WO2007029081A1 - Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use for the manufacture of a medicament - Google Patents

Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use for the manufacture of a medicament Download PDF

Info

Publication number
WO2007029081A1
WO2007029081A1 PCT/IB2006/002406 IB2006002406W WO2007029081A1 WO 2007029081 A1 WO2007029081 A1 WO 2007029081A1 IB 2006002406 W IB2006002406 W IB 2006002406W WO 2007029081 A1 WO2007029081 A1 WO 2007029081A1
Authority
WO
WIPO (PCT)
Prior art keywords
galantamine
controlled release
water insoluble
rate
core
Prior art date
Application number
PCT/IB2006/002406
Other languages
French (fr)
Inventor
Rajan Kumar Verma
Narayanan Badri Viswanathan
Rajeev Singh Raghuvanshi
Ashok Rampal
Original Assignee
Ranbaxy Laboratories Limited
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
Application filed by Ranbaxy Laboratories Limited filed Critical Ranbaxy Laboratories Limited
Priority to EP06795399A priority Critical patent/EP1933816A1/en
Priority to US12/065,773 priority patent/US20090087488A1/en
Publication of WO2007029081A1 publication Critical patent/WO2007029081A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • A61K9/5047Cellulose ethers containing no ester groups, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5073Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings
    • A61K9/5078Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings with drug-free core

Definitions

  • the present invention relates to controlled release oral dosage forms of galantamine or acceptable salts thereof and processes for the preparation thereof.
  • Galantamine is a known reversible, competitive acetylcholinesterase inhibitor. Chemically, it is 4aS, 6R, 8a-4a, 5, 9, 10, 11, ⁇ -hexahydro-S-methoxy-l l-methyl- ⁇ H- benzofuro [3a, 3,2-ef [2] benzazepin-6-ol) and it is isolated from both the bulbs of the Caucasian snowdrops Galanthus woronowi and also from the common snowdrop
  • Galanthus nivalis Galantamine have been used as a curare reversal agent in anesthetic practice in Eastern bloc countries and also experimentally in the West. Galantamine and its salts have been tested in the treatment of mania, chronic fatigue syndrome, schizophrenia, alcoholism, nicotine dependence and Alzheimer's disease. Galantamine hydrobromide is currently available in tablets and capsules for use in the treatment of Alzheimer's disease as RAZADYNETM.
  • Galantamine has an affinity for nicotinic receptors but not for muscarinic receptors, and it is capable of passing the blood-brain barrier.
  • Galantamine salts such as hydrobromide are given as immediate release tablets administered two or three times a day.
  • the therapy with an immediate release dosage form leads to undesired peaks in the plasma profiles of galantamine and a sharp decrease in concentration after about 6 to 8 hours. These fluctuations ranging from high to low plasma concentrations are undesirable and may lead to side effects, such as nausea, vomiting or headaches. These side effects generally precipitate when galantamine is administered in high doses.
  • a typical therapy with galantamine starts with a lower dose for 3 - 4 weeks and the dose is gradually increased till maximum tolerable dosage is achieved. If treatment with galantamine is interrupted for several days or longer, the patient will need to start over again at the lowest dose, increasing the dose at 4-week intervals until the former dose is achieved.
  • galantamine is administered two or three times a day. It would be desirable to reduce the frequency of dosing to once a day which would result in reduced plasma fluctuations in the in the plasma concentration of galantamine. This objective may be achieved by administering galantamine in controlled release form such that the constant plasma levels of galantamine are maintained over an extended period of time.
  • WO 0038686 discloses a controlled release formulation containing galantamine as the active ingredient, wherein it includes particles comprising galantamine or a pharmaceutically acceptable acid addition salt thereof, a water soluble pharmaceutically acceptable excipient and optionally other pharmaceutically acceptable excipients.
  • the particles are coated by a release rate controlling membrane coating.
  • the water soluble excipient has been described as a water soluble polymer.
  • the controlled release dosage form includes a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and a rate-controlling coating.
  • Embodiments of the controlled release dosage form may include one or more of the following features.
  • the one or more water insoluble excipients may be one or more of silicon dioxide, ethylcellulose, crosp.ovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
  • the core may include an inert sphere onto which one or more water insoluble excipients are coated with galantamine dispersed in it.
  • the core may include galantamine dispersed within it.
  • the rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
  • the water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
  • the water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose.
  • the one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
  • the controlled release formulation may include a seal coat between the core and the release rate-controlling coating.
  • the formulation may further include an immediate release portion which includes from about 10% to about 50% of the total dose of galantamine present.
  • the immediate release portion may be a coating over the rate- controlling coating or as galantamine core devoid of rate-controlling coating.
  • a process for the preparation of controlled release formulation of galantamine includes admixing galantamine or a pharmaceutically acceptable salt thereof with one or more water insoluble excipients to form a drug core; optionally applying a seal coat to the drug core; and applying a release-controlling coating over the core.
  • the one or more water insoluble excipients may be silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
  • the rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
  • the water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
  • the water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose.
  • the one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
  • a method of treating alzheimer's dementia in a patient in need thereof includes administering a controlled release dosage form of galantamine.
  • the controlled release dosage form of galantamine includes a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and a rate-controlling coating.
  • Embodiments of the method may include one or more of the following features.
  • the one or more water insoluble excipients may be silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
  • the rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
  • the water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
  • the water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose.
  • the one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol - A - Detailed Description of the Invention
  • a controlled release dosage form of galantamine may be prepared by using a core that includes galantamine and one or more water insoluble excipients, wherein the core is coated with a rate controlling layer.
  • controlled release dosage forms provide steady plasma levels of galantamine over a period of 12 to 24 hours.
  • the dosage forms may preferably be given once a day and would improve patient compliance.
  • Embodiments of the dosage form may include one or more of the following features.
  • the water insoluble excipients may include one or more of silicon dioxide, water insoluble polymers, such as ethylcellulose, crospovidone, sodium starch glycolate, cross-carmelose sodium, microcrystalline cellulose, water insoluble methacrylate polymers (ammonio methacrylate copolymer; e.g., Eudragit RLPO, Eudragit RSPO) and mixtures thereof.
  • the water insoluble polymers may be present throughout the core or as a coating on a core that includes galantamine dispersed in it.
  • the core may be sugar spheres and be composed of sucrose, starch, mannitol, lactose, microcrystalline cellulose, sodium carboxymethyl cellulose, silica, and glass.
  • a layer of galantamine may be dispersed along with one or more water insoluble polymer(s) and one or more pharmaceutically acceptable excipients to form a coating.
  • the core may be prepared by conventional techniques known in the art including granulation, extrusion and spheronization and the galantamine may be dispersed within the core or coated onto the core.
  • the rate-controlling coating may include one or more water insoluble polymers.
  • the water insoluble polymer maybe ethylcellulose, cellulose acetate, ammonio methacrylate copolymers, and methacrylic acid copolymer.
  • the rate controlling coating in addition to one or more water insoluble polymers may include one or more plasticizers, such as triethyl citrate, dibutyl phthalate, diethyl phthalate, triacetin, and PEG-400.
  • the rate-controlling coating may include a water soluble polymer such as polyvinylpyrrolidone or hydroxypropyl methylcellulose.
  • An optional seal coat may also be present between the core and the rate-controlling coating.
  • the seal coat may include water soluble polymers such as hydroxypropyl methylcellulose.
  • coated cores may be filled into capsules or compressed into tablets.
  • the coated core may be formulated to include a unit dose of galantamine and administered as such.
  • an immediate release portion sufficient to provide an initial therapeutic plasma levels may also be included in the dosage form.
  • the immediate release portion may be cores of the invention devoid of rate-controlling coating or it may be a galantamine containing coating over the rate-controlling coating and is readily soluble in aqueous media.
  • the immediate release portion may be at a concentration of between about 10% to about 50% of total dose of galantamine.
  • galantamine as defined herein includes galantamine base and pharmaceutically acceptable salts thereof.
  • the pharmaceutically acceptable salt of galantamine may be the hydrobromide salt.
  • the controlled release dosage form may provide therapeutic plasma levels of galantamine from about 12 to about 24 hours. For example, the dosage form maybe administered once a day.
  • Also provided in the present invention are processes for the preparation of controlled release dosage forms of galantamine.
  • the process includes admixing galantamine or a pharmaceutically acceptable salt form thereof with one or more water insoluble excipients to form a drug core and applying the release rate controlling membrane coating.
  • Embodiments of the process include one or more of the following features.
  • the process may include dispersing galantamine, the one or more water insoluble ⁇ olymer(s), and one or more excipients in a pharmaceutically acceptable solvent system and coated on to the inert core by conventional coating processes.
  • the coated core may then be further coated with a rate-controlling coating.
  • An optional seal coat may also be coated prior to applying the rate-controlling coating.
  • galantamine may be mixed with one or more water insoluble excipients and granulated either with water alone or with a water insoluble binder solution. The granules are dried and may be compressed into cores and - 6 - coated with the rate-controlling coating.
  • the process may include granulating a mixture of galantamine and one or more water insoluble excipients, extruding and spheronizing the wet mass into round spheroids, drying the spheroids and coating with the rate-controlling coating. An optional seal coat may also be coated prior to applying the rate-controlling coating.
  • galantamine may be sprayed onto the non-peril core and coated with a rate-controlling coating.
  • Galantamine hydrobromide, Eudragit RLPO, crospovidone and colloidal silicon dioxide were dispersed in isopropyl alcohol/water mixture (75/25).
  • Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
  • the above drug layered cores were coated with the coating composition which includes ethyl cellulose, hydroxypropylmethylcellulose and triacetin dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
  • the cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 8.8%; 10%; 11%, and 12% w/w.
  • Example 1 The coated cores of Example 1 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 1.
  • Table 1 Dissolution profile of coated cores of Example 1 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
  • Galantamine hydrobromide and Eudragit RLPO were dispersed in an isopropyl alcohol/water mixture (50/50 weight by weight) .
  • Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
  • the above drug layered cores were coated with the coating composition containing ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in isopropyl alcohol/water mixture (90/10 weight by weight).
  • the cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 6.4%, 9%, and 11.3% w/w.
  • Example 2 The coated cores of Example 2 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 2. Table 2: Dissolution profile of coated cores of Example 2 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer ofpH 6.8 at 50 rpm.
  • Galantamine hydrobromide and Eudragit RLPO were dispersed in isopropyl alcohol/water mixture (50/50 weight by weight).
  • Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
  • the above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
  • the cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 9%, 10%, 11%, and 12% w/w.
  • Example 3 The coated cores of Example 3 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 3.
  • Table 3 Dissolution profile of coated cores of example 3 with varying amounts of rate controlling coating in USP II dissolution apparatus in 90OmL phosphate buffer ofpH 6.8 at 50rpm.
  • Galantamine hydrobromide and Eudragit RLPO were dispersed in an isopropyl alcohol/water mixture (50/50 weight by weight) .
  • Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
  • the above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
  • the cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 5.1%, 6.1%, and 6.4% w/w.
  • Example 4 The coated cores of Example 4 were subjected to dissolution studies with USP II dissolution apparatus in 900 niL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 4. Table 4: Dissolution profile of coated cores of example 4 with varying amounts of rate controlling coating in USP ⁇ I dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
  • Galantamine hydrobromide, Eudragit RLPO, crospovidone and colloidal silicon dioxide were dispersed in an isopropyl alcohol/water mixture (75/25 weight by weight).
  • Non-peril sugar beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
  • the above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and triacetin dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
  • the beads were coated to a weight gain of 5.3% w/w.
  • Example 5 The coated cores of Example 5 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 5. -
  • Table 5 Dissolution profile of coated cores of Example 5 in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
  • the resultant spheroids were dried in fluid bed drier at 6O 0 C.
  • Spheroids were placed in fluid-bed coating apparatus and coated with above coating solution.
  • the spheroids with varying percentages of rate controlling coating were prepared i.e. spheroids had rate controlling coating in amounts of 6.1%, 7.5%, 8.0%, and 8.6% w/w.
  • Example 6 The coated spheroids of Example 6 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 6.
  • Table 6 Dissolution profile of coated spheroids of Example 6 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to controlled release oral dosage forms of galantamine or acceptable salts thereof and processes for the preparation thereof.

Description

GALANTAMINE-CONTAINING CONTROLLED RELEASE ORAL DOSAGE FORMS, PROCESSES FOR THE PREPARATION THEREOF AND USE FOR THE MANUFACTURE OF A MEDICAMENT
Technical Field of the Invention
The present invention relates to controlled release oral dosage forms of galantamine or acceptable salts thereof and processes for the preparation thereof.
Background of the Invention
Galantamine is a known reversible, competitive acetylcholinesterase inhibitor. Chemically, it is 4aS, 6R, 8a-4a, 5, 9, 10, 11, ^-hexahydro-S-methoxy-l l-methyl-βH- benzofuro [3a, 3,2-ef [2] benzazepin-6-ol) and it is isolated from both the bulbs of the Caucasian snowdrops Galanthus woronowi and also from the common snowdrop
Galanthus nivalis. Galantamine have been used as a curare reversal agent in anesthetic practice in Eastern bloc countries and also experimentally in the West. Galantamine and its salts have been tested in the treatment of mania, chronic fatigue syndrome, schizophrenia, alcoholism, nicotine dependence and Alzheimer's disease. Galantamine hydrobromide is currently available in tablets and capsules for use in the treatment of Alzheimer's disease as RAZADYNE™.
Galantamine has an affinity for nicotinic receptors but not for muscarinic receptors, and it is capable of passing the blood-brain barrier. Galantamine salts such as hydrobromide are given as immediate release tablets administered two or three times a day. The therapy with an immediate release dosage form leads to undesired peaks in the plasma profiles of galantamine and a sharp decrease in concentration after about 6 to 8 hours. These fluctuations ranging from high to low plasma concentrations are undesirable and may lead to side effects, such as nausea, vomiting or headaches. These side effects generally precipitate when galantamine is administered in high doses. A typical therapy with galantamine starts with a lower dose for 3 - 4 weeks and the dose is gradually increased till maximum tolerable dosage is achieved. If treatment with galantamine is interrupted for several days or longer, the patient will need to start over again at the lowest dose, increasing the dose at 4-week intervals until the former dose is achieved.
Currently galantamine is administered two or three times a day. It would be desirable to reduce the frequency of dosing to once a day which would result in reduced plasma fluctuations in the in the plasma concentration of galantamine. This objective may be achieved by administering galantamine in controlled release form such that the constant plasma levels of galantamine are maintained over an extended period of time.
WO 0038686 discloses a controlled release formulation containing galantamine as the active ingredient, wherein it includes particles comprising galantamine or a pharmaceutically acceptable acid addition salt thereof, a water soluble pharmaceutically acceptable excipient and optionally other pharmaceutically acceptable excipients. The particles are coated by a release rate controlling membrane coating. The water soluble excipient has been described as a water soluble polymer.
Summary of the Invention In one general aspect there is provided a controlled release dosage form of galantamine. The controlled release dosage form includes a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and a rate-controlling coating.
Embodiments of the controlled release dosage form may include one or more of the following features. For example, the one or more water insoluble excipients may be one or more of silicon dioxide, ethylcellulose, crosp.ovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
The core may include an inert sphere onto which one or more water insoluble excipients are coated with galantamine dispersed in it. Alternatively, the core may include galantamine dispersed within it.
The rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers. The water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer. The water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose. The one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
The controlled release formulation may include a seal coat between the core and the release rate-controlling coating. The formulation may further include an immediate release portion which includes from about 10% to about 50% of the total dose of galantamine present. The immediate release portion may be a coating over the rate- controlling coating or as galantamine core devoid of rate-controlling coating. In another general aspect there is provided a process for the preparation of controlled release formulation of galantamine. The process includes admixing galantamine or a pharmaceutically acceptable salt thereof with one or more water insoluble excipients to form a drug core; optionally applying a seal coat to the drug core; and applying a release-controlling coating over the core.
The one or more water insoluble excipients may be silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers. The rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers. The water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer. The water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose. The one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
In another general aspect there is provided a method of treating alzheimer's dementia in a patient in need thereof. The method includes administering a controlled release dosage form of galantamine. The controlled release dosage form of galantamine includes a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and a rate-controlling coating.
Embodiments of the method may include one or more of the following features. For example, the one or more water insoluble excipients may be silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers. The rate-controlling coating may include one or more water insoluble polymers, a water soluble polymer and one or more plasticizers. The water insoluble polymer may be ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer. The water soluble polymer may be polyvinylpyrrolidone and hydroxypropylmethylcellulose. The one or more plasticizers may be triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol - A - Detailed Description of the Invention
In the present invention the inventors have surprisingly found that a controlled release dosage form of galantamine may be prepared by using a core that includes galantamine and one or more water insoluble excipients, wherein the core is coated with a rate controlling layer.
These controlled release dosage forms provide steady plasma levels of galantamine over a period of 12 to 24 hours. The dosage forms may preferably be given once a day and would improve patient compliance.
Embodiments of the dosage form may include one or more of the following features. For example, the water insoluble excipients may include one or more of silicon dioxide, water insoluble polymers, such as ethylcellulose, crospovidone, sodium starch glycolate, cross-carmelose sodium, microcrystalline cellulose, water insoluble methacrylate polymers (ammonio methacrylate copolymer; e.g., Eudragit RLPO, Eudragit RSPO) and mixtures thereof. The water insoluble polymers may be present throughout the core or as a coating on a core that includes galantamine dispersed in it.
The core may be sugar spheres and be composed of sucrose, starch, mannitol, lactose, microcrystalline cellulose, sodium carboxymethyl cellulose, silica, and glass. Upon these cores a layer of galantamine may be dispersed along with one or more water insoluble polymer(s) and one or more pharmaceutically acceptable excipients to form a coating. Alternatively, the core may be prepared by conventional techniques known in the art including granulation, extrusion and spheronization and the galantamine may be dispersed within the core or coated onto the core.
The rate-controlling coating may include one or more water insoluble polymers. The water insoluble polymer maybe ethylcellulose, cellulose acetate, ammonio methacrylate copolymers, and methacrylic acid copolymer. The rate controlling coating in addition to one or more water insoluble polymers may include one or more plasticizers, such as triethyl citrate, dibutyl phthalate, diethyl phthalate, triacetin, and PEG-400.
Optionally, in order to adjust the rate of release of galantamine the rate-controlling coating may include a water soluble polymer such as polyvinylpyrrolidone or hydroxypropyl methylcellulose. An optional seal coat may also be present between the core and the rate-controlling coating. The seal coat may include water soluble polymers such as hydroxypropyl methylcellulose.
These coated cores may be filled into capsules or compressed into tablets. Alternatively, the coated core may be formulated to include a unit dose of galantamine and administered as such.
In another embodiment, an immediate release portion sufficient to provide an initial therapeutic plasma levels may also be included in the dosage form. The immediate release portion may be cores of the invention devoid of rate-controlling coating or it may be a galantamine containing coating over the rate-controlling coating and is readily soluble in aqueous media. The immediate release portion may be at a concentration of between about 10% to about 50% of total dose of galantamine.
The term "galantamine" as defined herein includes galantamine base and pharmaceutically acceptable salts thereof. The pharmaceutically acceptable salt of galantamine may be the hydrobromide salt. The controlled release dosage form may provide therapeutic plasma levels of galantamine from about 12 to about 24 hours. For example, the dosage form maybe administered once a day.
Also provided in the present invention are processes for the preparation of controlled release dosage forms of galantamine. The process includes admixing galantamine or a pharmaceutically acceptable salt form thereof with one or more water insoluble excipients to form a drug core and applying the release rate controlling membrane coating.
Embodiments of the process include one or more of the following features. The process may include dispersing galantamine, the one or more water insoluble ρolymer(s), and one or more excipients in a pharmaceutically acceptable solvent system and coated on to the inert core by conventional coating processes. The coated core may then be further coated with a rate-controlling coating. An optional seal coat may also be coated prior to applying the rate-controlling coating.
In another embodiment of the process, galantamine may be mixed with one or more water insoluble excipients and granulated either with water alone or with a water insoluble binder solution. The granules are dried and may be compressed into cores and - 6 - coated with the rate-controlling coating. In an alternative step, the process may include granulating a mixture of galantamine and one or more water insoluble excipients, extruding and spheronizing the wet mass into round spheroids, drying the spheroids and coating with the rate-controlling coating. An optional seal coat may also be coated prior to applying the rate-controlling coating.
In yet another embodiment of the process, galantamine may be sprayed onto the non-peril core and coated with a rate-controlling coating.
It is apparent from the text that various modifications and combinations of the formulations can be made without departing from the spirit and scope of the invention. For example, a water soluble or insoluble core may be used. Similarly, concentration of different excipients may vary as is apparent from the examples.
EXAMPLE 1
Figure imgf000007_0001
Procedure:
1. Galantamine hydrobromide, Eudragit RLPO, crospovidone and colloidal silicon dioxide were dispersed in isopropyl alcohol/water mixture (75/25). 2. Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
3. The above drug layered cores were coated with the coating composition which includes ethyl cellulose, hydroxypropylmethylcellulose and triacetin dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
The cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 8.8%; 10%; 11%, and 12% w/w.
The coated cores of Example 1 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 1.
Table 1: Dissolution profile of coated cores of Example 1 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
Figure imgf000008_0001
EXAMPLE 2
Figure imgf000009_0001
Procedure:
1. Galantamine hydrobromide and Eudragit RLPO were dispersed in an isopropyl alcohol/water mixture (50/50 weight by weight) .
2. Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
3. The above drug layered cores were coated with the coating composition containing ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in isopropyl alcohol/water mixture (90/10 weight by weight).
The cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 6.4%, 9%, and 11.3% w/w.
The coated cores of Example 2 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 2. Table 2: Dissolution profile of coated cores of Example 2 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer ofpH 6.8 at 50 rpm.
Figure imgf000010_0001
EXAMPLE 3
Figure imgf000010_0002
Procedure:
1. Galantamine hydrobromide and Eudragit RLPO were dispersed in isopropyl alcohol/water mixture (50/50 weight by weight).
2. Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
3. The above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
The cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 9%, 10%, 11%, and 12% w/w.
The coated cores of Example 3 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 3.
Table 3: Dissolution profile of coated cores of example 3 with varying amounts of rate controlling coating in USP II dissolution apparatus in 90OmL phosphate buffer ofpH 6.8 at 50rpm.
Figure imgf000011_0001
EXAMPLE 4
Figure imgf000012_0001
Procedure:
1. Galantamine hydrobromide and Eudragit RLPO were dispersed in an isopropyl alcohol/water mixture (50/50 weight by weight) .
2. Microcrystalline cellulose beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
3. The above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and diethyl phthalate dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight).
The cores with varying percentages of rate controlling coating were prepared i.e. beads had rate controlling coating in amounts of 5.1%, 6.1%, and 6.4% w/w.
The coated cores of Example 4 were subjected to dissolution studies with USP II dissolution apparatus in 900 niL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 4. Table 4: Dissolution profile of coated cores of example 4 with varying amounts of rate controlling coating in USP ΪI dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
Figure imgf000013_0001
EXAMPLE 5
Figure imgf000013_0002
Procedure:
1. Galantamine hydrobromide, Eudragit RLPO, crospovidone and colloidal silicon dioxide were dispersed in an isopropyl alcohol/water mixture (75/25 weight by weight).
2. Non-peril sugar beads were loaded in a fluid bed coating apparatus and sprayed with dispersion of step 1.
3. The above drug layered cores were coated with the coating composition that includes ethyl cellulose, hydroxypropylmethylcellulose and triacetin dissolved in an isopropyl alcohol/water mixture (90/10 weight by weight). The beads were coated to a weight gain of 5.3% w/w.
The coated cores of Example 5 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 5. -
Table 5: Dissolution profile of coated cores of Example 5 in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
Figure imgf000014_0001
EXAMPLE 6 Preparation of Spheroids
Figure imgf000015_0001
1. All the ingredients were blended in a granulator.
2. Water was added to the above blend and mixed till a wet mass was obtained.
3. The wet mass was extruded to give cylindrical extrudes.
4. The damp extrudes were placed in spheronizer and rotated till spheroids were formed.
5. The resultant spheroids were dried in fluid bed drier at 6O0C.
6. The dried spheroids were screened to give fraction between #16-25 BSS screen.
The above spheroids were coated with following coating composition:
Figure imgf000015_0002
Procedure
1. AU the ingredients were dissolved in an isopropyl alcohol-water mixture (90/10 weight by weight).
2. Spheroids were placed in fluid-bed coating apparatus and coated with above coating solution. The spheroids with varying percentages of rate controlling coating were prepared i.e. spheroids had rate controlling coating in amounts of 6.1%, 7.5%, 8.0%, and 8.6% w/w.
The coated spheroids of Example 6 were subjected to dissolution studies with USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm. The results are given in Table 6.
Table 6: Dissolution profile of coated spheroids of Example 6 with varying amounts of rate controlling coating in USP II dissolution apparatus in 900 mL phosphate buffer of pH 6.8 at 50 rpm.
Figure imgf000016_0001
While the present invention has been described in terms of its specific embodiments, certain modifications and equivalents will be apparent to those skilled in the art and are included within the scope of the present invention. The examples are provided to illustrate particular aspects of the disclosure and do not limit the scope of the present invention as defined by the claims.

Claims

We claim: 1. A controlled release dosage form of galantamine comprising: a. a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and b. a rate-controlling coating.
2. The controlled release formulation according of claim 1, wherein the one or more water insoluble excipients comprise silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
3. The controlled release formulation according to claim 1, wherein the core comprises an inert sphere onto which one or more water insoluble excipients is coated with galantamine dispersed in it.
4. The controlled release formulation according to claim 1, wherein the core comprises galantamine dispersed within it.
5. The controlled release formulation according to claim 1, wherein the rate- controlling coating comprises one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
6. The controlled release formulation according to claim 5, wherein the water insoluble polymer comprises ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
7. The controlled release formulation according to claim 5, wherein the water soluble polymer comprises polyvinylpyrrolidone and hydroxypropylmethylcellulose.
8. The controlled release formulation according to claim 5, wherein the one or more plasticizers comprise triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
9. The controlled release formulation according to claim 1, wherein a seal coat lies between the core and the release rate-controlling coating.
10. The controlled release formulation according to claim 1, further comprising an immediate release portion comprising from about 10% to about 50% of the total dose of galantamine present.
11. The controlled release formulation according to claim 10, wherein the immediate release portion comprises a coating over the rate-controlling coating or as galantamine core devoid of rate-controlling coating.
12. A process for the preparation of controlled release formulation of galantamine, the process comprising: a. admixing galantamine or a pharmaceutically acceptable salt thereof with one or more water insoluble excipients to form a drug core; b. optionally applying a seal coat to the drug core; and c. applying a release-controlling coating over the core.
13. The process according of claim 12, wherein the one or more water insoluble excipients comprise silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
14. The process according to claim 12, wherein the rate-controlling coating comprises one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
15. The process according to claim 14, wherein the water insoluble polymer comprises ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
16. The process according to claim 14, wherein the water soluble polymer comprises polyvinylpyrrolidone and hydroxypropylmethylcellulose.
17. The process according to claim 14, wherein the one or more plasticizers comprise triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
18. A method of treating alzheimer ' s dementia in a patient in need thereof, the method comprising administering a controlled release dosage form of galantamine comprising: a. a core comprising galantamine or a pharmaceutically acceptable salt and one or more of water insoluble excipients; and b. a rate-controlling coating.
19. The method according of claim 18 , wherein the one or more water insoluble excipients comprise silicon dioxide, ethylcellulose, crospovidone, sodium starch glycolate, crosscarmellose sodium, microcrystalline cellulose and methacrylate polymers.
20. The method according to claim 18, wherein the rate-controlling coating comprises one or more water insoluble polymers, a water soluble polymer and one or more plasticizers.
21. The method according to claim 18, wherein the water insoluble polymer comprises ethylcellulose, cellulose acetate, ammonio methacrylate copolymer and methacrylic acid copolymer.
22. The method according to claim 18, wherein the water soluble polymer comprises polyvinylpyrrolidone and hydroxypropylmethylcellulose.
23. The method according to claim 18, wherein the one or more plasticizers comprise triacetin, triethyl citrate, dibutyl phthalate, diethyl phthalate and polyethylene glycol.
PCT/IB2006/002406 2005-09-05 2006-09-01 Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use for the manufacture of a medicament WO2007029081A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP06795399A EP1933816A1 (en) 2005-09-05 2006-09-01 Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use for the manufacture of a medicament
US12/065,773 US20090087488A1 (en) 2005-09-05 2006-09-01 Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use of the manufacture of a medicament

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN2376/DEL/2005 2005-09-05
IN2376DE2005 2005-09-05

Publications (1)

Publication Number Publication Date
WO2007029081A1 true WO2007029081A1 (en) 2007-03-15

Family

ID=37682551

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2006/002406 WO2007029081A1 (en) 2005-09-05 2006-09-01 Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use for the manufacture of a medicament

Country Status (3)

Country Link
US (1) US20090087488A1 (en)
EP (1) EP1933816A1 (en)
WO (1) WO2007029081A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008048469A2 (en) * 2006-10-13 2008-04-24 Actavis Group Pct Hf Controlled-release coated dosage forms containing galantamine
WO2008140853A2 (en) * 2007-05-08 2008-11-20 Dow Global Technologies Inc. Water dispersible polymer compositions
EP2010158A1 (en) * 2006-04-26 2009-01-07 Alphapharm Pty Ltd. Controlled release formulations comprising uncoated discrete unit(s) and an extended release matrix
WO2009024858A1 (en) * 2007-08-22 2009-02-26 Aurobindo Pharma Limited Controlled release dosage form of galantamine
EP2116232A1 (en) 2008-05-09 2009-11-11 Ratiopharm GmbH Medicine containing galanthamine with controlled release
EP2152865A1 (en) * 2007-05-11 2010-02-17 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
WO2011093535A1 (en) * 2010-01-26 2011-08-04 현대약품 주식회사 Matrix pharmaceutical composition containing galantamine
WO2013013776A1 (en) 2011-07-26 2013-01-31 Pharmathen S.A. Prolonged release pharmaceutical composition comprising galantamine and method for the preparation thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000038686A1 (en) * 1998-12-24 2000-07-06 Janssen Pharmaceutica N.V. Controlled release galantamine composition
WO2005065661A2 (en) * 2003-12-31 2005-07-21 Actavis Group Hf Immediate, controlled and sustained release formulations of galanthamine

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4248856A (en) * 1979-07-10 1981-02-03 American Home Products Corporation Sustained release pharmaceutical compositions

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000038686A1 (en) * 1998-12-24 2000-07-06 Janssen Pharmaceutica N.V. Controlled release galantamine composition
WO2005065661A2 (en) * 2003-12-31 2005-07-21 Actavis Group Hf Immediate, controlled and sustained release formulations of galanthamine
US20050191349A1 (en) * 2003-12-31 2005-09-01 Garth Boehm Galantamine formulations

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2010158A1 (en) * 2006-04-26 2009-01-07 Alphapharm Pty Ltd. Controlled release formulations comprising uncoated discrete unit(s) and an extended release matrix
EP2010158B1 (en) 2006-04-26 2016-02-17 Alphapharm Pty Ltd. Controlled release formulations comprising uncoated discrete unit(s) and an extended release matrix
EP2010158A4 (en) * 2006-04-26 2012-10-03 Alphapharm Pty Ltd Controlled release formulations comprising uncoated discrete unit(s) and an extended release matrix
US7955622B2 (en) 2006-10-13 2011-06-07 Actavis Group Ptc Hf Controlled-release galantamine formulations
WO2008048469A2 (en) * 2006-10-13 2008-04-24 Actavis Group Pct Hf Controlled-release coated dosage forms containing galantamine
WO2008048469A3 (en) * 2006-10-13 2008-06-05 Actavis Group Pct Hf Controlled-release coated dosage forms containing galantamine
WO2008140853A3 (en) * 2007-05-08 2010-07-15 Dow Global Technologies Inc. Water dispersible polymer compositions
CN101896171B (en) * 2007-05-08 2012-05-23 陶氏环球技术公司 Water dispersible polymer compositions
WO2008140853A2 (en) * 2007-05-08 2008-11-20 Dow Global Technologies Inc. Water dispersible polymer compositions
EP2152865A1 (en) * 2007-05-11 2010-02-17 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
EP2152865A4 (en) * 2007-05-11 2010-11-03 Univ Jefferson Methods of treatment and prevention of neurodegenerative diseases and disorders
WO2009024858A1 (en) * 2007-08-22 2009-02-26 Aurobindo Pharma Limited Controlled release dosage form of galantamine
EP2116232A1 (en) 2008-05-09 2009-11-11 Ratiopharm GmbH Medicine containing galanthamine with controlled release
WO2011093535A1 (en) * 2010-01-26 2011-08-04 현대약품 주식회사 Matrix pharmaceutical composition containing galantamine
WO2013013776A1 (en) 2011-07-26 2013-01-31 Pharmathen S.A. Prolonged release pharmaceutical composition comprising galantamine and method for the preparation thereof

Also Published As

Publication number Publication date
EP1933816A1 (en) 2008-06-25
US20090087488A1 (en) 2009-04-02

Similar Documents

Publication Publication Date Title
RU2328274C2 (en) Medicinal agent lamotrigin with regulated release
TWI425944B (en) Sustained-release formulation of zonisamide
US20060093671A1 (en) Controlled release galantamine composition
US20090087488A1 (en) Galantamine-containing controlled release oral dosage forms, processes for the preparation thereof and use of the manufacture of a medicament
US20050169985A1 (en) Extended release formulation of venlafaxine hydrochloride
WO2009034541A2 (en) Controlled release pharmaceutical dosage forms of trimetazidine
CA2742680C (en) Pharmaceutical compositions for release control of methylphenidate
JP3017040B2 (en) Sustained-release pharmaceutical composition for oral administration of trimetazidine
JP6148252B2 (en) New formulation
EP3813831B1 (en) Extended release compositions comprising trihexyphenidyl
WO2004056336A2 (en) Controlled release, multiple unit drug delivery systems
AU2011235222B2 (en) Stabilized formulations of CNS compounds
US20110244050A1 (en) Pulsed-release sildenafil composition and method for preparing said composition
PL192950B1 (en) Multiparticulate pharmaceutical form with programmed and timed release and preparation method
EP3981390A1 (en) Lacosamide pharmaceutical composition and pharmaceutical preparation thereof
EP2698150A1 (en) Oral solid preparation of compound antituberculosis drug and preparation method thereof
KR102241487B1 (en) Pharmaceutical composition consisting of sustained-release pellets
KR20070044911A (en) Controlled-release formulation containing tamsulosin hydrochloride
CA2370355A1 (en) Multiparticulate formulations of lithium salts for oral administration suitable for once-a-day administration
WO2015037019A2 (en) Modified release pharmaceutical formulations
US20150037405A1 (en) Pharmaceutical compositions of levodopa and carbidopa
EP3796908B1 (en) Controlled release propiverine formulations
KR20160127405A (en) A sustained releasing Pharmaceutical Composition comprising Rivastigmine
EP2736496B1 (en) Pharmaceutical composition containing an antimuscarinic agent and method for the preparation thereof
KR20110122287A (en) Release controlled pharmaceutical composition comprising tramadol or pharmaceutically acceptable salts thereof, and oral dosage form comprising the composition

Legal Events

Date Code Title Description
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: 1248/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006795399

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 2006795399

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12065773

Country of ref document: US