcellulose (Surelease®) and then seal coated to a weight gain of 2% with the above seal coat.
15. A method of treatment and/or prophylaxis of dementia, including Alzheimer's disease, in mammals by administering an effective amount of a controlled release oral dosage form according to claim 1 , to a sufferer in need thereof.
16. A method for enhancing amyloid precursor protein processing along a non- amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease by administering an effective amount of a controlled release oral dosage form according to claim 1, to a sufferer in need thereof.
17. The use of a controlled release oral dosage form according to claim 1, in the manufacture of a medicament for treatment and/or prophylaxis of dementia, including Alzheimer's disease, in mammals.
18. The use of a controlled release oral dosage form according to claim 1, in the manufacture of a medicament for enhancing amyloid precursor protein processing along a non-amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease.
19. A pharmaceutical composition for treatment and/or prophylaxis of dementia, including Alzheimer's disease, in mammals which comprises a controlled release oral dosage form according to claim 1.
20. A pharmaceutical composition for enhancing amyloid precursor protein processing along a non-amyloidogenic pathway in patients suffering from, or at risk of developing, Alzheimer's disease which comprises a controlled release oral dosage form according to claim 1.
21. A dosage form, method, use or composition according to any preceding claim in which release in the gastro-intestinal tract takes place predominantly over the first eight to twelve hours following ingestion.
22. A dosage form, method, use or composition according to any preceding claim containing [R-(Z)]-α-(methoxyimino)-α-(l-azabicyclo [2.2.2]oct-3-yl)acetonitrile monohydrochloride.
23. A dosage form, method, use or composition according to any of claims 1 to 22 containing 5μg compound X (calculated as free base).
24. A dosage form, method, use or composition according to any of claims 1 to 22 containing 12.5μg compound X (calculated as free base).
25. A dosage form, method, use or composition according to any of claims 1 to 22 containing 25 μg compound X (calculated as free base).
26. A dosage form, method, use or composition according to any of claims 1 to 22 containing 50μg compound X (calculated as free base).
27. A dosage form, method, use or composition according to any of claims 1 to 22 containing 75μg compound X (calculated as free base).
28. A dosage form, method, use or composition according to any of claims 1 to 22 containing lOOμg compound X (calculated as free base).
PCT/GB1997/002418 1996-09-12 1997-09-08 Controlled release dosage form of r-(z)-alpha-methoxyimino-alpha-(1-azabicyclo2.2oct-c-yl)acetonitrile monohydrochloride WO1998010762A2 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
NZ334268A NZ334268A (en) 1996-09-12 1997-09-08 Controlled release dosage form of [R-(Z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo [2.2.2]oct-3-yl) acetonitrile monohydrochloride
CZ99832A CZ83299A3 (en) 1996-09-12 1997-09-08 Dosage form of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyklo[2,2,2]okt-3-yl)acetonitrile monohydrochloride with controlled release
AU41288/97A AU724086B2 (en) 1996-09-12 1997-09-08 Controlled release dosage form of (R-(Z))-alpha- (methoxyimino)-alpha-(1-azabicyclo(2.2.2)oct-3-yl) acetonitrile monohydrochloride
PL97332074A PL332074A1 (en) 1996-09-12 1997-09-08 Dosable administration form of controllable release of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo-[2,2,2]oct-3-yl) acetonitrile monohydrochloride
HU9904401A HUP9904401A3 (en) 1996-09-12 1997-09-08 Controlled release dosage form of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo[2.2.2]oct-3-yl)acetonitrile monohydrochloride
JP10513352A JP2001500150A (en) 1996-09-12 1997-09-08 Controlled release dosage form of [R- (Z)]-α- (methoxyimino) -α- (1-azabicyclo [2.2.2] oct-3-yl) acetonitrile monohydrochloride
IL12878197A IL128781A0 (en) 1996-09-12 1997-09-08 Controlled release dosage form of (R-(Z))-alpha-(methoxyimino)-alpha-(1-azabicyclo(2.2.2)oct-3-yl) acetonitrile monohydrochloride
BR9711734A BR9711734A (en) 1996-09-12 1997-09-08 Controlled-release dosage form of ¬r- (z) alfa-alpha- (metÄxiimino) -alpha- (1-azabicyclo¬2.2.2¾oct-3-yl) acetonitrile monochlorochloride
EP97939064A EP0929301A2 (en) 1996-09-12 1997-09-08 Controlled release dosage form of r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo 2.2.2]oct-3-yl)acetonitrile monohydrochloride
CA002265661A CA2265661A1 (en) 1996-09-12 1997-09-08 Controlled release dosage form of r-(z)-alpha-methoxyimino-alpha-(1-azabicyclo2.2.2oct-c-yl)acetonitrile monohydrochloride
NO991194A NO991194L (en) 1996-09-12 1999-03-11 Controlled-release dosage form of [R- (Z) - <alfa> - (methoxyimino) - <alfa> - (azabicyclo [2,2,2] oct-3-yl) acetonitrile monohydrochloride

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9619074.9 1996-09-12
GBGB9619074.9A GB9619074D0 (en) 1996-09-12 1996-09-12 Composition

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09254621 A-371-Of-International 1999-03-11
US09/759,576 Continuation US20010003588A1 (en) 1996-09-12 2001-01-12 Controlled release dosage form of [R-(Z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo[2.2.2.]oct-3-yl)acetonitrile monohydrochloride

Publications (2)

Publication Number Publication Date
WO1998010762A2 true WO1998010762A2 (en) 1998-03-19
WO1998010762A3 WO1998010762A3 (en) 1998-06-04

Family

ID=10799833

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1997/002418 WO1998010762A2 (en) 1996-09-12 1997-09-08 Controlled release dosage form of r-(z)-alpha-methoxyimino-alpha-(1-azabicyclo2.2oct-c-yl)acetonitrile monohydrochloride

Country Status (22)

Country Link
EP (1) EP0929301A2 (en)
JP (1) JP2001500150A (en)
KR (1) KR20000036039A (en)
CN (2) CN1235544A (en)
AR (1) AR008176A1 (en)
AU (1) AU724086B2 (en)
BR (1) BR9711734A (en)
CA (1) CA2265661A1 (en)
CO (1) CO5031291A1 (en)
CZ (1) CZ83299A3 (en)
GB (1) GB9619074D0 (en)
HU (1) HUP9904401A3 (en)
ID (1) ID19589A (en)
IL (1) IL128781A0 (en)
MA (1) MA24359A1 (en)
NO (1) NO991194L (en)
NZ (1) NZ334268A (en)
PE (1) PE2499A1 (en)
PL (1) PL332074A1 (en)
TR (1) TR199900505T2 (en)
WO (1) WO1998010762A2 (en)
ZA (1) ZA978133B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999044589A1 (en) * 1998-03-04 1999-09-10 Gattefossé S.A. Oral pellet with immediate release comprising glycerides subjected to polyglycolysis, method for making same
WO1999045924A1 (en) * 1998-03-11 1999-09-16 Smithkline Beecham Plc Composition
WO2000064415A1 (en) * 1999-04-22 2000-11-02 Euroceltique S.A. Method for producing a water-insoluble amorphous or partially amorphous controlled release matrix
WO2001006982A2 (en) * 1999-07-26 2001-02-01 Laboratoires Des Produits Ethiques Ethypharm Low-dose tablets and preparation method
WO2002045571A2 (en) * 2000-12-06 2002-06-13 Wyeth Fast dissolving tablet
WO2004100932A1 (en) * 2003-05-14 2004-11-25 Eurand Pharmaceuticals Ltd. Controlled drug release composition resistant to in vivo mechanic stress
US8377977B2 (en) 2004-08-13 2013-02-19 Boehringer Ingelheim International Gmbh Extended release tablet formulation containing pramipexole or a pharmaceutically acceptable salt thereof
US8679533B2 (en) 2002-07-25 2014-03-25 Pharmacia Corporation Pramipexole once-daily dosage form
US8715728B2 (en) 2004-08-13 2014-05-06 Boehringer Ingelheim International Gmbh Extended release pellet formulation containing pramipexole or a pharmaceutically acceptable salt thereof
US9132096B1 (en) 2014-09-12 2015-09-15 Alkermes Pharma Ireland Limited Abuse resistant pharmaceutical compositions

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1617838A4 (en) * 2003-04-25 2011-07-06 Mitsubishi Tanabe Pharma Corp Composition for oral administration containing alkylene dioxybenzene derivative
US20050142191A1 (en) * 2003-06-23 2005-06-30 Neurochem (International) Limited Pharmaceutical formulations of amyloid inhibiting compounds
JP2005272347A (en) * 2004-03-24 2005-10-06 Ohara Yakuhin Kogyo Kk Method for producing solid preparation
EP1970056A1 (en) * 2007-03-15 2008-09-17 Polichem S.A. Time-specific delayed/pulsatile release dosage forms

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0392803A1 (en) * 1989-04-13 1990-10-17 Beecham Group p.l.c. Novel compounds
WO1995031457A1 (en) * 1994-05-14 1995-11-23 Smithkline Beecham Plc Derivatives of quinuclidine n-oxide as muscarinic receptor ligands
WO1996012486A1 (en) * 1994-10-25 1996-05-02 Smithkline Beecham P.L.C. Use of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo [2.2.2] oct-3-yl) acetonitrile to reduce amyloid beta a4 formation in alzheimer's disease
WO1997004750A2 (en) * 1995-07-29 1997-02-13 Smithkline Beecham Plc Process for preparing solid dosage forms of very low-dose drugs

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0392803A1 (en) * 1989-04-13 1990-10-17 Beecham Group p.l.c. Novel compounds
WO1995031457A1 (en) * 1994-05-14 1995-11-23 Smithkline Beecham Plc Derivatives of quinuclidine n-oxide as muscarinic receptor ligands
WO1996012486A1 (en) * 1994-10-25 1996-05-02 Smithkline Beecham P.L.C. Use of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo [2.2.2] oct-3-yl) acetonitrile to reduce amyloid beta a4 formation in alzheimer's disease
WO1997004750A2 (en) * 1995-07-29 1997-02-13 Smithkline Beecham Plc Process for preparing solid dosage forms of very low-dose drugs

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999044589A1 (en) * 1998-03-04 1999-09-10 Gattefossé S.A. Oral pellet with immediate release comprising glycerides subjected to polyglycolysis, method for making same
WO1999045924A1 (en) * 1998-03-11 1999-09-16 Smithkline Beecham Plc Composition
US6451343B1 (en) 1998-03-11 2002-09-17 Smithkline Beecham Corporation Composition for treating dementia and Alzheimer's disease
WO2000064415A1 (en) * 1999-04-22 2000-11-02 Euroceltique S.A. Method for producing a water-insoluble amorphous or partially amorphous controlled release matrix
EP1839651A1 (en) * 1999-04-22 2007-10-03 Euro-Celtique S.A. Method for producing a water-insoluble amorphous or partially amorphous controlled release matrix
AU763609B2 (en) * 1999-04-22 2003-07-31 Euro-Celtique S.A. Method for producing a water-insoluble amorphous or partially amorphous controlled release matrix
JP2007211019A (en) * 1999-04-22 2007-08-23 Euro-Celtique Sa Method for producing pharmaceutical form or preliminary stage thereof, pharmaceutical form and method for using pharmaceutical form
WO2001006982A2 (en) * 1999-07-26 2001-02-01 Laboratoires Des Produits Ethiques Ethypharm Low-dose tablets and preparation method
FR2796840A1 (en) * 1999-07-26 2001-02-02 Ethypharm Lab Prod Ethiques LOW DOSAGE TABLETS AND METHOD OF PREPARATION
WO2001006982A3 (en) * 1999-07-26 2001-04-12 Ethypharm Lab Prod Ethiques Low-dose tablets and preparation method
US8337892B1 (en) 1999-07-26 2012-12-25 Ethypharm Low-dose tablets and preparation method
AU2002230589B2 (en) * 2000-12-06 2005-11-10 Pf Consumer Healthcare 1 Llc Fast dissolving tablet
US6733781B2 (en) 2000-12-06 2004-05-11 Wyeth Fast dissolving tablet
WO2002045571A3 (en) * 2000-12-06 2002-08-29 Wyeth Corp Fast dissolving tablet
WO2002045571A2 (en) * 2000-12-06 2002-06-13 Wyeth Fast dissolving tablet
US8679533B2 (en) 2002-07-25 2014-03-25 Pharmacia Corporation Pramipexole once-daily dosage form
WO2004100932A1 (en) * 2003-05-14 2004-11-25 Eurand Pharmaceuticals Ltd. Controlled drug release composition resistant to in vivo mechanic stress
US8715728B2 (en) 2004-08-13 2014-05-06 Boehringer Ingelheim International Gmbh Extended release pellet formulation containing pramipexole or a pharmaceutically acceptable salt thereof
US8377977B2 (en) 2004-08-13 2013-02-19 Boehringer Ingelheim International Gmbh Extended release tablet formulation containing pramipexole or a pharmaceutically acceptable salt thereof
US9132096B1 (en) 2014-09-12 2015-09-15 Alkermes Pharma Ireland Limited Abuse resistant pharmaceutical compositions
US9452163B2 (en) 2014-09-12 2016-09-27 Recro Gainesville Llc Abuse resistant pharmaceutical compositions
US9486451B2 (en) 2014-09-12 2016-11-08 Recro Gainesville Llc Abuse resistant pharmaceutical compositions
US9713611B2 (en) 2014-09-12 2017-07-25 Recro Gainesville, LLC Abuse resistant pharmaceutical compositions
US10092559B2 (en) 2014-09-12 2018-10-09 Recro Gainesville Llc Abuse resistant pharmaceutical compositions
US10960000B2 (en) 2014-09-12 2021-03-30 Recro Gainesville Llc Abuse resistant pharmaceutical compositions

Also Published As

Publication number Publication date
AR008176A1 (en) 1999-12-09
WO1998010762A3 (en) 1998-06-04
GB9619074D0 (en) 1996-10-23
CO5031291A1 (en) 2001-04-27
EP0929301A2 (en) 1999-07-21
HUP9904401A3 (en) 2001-03-28
CN1446535A (en) 2003-10-08
AU4128897A (en) 1998-04-02
KR20000036039A (en) 2000-06-26
NZ334268A (en) 2000-10-27
BR9711734A (en) 1999-08-24
ID19589A (en) 1998-07-23
AU724086B2 (en) 2000-09-14
JP2001500150A (en) 2001-01-09
CN1235544A (en) 1999-11-17
HUP9904401A2 (en) 2000-06-28
IL128781A0 (en) 2000-01-31
PL332074A1 (en) 1999-08-30
CZ83299A3 (en) 1999-08-11
MA24359A1 (en) 1998-07-01
NO991194D0 (en) 1999-03-11
ZA978133B (en) 1999-04-12
TR199900505T2 (en) 1999-06-21
NO991194L (en) 1999-03-11
CA2265661A1 (en) 1998-03-19
PE2499A1 (en) 1999-03-24

Similar Documents

Publication Publication Date Title
US6468560B2 (en) Controlled release dosage form of [R-(Z)]-α-(methoxyimino)-α-(1-azabicyclo[2.2.2]oct-3yl) acetonitrile monohydrochloride
US5807579A (en) Pseudoephedrine combination pharmaceutical compositions
US7790200B2 (en) Formulation and process for drug loaded cores
EP0605174B1 (en) Delayed, Sustained-release pharmaceutical preparation
US20110008426A1 (en) Modified release pharmaceutical compositions comprising mycophenolate and processes thereof
US20030133982A1 (en) Zero-order sustained release dosage forms and method of making same
US20130230596A1 (en) Abuse-Resistant Oral Dosage Forms And Method Of Use Thereof
US20040228915A1 (en) Oral extended release compressed tablets of multiparticulates
EP1978935B1 (en) Coated formulations
AU2008288106B2 (en) Extended release compositions comprising mycophenolate sodium and processes thereof
WO2010140111A1 (en) Pharmaceutical compositions containing a combination of an antihistamine and a decongestant
KR20090091084A (en) Controlled-release pharmaceutical formulation
US20020156133A1 (en) Oral administration forms for administering a fixed tramadol and diclofenac combination
AU724086B2 (en) Controlled release dosage form of (R-(Z))-alpha- (methoxyimino)-alpha-(1-azabicyclo(2.2.2)oct-3-yl) acetonitrile monohydrochloride
US20030224045A1 (en) Combination immediate release sustained release levodopa/carbidopa dosage forms
US20030129236A1 (en) Multiple pulse extended release formulations of clindamycin
US20030099710A1 (en) Granule modulating hydrogel system
US20090220593A1 (en) Extended release dosage forms of quetiapine
JP2003500348A (en) Multiparticulate controlled release selective serotonin reuptake inhibitor formulation
US7060293B1 (en) Powder-layered oral dosage forms
US20040146556A1 (en) Oral extended release tablets and methods of making and using the same
KR20030076634A (en) Medicinal composition
US8173164B2 (en) Oral administration forms for administering a fixed tramadol and diclofenac combination
MXPA99002404A (en) Controlled release dosage form of [r-(z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo[2.2. 2]oct-3-yl)acetonitrile monohydrochloride
JP2003507416A (en) Pharmaceutical preparations

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 97199411.0

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 334268

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1997939064

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1999/00505

Country of ref document: TR

WWE Wipo information: entry into national phase

Ref document number: PV1999-832

Country of ref document: CZ

ENP Entry into the national phase

Ref document number: 2265661

Country of ref document: CA

Ref document number: 2265661

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/1999/002404

Country of ref document: MX

Ref document number: 1019997002030

Country of ref document: KR

Ref document number: 09254621

Country of ref document: US

ENP Entry into the national phase

Ref document number: 1998 513352

Country of ref document: JP

Kind code of ref document: A

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1997939064

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1999-832

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1019997002030

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1997939064

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1019997002030

Country of ref document: KR

WWR Wipo information: refused in national office

Ref document number: PV1999-832

Country of ref document: CZ