CA2537103C - Once daily dosage forms of trospium - Google Patents

Once daily dosage forms of trospium Download PDF

Info

Publication number
CA2537103C
CA2537103C CA002537103A CA2537103A CA2537103C CA 2537103 C CA2537103 C CA 2537103C CA 002537103 A CA002537103 A CA 002537103A CA 2537103 A CA2537103 A CA 2537103A CA 2537103 C CA2537103 C CA 2537103C
Authority
CA
Canada
Prior art keywords
component
trospium
trospium chloride
composition
release
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002537103A
Other languages
French (fr)
Other versions
CA2537103A1 (en
Inventor
Argaw Kidane
Henry H. Flanner
Padmanabh Bhatt
Arash Raoufinia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Supernus Pharmaceuticals Inc
Original Assignee
TCD ROYALTY SUB LLC
Supernus Pharmaceuticals Inc
Shire Laboratories Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34594857&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2537103(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by TCD ROYALTY SUB LLC, Supernus Pharmaceuticals Inc, Shire Laboratories Inc filed Critical TCD ROYALTY SUB LLC
Publication of CA2537103A1 publication Critical patent/CA2537103A1/en
Application granted granted Critical
Publication of CA2537103C publication Critical patent/CA2537103C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, 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
    • A61K9/14Particulate 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
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • 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
    • A61K31/468-Azabicyclo [3.2.1] octane; Derivatives thereof, e.g. atropine, cocaine
    • 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
    • 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
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; 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
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5026Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • A61K9/5047Cellulose ethers containing no ester groups, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5073Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings
    • A61K9/5078Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals having two or more different coatings optionally including drug-containing subcoatings with drug-free core
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/06Anti-spasmodics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder

Abstract

A pharmaceutical composition of a pharmaceutically acceptable trospium salt, with upon administration to a human patient generates an average steady state blood levels of trospium with a minimum (Cmin) and maximum (Cmax) blood levels of about 0.5-2.5 ng/ml and about 2.0-6.0 ng/ml, respectively.

Description

50399-18(S) Once Daily Dosage Forms of Trospium Field of the invention The present invention is directed to pharmaceutical compositions that allow for once daily dosage forms of trospium. Trospium is indicated in the treatment of urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and detrusor hyperreflexia. These compositions are useful in treating the afore-mentioned conditions with once-a-day administration.
Backqround of the invention Trospium is a quaternary ammonium derivative of tropine, and has anticholinergic properties. The hydrophilicity of the molecule, due to its permanent positive charge, limits its lipid solubility. Trospium chloride has been shown to antagonize acetylcholine on excised strips of human bladder muscle.
Antispasmodic activity has been shown in the bladder, the small intestine, and on contractility of the gall bladder. Trospium chloride exhibits parasympatholytic action by reducing smooth muscle tone, such as is found in the urogenital and gastrointestinal tracts.
This mechanism enables the detrusor to relax, thus inhibiting the evacuation of the bladder. Lowering the maximum detrusor pressure results in improved adaptation of the detrusor to the contents of the bladder, which in turn leads to enhanced bladder compliance with increased bladder capacity.

Trospium chloride was introduced into the market as a spasmolytic agent in 1967 (German patent 1 194 422). Trospium chloride has been available in an orally administrable, solid administration form (tablets and dragees), for intravenous or intramuscular injection as a solution, and for rectal administration as suppositories, and is primarily used for the treatment of bladder dysfunctions (urge incontinence, detrusor hyperrefiexia). The product has been on the market in Germany and several other European countries for a number of years for specific therapeutic indications including urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and detrusor hyperreflexia.

Currently, in the European market there is an immediate release trospium chloride tablet (Spasmo-lyt ), which is indicated for the treatment of urge incontinence and detrusor hyperreflexia and is used as a 20 mg tablet taken twice daily or bid (a total dose of 40 mg per day). In common with other quaternary ammonium compounds, orally administered,trospium chloride is slowly absorbed, with the maximum blood level achieved after 5-6 hrs. The oral bioavailability is approximately 10%, and is significantly reduced with the intake of high-fat food-.
There are side effects associated with the use of the twice-daily trospium chloride regimen, such as dry mouth, headache, constipation, dyspepsia, and abdominal pain. These side effects are associated with a high blood concentration of trospium chloride. Moreover, studies in which a 40 mg immediate release dose,was given once daily resulted in higher overall incidence of adverse events as compared to 20 mg given twice daily.

A once-a-day administration of trospium is advantageous over the twice-a-day administration in terms of both patient compliance and reduced adverse events, thus providing better treatment of the conditions for which trospium chloride is indicated.
2 In order to provide for an effective once-a-day form of trospium, there is a need for unique formulation approaches that provide the desired therapeutic effects while minimizing, if not eliminating, the undesired side effects mentioned above.
This means that the minimum blood trospium concentration (Cmin) at steady state should be above the minimum therapeutically effective blood concentration and the maximum blood trospium concentration (Cma.x) also at steady state should be below the maximum toxic blood concentration over the treatment period. Trospium chloride and other quaternary ammonium compounds exhibit a limited window of absorption in the human gastrointestinal tract, presenting a significant challenge to formulating a once-a-day composition.

Summary of the Invention It is an object of the present invention to provide a pharmaceutical composition of any pharmaceutically acceptable trospium salt, typically trospium chloride, which can be given once a day yet meet the steady state blood levels required for the treatment or prevention of diseases or conditions that would benefit from its spasmolytic activity. Such a disease or condition includes, and the present invention is primarily directed to, such bladder dysfunctions as urge incontinence or detrusor hyperreflexia, nocturia, and urinary frequency.

Such once-daily compositions of a trospium salt are targeted to result in average steady state blood levels of trospium with a minimum (Cmin) and maximum (Cmax) blood levels of about 0.5-2.5 ng/ml and about 2.0-6.0 ng/ml, respectively, which blood levels have been shown to be safe and effective. Steady state blood levels preferably average between a Cm;n of about 0.75 ng/ml and a Cmax of about 5.0
3 50399-16(S) ng/ml, for dosage forms of the present invention that correspond to a 20 mg bid regimen.

In one aspect of the invention an extended release (XR) pharmaceutical composition is provided, which contains between about 25 mg and about 60 mg trospium chloride for once-a-day or qd administration, and which is characterized by having the following in vitro release profile in phosphate buffer (pH 7.5) dissoiution medium: about 0-40% released in about 1 hour, about 20-85% released in about 4 hours and greater than 70% released in about 12 hours.

In yet another aspect of this invention a delayed release (DR) pharmaceutical composition is provided, which contains between about 25 mg and about 80 mg trospium chloride for once-a-day or qd administration, depending on the length of the lag phase. The in vivo delay in the release can be tailored to a particular application, but generally is from about 0.5 hour to about 6 hours, more preferably from about 2.5 hours to about 5 hours, during which time there should minimal, if any, detectable trospium in the blood. The in vitro release profile of such a formulation is generally characterized by having less than about 10% released in acidic media within 2 hours and more than about 80% released in buffer media of pH 6.8 and higher within 1 hour.

In still another aspect of the present invention an immediate release (IR) composition is provided, which contains no more than about 20 mg active drug, combined with a delayed release composition that is designed to dissolve at a pH of about 7.0 (i.e.,in the lower part of the GI tract), such as the DR2 composition of the examples, to form a single once-a-day trospium chloride formulation containing in total about 80 mg drug,
4 50399-18(S) Another aspect of the present invention relates to an oral pharmaceutical composition comprising (1) between 25 mg and 80 mg of trospium chloride and (2) at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH

above 4.5, and an extended release (XR) component comprising from lo to 40% of a release controlling polymer, wherein at least a portion of said trospium chloride is contained in said at least one component whereby at a once daily dosage the composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

Another aspect of the present invention relates to use of the trospium composition as described herein in a mammal, in the manufacture of a medicament which provides a daily dose for treating a urinary incontinence disease or condition in the mammal.

Another aspect of the present invention relates to a pharmaceutical composition for a once-a-day administration of trospium comprising (1) a total amount of trospium chloride which is less than 80 mg and (2) at least one component selected from an extended release (XR) component comprised of extended release particulates comprising from lo to 40% of a release controlling polymer; and a delayed release (DR) component comprised of delayed release particulates comprising an enteric polymer that dissolves at pH above 4.5, wherein at least a portion of said trospium chloride is contained in the particulates of said at least one component whereby once-a-day administration of said pharmaceutical composition provides steady state blood levels of trospium that are substantially equivalent to steady state blood levels of trospium achieved with twice 4a '50399-18(S) daily administration of 20 mg immediate release trospium chloride tablets.

Another aspect of the present invention relates to use of a once-a-day formulation comprising a therapeutically effective amount of trospium chloride in the form of at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH
above 4.5, and an extended release (XR) component comprising from lo to 40% of a release controlling polymer, which provides steady state blood levels of trospium substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg immediate release trospium chloride tablets and with a lessening of side effects, in the manufacture of a medicament for treating a mammal suffering from overactive bladder that would benefit from a once daily administration of a therapeutically effective amount of trospium chloride.

Another aspect of the present invention relates to use of a once-a-day formulation comprising a therapeutically effective amount of trospium chloride in the form of at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH
above 4.5, and an extended release (XR) component comprising from lo to 400 of a release controlling polymer, which provides steady state blood levels of trospium substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg immediate release trospium chloride tablets and with a lessening of side effects, for treating a mammal suffering from overactive bladder that would benefit from a once daily administration of a therapeutically effective amount of trospium chloride.
4b '50399-18 (S) A further aspect of the present invention relates to use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride at a pH
of about 7.0 for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

A further aspect of the present invention relates to use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride in the lower intestine for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

A further aspect of the present invention relates to use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride in the colon for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

Another aspect of the present invention relates to a pharmaceutical composition comprising from 25 to 80 mg of trospium chloride in which at least a portion of said trospium chloride is contained in a delayed release 4c 50399-18(S) formulation, which comprises an enteric polymer that dissolves at a pH of about 7.0, releasing trospium chloride, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

Another aspect of the present invention relates to a pharmaceutical composition comprising from 25 to 80 mg trospium chloride in which at least a portion of said trospium chloride is contained in a delayed release formulation, which comprises an enteric polymer that dissolves in the lower intestine, colon, or both, releasing trospium chloride, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.

Another aspect of the present invention relates to a process of preparation of a once daily dosage unit of trospium chloride, which will give steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, comprising the steps of: 1. preparing at least one component selected from an extended release (XR) component and a delayed release (DR) component; 2.
optionally, combining the result of step 1 with at least one component selected from an XR component, a DR component and an immediate release (IR) component; and 3. encapsulating the result of step 1 or step 2 into a capsule, wherein the total dose of trospium chloride is less than about 80 mg.
Another aspect of the present invention relates to an oral pharmaceutical composition of trospium comprising 25 to 80 mg of trospium chloride and a carrier, wherein at least part of the trospium chloride is contained in an 4d '50399-18(S) extended release (XR) component or in a delayed release (DR) component, and wherein said composition at a once daily dosage provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.
Another aspect of the present invention relates to an oral pharmaceutical composition of trospium comprising at least one component selected from a delayed release (DR) component and an extended release (XR) component, and a carrier, wherein said composition contains between 25 mg and 80 mg of trospium chloride and at once daily dosage provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, while minimizing the occurrence of adverse side effects as compared to an immediate release formulation containing 40 mg of trospium chloride.

Another aspect of the present invention relates to an oral pharmaceutical composition of trospium, containing a once daily dose of trospium chloride and comprising a combination of a delayed release (DR) component comprising an enteric polymer that dissolves at pH above 4.5, and an extended release (XR) component comprising from 1% to 40% of a release controlling polymer, wherein said DR component is composed of enteric coated pellets consisting essentially of: 1. trospium chloride, 2. a sugar core, 3. hydroxypropyl methylcellulose, 4. a coating of the enteric polymer that delays release of the trospium chloride from the pellet for a period of time after administration, 5. triethyl citrate, 6. a protective overcoating, and 7. talc; and said XR
component is composed of extended release pellets consisting essentially of: 1. trospium chloride, 2. a sugar core, 3.
hydroxypropyl methylcellulose, 4. a surface coating that 4e '50399-18 (S) controls a release profile of the trospium chloride from the pellet after administration, 5. a protective overcoating, and 6. talc; and the once daily dose provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.

A further aspect of the present invention relates to an oral pharmaceutical composition of trospium, containing a once daily dose of trospium chloride and comprising a mixture of enteric coated pellets and extended release pellets in a ratio of 1:1, the enteric coated pellets consisting essentially of trospium chloride, sugar spheres, hydroxypropyl methylcellulose, methacrylic acid copolymer, triethyl citrate, and talc and wherein the extended release pellets consist essentially of trospium chloride, sugar spheres, hydroxypropyl methylcellulose, ethyl cellulose, OPADRY WHITETM and talc; and the once daily dose provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.
4f Another aspect of the present invention is to provide a method for treating urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and detrusor hyperreflexia with once-a-day administration of trospium chloride.

Yet another aspect of the invention provides a single dosage form that allows for an additional release, or pulse, of a drug with a short half-life at about the half-life (t,12) thereof. Such dosage forms are a significant challenge to develop when the drug is one, such as tropsium, that has a defined region of absorption in the upper GI
tract, and is more poorly absorbed in the lower GI tract (i.e. the ileum area and colon).

The invention is also directed to a method of enteral administration of a pharmaceutical composition comprising an effective amount of a salt of trospium (e.g., trospium chloride), in which an improvement comprises including a delayed release formulation of said salt of trospium, which releases trospium at a pH
of about 7Ø In another embodiment of the invention, a method is provided for an enteral administration of a pharmaceutical composition comprising an effective amount of a trospium salt (e.g., trospium chloride), in which an improvement comprises including a delayed release formulation of said trospium salt, which releases trospium in the lower intestine, preferably in the colon. Accordingly, the invention is further directed to a pharmaceutical composition comprising a trospium salt (preferably, trospium chloride) as at least one active pharmaceutical ingredient in which at least a portion of said trospium salt is contained in a delayed release formulation, which releases trospium at a pH of about 7Ø In an alternative embodiment, the invention is still further directed to a pharmaceutical composition comprising a trospium salt (preferably, trospium chloride) as at least one active pharmaceutical ingredient in
5 which at least a portion of said trospium salt is contained in a delayed release formulation, which releases trospium in the lower intestine, colon, or both.

Finally, another aspect of the invention is to provide processes for preparing the once daily compositions of the present invention, and methods of treatment using them.

Brief Description of the Drawings Figure 1 shows the dissolution profiles for the immediate release trospium chloride pellets in 0.1 N HCI, pH 1.1. The profiles show a release that reaches completion in about 15 minutes.

Figure 2 shows the dissolution profiles for ethylcellulose coated (extended release or "XR") trospium pellets.

Figure 3 shows the dissolution profiles for trospium chloride delayed release ("DR") pellets.

Figure 4 shows the mean dissolution profile for 50 mg trospium chloride XR1-2 pellets.

Figure 5 shows the mean dissolution profile for 40 mg trospium chloride XR1-1 pellets.

Figure 6 shows the mean dissolution profile for 35 mg trospium chloride DR1 pellets.

Figure 7 shows the mean dissolution profile for 40 mg trospium chloride DR2 pellets.

Figure 8 shows the mean dissolution profile for 60 mg trospium chloride XR1 /DR2 pellets.
6 50399-18(S) Figure 9 shows the pharmacokinetic profiles of four exemplary controlled release compositions versus two immediate release products.

Figure 10 shows the same data illustrated in Figure 4 with Formulation D
removed for ease of comparison.

Figure 11 shows the steady state pharmacokinetic profiles of four trospium chloride controlled release formulations Detailed Description of the Invention The present invention is primarily directed to once-daily, orally administrable forms of trospium, which due to its charged nature is usually found in the form of a salt, typically trospium chloride. Such formulations have not been previously known, most likely because trospium chloride presents challenges due to its high solubility and limited absorption window. Moreover, previous researchers have noted that due to the limited region of absorption, conventional modified release dosage forms were not thought practical. However, the present inventors have discovered oral dosage forms, which can be given once a day yet meet the steady state blood levels required for the treatment or prevention of diseases or conditions that would benefit from its spasmolytic activity.
The present invention is accomplished by providing an orally administered composition of trospium designed to provide certain steady state blood levels of the drug comparable to a twice-a-day regimen, preferably with some refinements, yet in a formulation that requires that the mammal, preferably human, take only one dosage a day. The preferred blood level of trospium is between about 0.5 and about 6.0 ng/ml at the steady state. Preferably, the blood levels stay within the preferred
7 50399-18(S) blood level, with once daily dosing, for the course of treatment. More preferably, the blood levels are between about 0.5 ng/ml and 5.0 ng/ml at the steady state. In addition, more preferably, a suitable once-a-day formulation exhibits a C,,, within 80 to 120% of the average CR,a,, of a corresponding twice-a-day formu-ation (typically one 20 mg IR twice a day, but could be titrated up or down) and a Cmiõ between and 120% of the average CR,;, of said twice daily regimen.

The concepts of the present invention may likewise be used to formulate controlled release compositions containing therapeutically active agents that exhibit similar solubility, limited absorption window and bioavailability characteristics as trospium. Examples of such compounds include, for instance, propantheline, emepronium, clidinium, and glycopyrrolate, which all are quaternary ammonium compounds.

As used herein, "about" means within the pharmaceutically acceptable limits found in the United States Pharmacopia (USP-NF 21), 2003 Annual Edition, for amount of active pharmaceutical ingredients. With respect to blood levels, "about" means within FDA acceptable guidelines.

The compositions of the present invention may be in the form of, among others, a granule, tablet (including matrix or osmotic), pellet, powder, sachet, capsule, gel, dispersion, solution or suspension. The only requirement is that the dosage forms be composed in such a manner as to achieve the profiles set forth herein.

In vivo profiles for trospium chloride that provide the appropriate blood (or, more particularly, plasma) concentration levels over time in order to meet the therapeutic requirements for once daily administration were determined in the
8 present invention. The method used herein for the plasma concentration determination was the liquid chromatography/mass spectrometry/mass spectrometry or LC/MS/MS method. With this technique, trospium is extracted from an aliquot of plasma using a solid phase extraction procedure. This extract is then analyzed using HPLC equipped with a mass spectrometer as a detector. These profiles are such that the mean blood trospium chloride levels provide an effective amount of the drug for the treatment of such conditions as urinary frequency, urgency, nocturia, and urge-incontinence due to detrusor instability, urge syndrome, and detrusor hyperreflexia, yet within such upper limits as to minimize the occurrence of adverse side effects typically associated with spikes in the plasma concentration that follow the multiple administration of immediate release formulations. The blood trospium chloride concentrations versus time profiles are characterized by a steady state Cmiõ
of from about 0.5 to about 1.5 ng/ml, and a steady state Cmax of from about 2.0 to about 6.0 ng/ml.

With the present invention, it was surprisingly found that once daily dosing of trospium chloride in a delayed release formulation provides the =required blood profile. Moreover, it was surprisingly found that once daily dosing with a dosage unit containing a combination of immediate release and delayed components provides a desired therapeutic blood profile. Still further, it was discovered that once daily dosing of trospium chloride in an extended release preparation also provides a desired therapeutically effective blood profile.

Thus, with the present invention it was found that an effective blood trospium chloride concentration at steady state could be achieved by formulating trospium chloride in several inventive ways. These dosage units are in the form of an
9 50399-18(S) extended release, a delayed release, or various combinations of immediate, extended and delayed release forms.

Immediate release composition By "immediate release composition" is meant a dosage form that is formulated to release substantiaily all the active ingredient on administration with no enhanced, delayed or extended release effect. Such a composition for purposes of the present invention is, at least initially, in the form of a pellet (a term used interchangeably with "bead "or "beadiet" herein). The immediate release pellet can be one component of a plurality of components of a dosage form. The immediate release pellet can also serve as a precursor to an extended or delayed release pellet.

The non-active ingredients and processes for preparing such immediate release pellets are well known in the art, and the present invention is not limited in these respects. See, for example, Remington's Pharmaceutical Sciences, 18tn Edition, A. Gennaro, Ed., Mack Pub. Co. (Easton, PA 1990), Chapters 88-91.

For instance, an immediate release pellet can be prepared by mixing the trospium salt (e.g., trospium chloride) with a bulking agent. Additionally, one can add disintegrating agents, antiadherants and glidants to the formulation.

Bulking agents employable in these compositions may be chosen from, among others: microcrystalline cellulose, for example, AVICEL0 (FMC Corp.) or EMCOCEL (Mendell Inc.), which also has binder properties; dicalcium phosphate, for example, EMCOMPRESS (Mendell Inc.); calcium sulfate, for example, COMPACTROL (Mendell Inc.); and starches, for example, Starch 1500; and polyethylene glycols (CARBOWAX ). Such bulking agents are typically present in the range of about 5% to about 75% (w/w), with a preferred range of about 25%
to about 50% (w/w).

Suitable disintegrants include, but are not limited to: crosslinked sodium carboxymethyl cellulose (AC-DI-SOL ), sodium starch glycolate (EXPLOTAB

PRIMOJEL ) and crosslinked polyvinylpolypyrrolidone (Plasone-XL ).
Disintegrants are used to facilitate disintegration of the pellet upon administration and are typically present in an amount of about 3% to about 15% (w/w), with a preferred range of about 5% to about 10% (w/w).

Antiadherants and glidants employable in such formulations can include talc, cornstarch, silicon dioxide, sodium lauryl sulfate, colloidal silica dioxide, and metallic stearates, among others.

In addition, the immediate release composition may contain one or more binders to give the pellets cohesiveness. Such binders are well known in the art, and include such substances as microcrystalline cellulose, polyvinyl pyrrolidone, starch, Maltrin, methylcellulose, hydroxypropyl methylcellulose, carboxymethyl cellulose, sucrose solution, dextrose solution, acacia, tragacanth and locust bean gum, which may be applied wet. The binding agent may be present in the composition in an amount of from about 0.2 wt% to about 20 wt%, preferably from about 5 wt% to about 15 wt%.

The pellets can be made by, for example, simple granulation such as wet granulation or dry granulation, followed by sieving; extrusion and marumerization (spheronization); rotogranulation; or any agglomeration process that results in a pellet of reasonable size and robustness. For extrusion and marumerization, the drug and other additives are granulated by addition of a binder solution. The wet mass is passed through an extruder equipped with a certain size screen, and the extrudates are spheronized in a marumerizer. The resulting pellets are dried and sieved for further applications.

One may also use high-shear granulation, wherein the drug and other additives are dry-mixed and then the mixture is wetted by addition of a binder solution in a high shear-granulator/mixer. The granules are kneaded after wetting by the combined actions of mixing and milling. The resulting granules or pellets are dried and sieved for further applications.

Alternatively, and preferably, the immediate release beadlets or pellets are prepared by solution or suspension layering, whereby a drug solution or dispersion, with or without a binder and optionally an anti-tacking agent such as talc, is sprayed onto a core or starting seed (either prepared or a commercially available product) in a fluid bed processor or other suitable equipment. The cores or starting seeds can be, for example, sugar spheres or spheres made from microcrystalline cellulose.

The binder in the formula can be present in amounts ranging from about 0% to about 5% by weight, and preferably about 0.5% to about 2% by weight. The amount of anti-tacking agent used can be from about 0% to about 5%, preferably about 0.5% to about 2% by weight. The drug thus is coated on the surface of the starting seeds.
The drug may also be layered onto the drug-containing pellets described above, if desired. Following drug layering, the resulting drug-loaded pellets are dried for further applications.

A protective layer, or overcoating, may be desired to ensure that the drug-loaded pellets do not aggregate during processing or upon storage. The protective coating layer may be applied immediately outside the core, either a drug-containing core or a drug-layered core, by conventional coating techniques such as pan coating or fluid bed coating using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. OPADRY , OPADRY II (Colorcon) and corresponding color and colorless grades from Colorcon can be used to protect the pellets from being tacky and provide colors to the product. The suggested levels of protective or color coating are from about 1% to about 6%, preferably about 2%
to about 3% (w/w). Many ingredients can be incorporated into the overcoating formula, for example to provide a quicker immediate release, such as plasticizers:
acetyltriethyl citrate, triethyl citrate, acetyltributyl citrate;
dibutylsebacate, triacetin, polyethylene glycols, propylene glycol and the others; lubricants: talc, colloidal silica dioxide, magnesium stearate, calcium stearate, titanium dioxide, magnesium silicate, and the like.

The immediate release pellets are contemplated as being used in combination with extended release pellets and/or delayed release pellets in a single dosage form.
Extended release composition (XR) Trospium chloride extended release pellets can be prepared, for example, by coating drug layered inert pellets with release controlling polymers. First, the inert .pellet is coated with the drug layer or a drug loaded granule is prepared, as described above. Then the active (drug loaded) pellet is coated with a release controlling polymeric membrane. The release controlling coating layer may be applied immediately outside the core (such as a drug-containing core or a drug-layered core), by conventional coating techniques, such as pan coating or fluid bed coating, using solutions of polymers in water or suitable organic solvents or by using aqueous polymer dispersions. As an alternative embodiment, the release controlling membrane can separate additional drug layers on the core; for instance, after coating with the release controlling substance, another drug layer can be applied, which is followed by another release controlling layer, etc. Suitable materials for the release controlling layer include EUDRAGITO RL, EUDRAGITO RS, cellulose derivatives such as ethylcellulose aqueous dispersions (AQUACOATO, SURELEASEO), hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, OPADRYO, and the like. The thickness of the coating affects the release profile, and so this parameter can be used to customize the profile. The suggested coating levels are from about 1% to about 40%, preferably about 5% to about 30% (w/w), and about 20% or about 25% as most preferred embodiments. A 20% w/w coating should release about 80% of the trospium chloride in 3.5 hours post ingestion, and a 25% w/w coating should result in the release of about 80% of the trospium chloride in 4.5 hours post-ingestion.

The extended release pellets contain between about 25 and about 60 mg trospium chloride, and may be used alone, or in combination with immediate release or delayed release -pellets to constitute a single daily dosage form.

Delayed release composition (DR) The delayed-release component has a coat applied to the surface of the active pellet that delays the release of the drug from the pellet after administration for a certain period of time. This delayed release is accomplished by applying a coating of enteric materials. "Enteric materials" are polymers that are substantially insoluble in the acidic environment of the stomach, but are predominantly soluble in intestinal fluids at various specific pHs. The enteric materials are non-toxic, pharmaceutically acceptable polymers, and include, for example, cellulose acetate phthalate (CAP), hydroxypropyl methylcellulose phthalate (HPMCP), polyvinyl acetate phthalate (PVAP), hydroxypropyl methylcellulose acetate succinate (HPMCAS), cellulose acetate trimellitate, hydroxypropyl methylcellulose succinate, cellulose acetate succinate, cellulose acetate hexahydrophthalate, cellulose propionate phthalate, copolymer of methylmethacrylic acid and methyl methacrylate, copolymer of methyl .5 acrylate, methylmethacrylate and methacrylic acid, copolymer of methylvinyl ether and maleic anhydride (Gantrez ES series), ethyl methyacrylate-methylmethacrylate-chlorotrimethylammonium ethyl acrylate copolymer, natural resins such as zein, shellac and copal collophorium, carboxymethyl ethylcellulose, co-polymerized methacrylic acid/methacrylic acid methyl esters such as, for instance, materials known under the trade name EUDRAGITOL12.5, L100, or EUDRAGITOS12.5, S100, and several commercially available enteric dispersion systems (e.g., EUDRAGITO L30D55, EUDRAGITO FS30D, EUDRAGITO L100-55, EUDRAGITO
S100 (Rohm Pharma), KOLLICOATO MAE30D and 30DP (BASF), ESTACRYLO
30D (Eastman,Chemical), AQUATERICO and AQUACOATO CPD30 (FMC)). The foregoing is a list of possible~ materials, but one of skill in the art would appreciate that there are other such materials that would meet the objectives of the present invention of providing for a delayed release profile including tailoring release based on the ambient pH environment, temporal considerations and other factors.

These coating materials can be employed in coating the surfaces in a range of from about 1.0% (w/w) to about 50% (w/w) of the pellet composition.
Preferably, these coating materials are in the range of from about 20 percent to about 40 percent (w/w). The pellets may be coated in a fluidized bed apparatus or pan coating, for example, in a conventional manner.

With the enteric-coated pellets, there is no substantial release of trospium in the acidic stomach environment of below about pH 4.5. The trospium becomes available when the pH-sensitive enteric layer dissolves at a higher pH in the GI tract, after a certain delayed time, or after the unit passes through the stomach.
The preferred delay time is in the range of about 0.5 to about 6 hours, but more preferable is about 0.5 to about 4 hours.

More particularly, preferred DR pellets are those that are coated with Eudragit L30D-55 (which dissolves at about pH 5.5 - 6.0, i.e., in the upper intestines), and others that are coated with Eudragit FS30D (which dissolves at about pH 7.0, i.e. in the lower intestine and colon).

As a variation of this embodiment, the DR pellet contains layers of the trospium, separated by protective (XR) or release-controlling (DR) layers, optionally surrounded by an IR layer, which will result in a pulsed dose delivery; in other words, a combination of an IR or XR with a DR in the same pellet. Such a dosage form is made as an alternative way to meet the blood level requirements of the release profile of the present invention, which may be comparable to separate IR/XR
and IR/DR pellets in the same capsule.

Preferably, the DR pellets are used in combination with XR pellets, but may also be used with IR pellets or a combination of all three.

Immediate Release (IR) /Delayed Release(DR) Dosage Units Pulsatile drug release can be achieved through a combination of immediate release and delayed release,components in a single dosage form. For instance, a combination of the immediate release (IR) pellets and delayed release (DR) pellets described herein can be employed. With this approach, pellets coated with enteric polymer (DR pellets) are combined with drug-coated pellets (IR pellets) to provide an immediate release followed by a pulsed release of trospium. Whereas the IR
portion provides a fast rise in the plasma-time profile, the DR portion helps ensure that an effective plasma level is maintained over a longer period of time, preferably a 24 hour period.

The ratio between the immediate-release component and the delayed-release component can be used to adjust the in vitro drug release profile and in vivo blood concentration profile. Moreover, the profile can be manipulated by the properties of the delayed-release coating. By providing the desired drug release profiles according to the present invention, the compositions eliminate the need for a second dose for the day. Additionally, the total dose of trospium is preferably at or below 80 mg to avoid undesirable side effects but more than 30 mg to achieve the desired antispasmodic effect.

Immediate Release (IR)/Extended Release (XR) Dosage Units As an alternative embodiment, the once-daily dosage unit may contain a combination of IR and XR pellets, in ratios designed to be substantially equivalent to a twice a day regimen, or otherwise provide a once-daily dosage form that will be safe and effective with minimized side effects.

The immediate release portion is designed to provide an effective plasma level at early time points. The extended release portion of the dosage form is designed to maintain the effective blood level throughout a 24-hour period, thus providing coverage for 24 hr. The IR portion provides about 20 mg or less of trospium to provide effective blood levels and yet avoid the side effects associated with spikes in the plasma profile. The extended release portion provides about mg to about 60 mg of trospium chloride, more preferably from about 20 to about mg in the extended release form.

Immediate Release (IR)/DelaYed Release (DR)/Extended Release (XR) Dosage Units Yet another embodiment of the present invention is a multiparticulate dosage form, which combines the three types of pellets. This type of dosage unit will provide multiple pulses of drug release, with the effect being a more or less sustained blood level of trospium within the acceptable range.

With this combination, the IR portion is designed to provide an effective blood level soon after ingestion, which is maintained by the DR and XR combinations.
The DR portion provides an immediate release after a delay. The XR portion provides an extended release profile that maintains the effective blood level of trospium throughout the course of the day. The total dose of trospium in this composition is no greater than 80 mg, preferably 60 mg with the IR portion accounting for a maximum,of about 20 mg of the total. The DR and XR portions account for 10 mg to 60 mg combined with ratios of XR to DR ranging form about 1:10 to about 10:1.
Delayed Release (DR)/Extended Release (XR) Dosage Units Yet another embodiment of the present invention is a multiparticulate dosage form, which combines the extended release pellets with delayed release pellets.
While the XR portion provides a sustained blood profile, the DR portion prevents the blood level form falling below the effective level at later time points. The XR is designed to provide between 10 mg to 40 mg, preferably between 20 mg to 40 mg, and more preferably 30 mg trospium chloride. The DR portion is preferably a longer delayed release with a delay of about 2-4 hrs and provides between 10 mg and mg, preferably between 20 mg and 40 mg, and more preferably 30 mg trospium chloride.

Dosage Forms As noted previously herein, the compositions of the present invention can be in a number of different forms, such as tablets, powders, suspensions, solutions, etc.
The composition is preferably in pellet/beadiet form, which can be incorporated into hard gelatin or other kinds of capsules, either with additional excipients, or alone.

Typical excipients to be added to a capsule formulation include, but are not limited to: fillers such as microcrystalline cellulose, soy polysaccharides, calcium phosphate dihydrate, calcium sulfate, lactose, sucrose, sorbitol, or any other inert filler. In addition, there can be flow aids such as fumed silicon dioxide, silica gel, magnesium stearate, calcium stearate or any other materials that impart good flow properties. A

lubricant can also be added if desired, such as polyethylene glycol, leucine, glyceryl behenate, magnesium stearate or calcium stearate. The multiparticulate capsules are preferred because they provide an increased surface area as opposed to a tablet, which allows for better release profiles and thus bioavailability.

However, the pellets described above can be incorporated into a tablet, in particular by incorporation into a tablet matrix, which rapidly disperses the particles after ingestion. In order to incorporate these particles into such a tablet, a filler/binder must be used in the tableting process that will not allow the destruction of the pellets during the tableting process. Materials that are suitable for this purpose include, but are not limited to, microcrystalline cellulose (AVICELO), soy polysaccharide (EMCOSOYO), pre-gelatinized starches (STARCHO 1500, NATIONAL 1551), and polyethylene glycols (CARBOWAX ). These materials should be present in the range of about 5%-75% (w/w), and preferably between about 25%-50% (w/w).

In addition, disintegrants are added to the tablets in order to disperse the beads once the tablet is ingested. Suitable disintegrants include, but are not limited to: crosslinked sodium carboxymethyl cellulose (AC-DI-SOL ), sodium starch glycolate (EXPLOTAB , PRIMOJEL&), and crosslinked polyvinylpolypyrrolidone (Plasone-XL). These materials should be present in the range of about 3 /o-15%
(w/w), with a preferred range of about 5%-10% (w/w).

Lubricants are also added to assure proper tableting, and these can include, but are not limited to: magnesium stearate, calcium stearate, stearic acid, polyethylene glycol, leucine, glyceryl behenate, and hydrogenated vegetable oil.
These lubricants should be present in amounts from about 0.1 %-10% (w/w), with a preferred range of about 0.3%-3.0% (w/w).

Tablets are formed, for example, as follows. The pellets are introduced into a blender along with AVICEM, disintegrants and lubricant, mixed for a set number of minutes to provide a homogeneous blend which is then put in the hopper of a tablet press with which tablets are compressed. The compression force used is adequate to form a tablet; however, it is not sufficient to fracture the beadlets or coatings.

A pharmaceutical formulation for the delivery of trospium chloride for the effective treatment of urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and/or detrusor hyperreflexia in a human patient comprising an extended release composition that provides an extended release upon oral administration to said patient; and a pharmaceutical acceptable carrier; wherein the pharmaceutical formulation is sufficient to maintain an effective level of trospium chloride in the patient over the course of at least 12 hours without further administration of trospium chloride . The total dosage of trospium chloride may be about 30 mg to 70 mg producing in a human patient a plasma concentration versus time curve having an area under the curve of about 30,000pg-Hr/ml to about 80,000pg-Hr/ml . The plasma concentration may have a maximum concentration of about 1.5ng/ml to about 6.Ong/ml . The plasma concentration may have a minimum concentration of about 0.5ng/ml to about 1.5ng/ml . The maximum concentration of value of the said plasma concentration curve may be reached in about 3 to about 24 hours after oral administration.

A pharmaceutical formulation for the delivery of trospium chloride for the effective treatment of urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and/or detrusor hyperreflexia in a human patient comprising an extended release composition that provides a delayed release upon oral administration to said patient; and a pharmaceutical acceptable carrier; wherein the pharmaceutical formulation is sufficient to maintain an effective level of trospium chloride in the patient over the course of at least 12 hours without further administration of trospium chloride . The total dosage of trospium chloride may be about 30 to 70 mg producing in a human patient a plasma concentration versus time curve having an area under the curve of about 30,000 pg/ml*hr I to about 80,000 pg/ml*hr. The plasma concentration may have a maximum concentration of about 1.5ng/ml to about 6.Ong/ml . The plasma concentration may have a minimum concentration of about 0.5ng/ml to about 1.5ng/ml . The maximum concentration of value of the said plasma concentration curve may be reached in about 3 to about 24 hours after oral administration.

A pharmaceutical formulation for the delivery of trospium chloride for the effective treatment of urinary frequency, urgency, nocturia, and urge-incontinence associated with detrusor instability, urge syndrome, and/or detrusor hyperreflexia in a human patient comprising an immediate release and/or an extended release composition that provides an immediate release and/or an extended release upon oral administration to said patient; a delayed release composition that provides delayed release upon oral administration to said patient; and a pharmaceutical acceptable carrier; wherein the pharmaceutical formulation is sufficient to maintain an effective level of trospium chloride in the patient over the course of at least 12 hours without further administration of trospium chloride , and a peak plasma concentration of the trospium chloride reached after release of said delayed release composition exceeds the peak plasma concentration previously reached after release of said immediate release composition or extended release composition.
The total dosage of trospium chloride may be about 30 to 70 mg producing in a human patient a plasma concentration versus time curve having an area under the curve of about 30,000 pg/ml*hr to about to about 80,000 pg/ml*hr. The plasma concentration may have a maximum concentration of about 1.5ng/ml to about 6.Ong/ml . The plasma concentration may have a minimum concentration of about 0.5ng/ml to about 1.5ng/mi. The maximum concentration of value of the said plasma concentration curve may be reached in about 3 to about 24 hours after oral administration.

A once a day pharmaceutical formulation of trospium chloride comprising an immediate release or an extended release composition combined with a delayed release composition wherein the formulation composition contains sufficient trospium chloride to obtain a mean blood plasma trospium concentration in a human patient is about 500 pg/mL to about 800 pg/mL within about 1-3 hour of oral administration; A plasma concentration versus time of the said once a day formulation has an area under the curve of about 30,000 pg/ml*hr I to about 80,000 pg/ml*hr. The maximum concentration pf said plasma concentration curve is about 1.5 ng/mL to about 6.0 ng/mL. The Tmax is about 5 and about 6 hours. The total trospium chloride dose is about 30 mg to 80 mg,per dose. The immediate release or the extended release composition has a release of trospium chloride equal to about 5% to about 20% of the total dose content within 2 hours as measured in an in vitro dissolution test using an USP Apparatus II at 50 RPM in 950m1 50mM phosphate buffer at a pH between 6.8 and 7.5 at 37 C . Said immediate release or an extended release composition combined with a delayed release composition may be in a single unit or in separate units. Said unit or units may be erodible matrix systems, coated systems, osmotic systems or combinations thereof.

The invention contemplates a pharmaceutical composition suitable for a once-a-day administration of trospium chloride comprising an amount of solid, trospium chloride-bearing particulates, each particulate including one or more trospium chloride-release-controlling substances, such that once-a-day administration of said pharmaceutical composition provides steady state (i.e., not single dose but after at least a few daily doses, or starting approximately between about 72 hours to about 120 hours of continuous once daily dosing) blood levels of trospium, which are substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of the available 20 mg immediate release trospium chloride tablets, provided that said solid, trospium chloride-bearing particulates cannot comprise trospium chloride-release-controlling substances selected exclusively from immediate release substances.

In a preferred embodiment of the invention the once-a-day administration of the controlled release pharmaceutical composition provides steady state blood levels of trospium falling in the range of about 0.5 ng/mi to about 6.0 ng/ml, and preferably, for the dosage level corresponding to the 20 mg bid regimen of trospium chloride, falling in the range of about 0.75 ng/ml to about 3.0 ng/ml. It will be understood by clinicians and others skilled in the art that patients may be titrated up or down from the conventional 20 mg bid trospium chloride dosage, in which case the dosage units of the present invention would be correspondingly adjusted.
The range of drug concentration in. the formulations of the present invention accounts for such adjustments, it being understood that the preferred drug ranges roughly correspond to the typical 20 mg bid dosage regimen.

The invention is also contemplated to provide a pharmaceutical composition in which once-a-day administration provides steady state blood CmaX levels of trospium falling in the range of about 2.5 ng/ml to about 4.5 ng/ml and Cm;,, levels of trospium falling in the range of about 0.5 ng/ml to about 1.5 ng/mI. Moreover, the invention provides pharmaceutical compositions in which once-a-day administration provides steady state areas under the curve falling in the range of about 30 to about 60 ng/ml*hr, preferably, falling in the range of about 35 to about 45 ng/ml*hr.

In a particular embodiment of the invention, pharmaceutical compositions are provided in which once-a-day administration provides steady state %F (i.e., relative bioavailability) values falling in the range of about 80 to about 120, preferably, falling in the range of about 90 to about 110.

Hence, the invention contemplates that a wide selection of one or more trospium chloride-release-controlling substances is selected for inclusion in the solid, trospium chloride-bearing particulates, including, for example, one or more trospium chloride-release-controlling substances selected from substances that provide for immediate release, delayed release, extended release, or pH-sensitive release of trospium chloride, provided that if an immediate release substance is selected, then the pharmaceutical composition also includes one or more delayed release, extended release, pH-sensitive release substances or combinations thereof.

Specific embodiments, described in further detail in the examples, include but are not limited to formulations that are designated DRI, DR2, XR, XR1-1, XR1-2, XR1 + DR2 and IR/DR2, to name a few.

The invention also contemplates a method of treating a mammal suffering from a condition that would benefit from a once daily administration of an effective amount of trospium chloride, comprising administering to a mammal in need thereof a once-a-day formulation comprising an effective amount of trospium chloride which provides steady state blood levels of trospium, which are substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg bid immediate release trospium chloride tablets (or a corresponding titrated dose) and which will lessen side effects. The invention now will be described in particularity with the following illustrative examples; however, the scope of the present invention is not intended to be, and shall not be, limited to the exemplified embodiments below.

This invention provides profiles that would make an acceptable once-a-day dosing regimen for trospium chloride. Trospium chloride is a highly water-soluble compound with a saturation solubility of 500 mg/mI. This invention overcomes the challenge imposed by highly water-soluble drugs as the active pharmaceutical ingredient in extended release preparations. Once-a-day dosing has a decided 50399-18(S) advantage over multiple dosing, increasing, for example, the rates of patient compliance. Also, once-a-day dosing with controlled release formulations reduces the side effects associated with spikes in the plasma concentration, which follow the multiple dose administration of immediate release formulations.

Thus, the present invention is also directed to methods of treating a mammal, preferably human, by administering once a day a composition according to the present invention, which will give average steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, and preferably between about 0.75 and about 5.0 ng/ml. Any of the various compositions described in this application can accomplish those blood levels, an achievement not previously thought possible. Schroder, S. et al., vide supra.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

Examples Example 1: Trospium chloride immediate release pellets Trospium chloride immediate release (IR) pellets were manufactured by coating 30/35-mesh sugar spheres with trospium chloride from a coating dispersion consisting of trospium chloride, hydroxypropylmethylcellulose (HPMC E5, a binder), talc (an anti-tacking agent), and water in a Glatt's GPCG-1 fluid bed coater.
Table 1 provides the formula composition of trospium chloride IR capsules, as well as modified release compositions, and Table 2 sets forth the composition of the pellets.
The drug layering dispersion is prepared by dissolving the HPMC E5 in water, dissolving the trospium chloride therein, then dispersing the talc, and stirring for 20 minutes. The resulting dispersion was stirred throughout the coating process to prevent settling of coating components. Coating parameters for Glatt's GPCG-1 are given in Table 3. The pellets generated contained about 20% w/w of trospium chloride.

The procedure followed to determine the dissolution profiles was:
Figure 1: USP Apparatus II, 50RPM. Media: 950ml 0.1 N HCI, pH 1.1 Figure 2: USP Apparatus II, 50RPM. Media: 950ml 50mM phosphate buffer, pH 7.5 Figure 3: USP Apparatus II, 50RPM. Media: 750m1 0.1 N HCI pH 1.1 for the first Hrs and then media adjusted to pH 6.8 at 2Hrs using phosphate buffer (total media volume = 950m1) Figure 4: USP Apparatus II, 50RPM. Media: 950m1 50mM phosphate buffer, pH 7.5 Figure 5: USP Apparatus II, 50RPM. Media: 950m1 50mM phosphate buffer, pH 7.5 Figure 6: USP Apparatus II, 50RPM. Media: 750ml 0.1 N HCI pH 1.1 for the first Hrs and then media adjusted to pH 6.8 at 2Hrs using phosphate buffer (total media volume = 950m1) Figure 7 USP Apparatus II, 50RPM. Media: 750ml 0.1 N HCI pH 1.1 for the first 2 Hrs and then media adjusted to pH 7.5 at 2Hrs using phosphate buffer (total media volume = 950m1) Figure 8: USP Apparatus II, 50RPM. Media: 750m10.1 N HCI pH 1.1 for the first 2 Hrs and then media adjusted to pH 7.5 at 2Hrs using phosphate buffer (total media volume = 950ml) Figure 1 shows the dissolution profiles for the immediate release trospium chloride pellets in 0.1 N HCI, pH 1.1. The profiles show a release greater than about 90% in about 15 minutes.

Figure 2 shows the dissolution profiles for the extended release trospium chloride pellets in phosphate buffer, pH 6.8. The profiles show a release between about 25% and about 80% in about 4 hours, between about 50% and about 95% in about 8 hours, between about 70% and about 98% in about 12 hours and between about 90% and about 100% in 24 hours.

Figure 3 shows the dissolution profiles for the delayed release trospium chloride pellets in 0.1 N HCI and phosphate buffer, pH 6.8. The profiles show a release below 1% in acidic media and a release greater than about 90% in about 15 minutes after changing the pH

Figure 4 shows the mean dissolution profiles for the extended release 50 mg trospium chloride pellets in phosphate buffer, pH 6.8. The profiles show a release of less tha 10 % in about 2 hour between about 20 % and about 30% in about 4 hours, between about 50% and about 60% in about 8 hours, between about 70% and about 80% in about 12 hours and between about 90% and about 100% in 24 hours.
Figure 5 shows the mean dissolution profiles for the extended release 40 mg trospium chloride pellets in phosphate buffer, pH 6.8. The profiles show a release of less tha 10 % in about 2 hours, between about 20 % and about 30% in about 2 hours, between about 40% and about 60% in about 4 hours, between about 80%

and about 90% in about 8 hours and between about 90% and about 100% in 12 hours.

Figure 6 shows the mean dissolution profile for delayed release 35 mg trospium chloride pellets in 0.1 N HCI and phosphate buffer, pH 6.8. The profiles show a release below 1% in acidic media and a release greater than about 40%
in about 15 minutes after changing the pH, greater than 80% in 30 minute after changing the pH and greater than 90% within an hour after changing the pH.

Figure 7 shows the mean dissolution profile for delayed release 35 mg trospium chloride pellets in 0.1 N HCI and phosphate buffer, pH 6.8. The profiles show a release below 1% in acidic media and a release greater than about 30%
in about 30 minutes after changing the pH, greater than 60% release in about 60 minutes after changing the pH and greater than 80% release within an hour after changing the pH and greater than 90% release within about 4 hour after changing the pH.

Figure 8 shows the mean dissolution profile for extended release/ delayed release 60 mg trospium chloride pellets in 0:1 N HCI and phosphate buffer, pH
6.8.
The profiles show a release between about 10% and about 20% during the first two hours in acidic media, and- a release between 30% and 40 % in about 30 minutes after changing the pH, between 60% and 70% release in about 1 hour after changing the pH, between 60% and 80% release in about 2 hours after changing the pH, between 70% and 80% release in about 4 hours after changing the pH, between 80% and 90% release in about 6 hours after changing the pH and greater than 90%
release after a period of about 8 hours after changing the pH.

All the dissolution profiles are generated at 37 C.

Table 1a. Percent weight composition of trospium chloride dosage forms Components Quantity % Quantity % Quantity % Quantity %
per unit per unit per unit per unit (mg) (mg) (mg) (mg) Trospium Chloride 40 13.64 40 9.13 40 9.23 40 11.97 Sugar Spheres, NF 152 51.84 152.1 34.71 152 35.07 152 45.48 Hydroxypropyl 2 0.68 2 0.46 2 0.46 2 0.6 methylcellulose, USP
(Methocel E5 Premium LV) Eudragit 'L30D-55 N/A N/A 110.4 25.19 N/A N/A N/A N/A
Eudragit FS30D N/A N/A N/A N/A 100 23.07 N/A N/A
Triethyl Citrate, NF N/A N/A 16.6 3.79 5.6 1.29 N/A N/A
Opadry White, YS-1- 4 1.36 10.9 2.49 10.8 2.49 8.8 2.63 Talc, USP 2 0.68 13 2.97 29.8 6.88 2 0.6 Ethyl Cellulose-based N/A N/A N/A N/A N/A N/A 36.2 10.83 Coating Dis ersion (Surelease Clear) Hard Gelatin 93.2 31.79 93.2 21.27 93.2 21.50 93.2 27.89 Capsules #0, White Opaque Total =293.2 100 438.2 100 433.4 100 334.2 100 Table 1 b. Percent weight composition of trospium chloride dosage forms 35mg DRI 40mg XR1-1 50mg XR1-2 60mg XR1; DR2 Capsules Capsules Capsules Capsules Component Quanti % Quantity % Quantity % Quanti %
fy per per unit per unit ty per unit unit (mg) (mg) (mg) (mg) Trospium Chloride 35 9.13 40 11.07 50 11.17 60 11.09 Sugar Spheres, NF 133 34.71 152 42.06 190 42.46 228 42.16 Hydroxypropyl 1.8 0.46 2 0.55 2.5 0.56 3 0.55 methylcellulose, USP (Methocel E5' Premium LV) Eudragit L30D-55 96.5 25.19 N/A N/A N/A N/A N/A N/A
Eudragit FS30D N/A N/A N/A N/A 75 13.87 Triethyl Citrate, NF 14.5 3.79 N/A N/A N/A N/A 4.2 0.78 Opadry White, YS- 9.5 2.49 9.1 2.52 11.9 2.66 14.7 2.72 Talc, USP 11.4 2.97 2 0.55 2.5 0.56 23.8 4.4 Ethyl Cellulose- N/A N/A 51.3 14.19 85.6 19.13 27.1 5.01 based Coating Dispersion Surelease Clear) Hard Gelatin 105 21.27 105 29.05 105 23.46 105 19.42 Capsules #Oel, White Opaque Total 406.7 100 361.4 100 433.4 100 540.8 100 Table 2. Percent weight composition of trospium chloride pellets Components % % % %
Trospium Chloride 20 11.59 11.76 16.6 Sugar Spheres, NF 76 44.09 44.68 63.08 Hydroxypropyl 1 0.58 0.59 0.83 methylcellulose, USP
(Methocel E5 Premium LV) Eudragit L30D-55 N/A 32 N/A N/A
Eudragit FS30D N/A N/A 29.39 N/A
Triethyl Citrate, NF N/A 4.81 1.65 N/A
Opadry White, YS-1- 2 3.16 3.17 3.66 Talc, USP 1 3.77 8.76 0.83 Ethyl Cellulose-based N/A N/A N/A 15 Coating Disersion (Surelease Clear) Total 100 100 100 100 Table 3. Coating process parameters Parameter GPCG-1 Inlet air temperature ( C) 50-55 Product temperature ( C) 40-42 Atomization air (bar) 1.5 Spray rate (g/min} 8-12 Example 2: Trospium chloride extended release pellets The composition of trospium chloride XR pellet filled capsules is provided in Table 1.
Trospium chloride XR pellets were manufactured by coating trospium chloride immediate release pellets with a Surelease Clear coating dispersion using a Glatt fluid bed coater. Surelease Clear is a 25% w/w aqueous dispersion supplied by Colorcon (West Point, PA). The Surelease Clear coating dispersion was prepared by adding water to Surelease Clear and mixing for 20 minutes to achieve a 20%w/w dispersion of Surelease Clear. This 20% w/w Surelease Clear dispersion was used for coating. The resulting dispersion was stirred throughout the coating process to prevent settling of coating components. Various coating levels of Surelease Clear were examined with the objective of achieving extended release pellets with different extents of delay in coating dissolution, which are shown in Table 4. Figure 2 shows the dissolution profiles for ethyicellulose coated trospium pellets.

Table 4. Composition of trospium chloride extended release pellets Material 15 o w/w 20% w/w 22.5% w/w 25% w/w 27.5% wlw 30 /a w/w Surelease Surelease Surelease Surelease Surelease Surelease Trospium chloride 16.6 16.0 15.5 15.0 14.5 14.0 Methocel E5 0.83 0.8 0.78 0.75 0.73 0.7 (HPMC) Sugar Spheres NF 63.08 60.8 58.9 57.0 55.1 53.2 30/35 mesh Altalc 300V (Talc 0.83 0.8 0.78 0.75 0.73 0.7 USP) Surelease Clear 15 20.0 22.5 25.0 27.5 30 Opadry White YS- 3.66 1.6 1.55 1.5 1.45 1.4 Example 3: Trospium chloride delayed release pellets The composition of trospium chloride DR pellet filled capsules is provided in Table 1. Table 2 provides the composition of delayed release pellets. Trospium chloride immediate release pellets were coated with Eudragit L30D55 from a coating dispersion consisting of Eudragit L30D55, triethylcitrate (a plasticizer), talc (an anti-tacking agent), and water using a Glatt" fluid bed coater. Eudragit is a 30% w/w aqueous dispersion supplied by Rohm America (Piscataway, NJ). The Eudragit L30D55 coating dispersion was prepared by dispersing, talc in water and mixing for at least 20 minutes. Eudragit L30D55 dispersion was sieved through an 80-mesh sieve. Triethylcitrate was added to the Eudragit"L30D55 dispersion and mixed for at least 30 minutes. The talc dispersion was then slowly poured into the Eudragit L30D55/TEC dispersion and mixed for at least 30 minutes. The resulting dispersion (an 11.7% w/w Eudragit L30D55 aqueous dispersion) was filtered through an 80-mesh sieve and stirred throughout the coating process to prevent settling of coating components. Various coating levels of Eudragit L30D55 were examined with the objective of achieving an acid resistant coating. Figure 3 shows the dissolution profiles for trospium chloride delayed release pellets.

Example 4 - Combination XR/DR

Extended release pellets were prepared as in Example 2, with a 15% w/w coating of SURELEASE . Delayed release pellets were manufactured by coating immediate release pellets with Eudragit FS30D, in a manner similar to Example 3.
Eudragit FS30D is a 30% aqueous dispersion supplied by Rohm America (Piscataway, NJ). The Eudragit FS30D coating dispersion is 18% w/w Eudragit FS30D. The enteric coated pellets are combined with extended release pellets in the XR pellets to DR pellets ratio of 1:1, to achieve a total trospium chloride dose of 60 mg.

Example 5. Trospium chloride delayed release at 35 mg strength Delayed release pellets manufactured by coating immediate release pellets with Eudragit L30D55 as described in Example 3 were filled into capsules at a fill weight that provides 35 mg trospium chloride in the capsule dosage unit.

Example 6 - Single Dose Human Pharmacokinetic Studies A human trial of four controlled release formulations was conducted. The study compared four controlled release dosage units described in the previous examples (DR1 40 mg, DR2 40 mg, XR 40 mg and a mixture of 20 mg IR:120 mg DR2) with a 40 mg IR capsule given as once daily single dose and the 20 mg IR
tablet (Spasmo-Lyt , Madaus), which was given twice a day at 12 hour intervals.

Figure 4 shows the pharmacokinetic profiles of the four once-a-day controlled release compositions versus the two immediate release products. Figure 5 shows the same data with Formulation D removed for ease of comparisons. These data demonstrate that the DR1, XR1 and the combination of IR/DR2 produced pharmacokinetic profiles and parameters that are similar to the commercial IR
twice a day product (Figures 4 and 5).

Table 5 presents single dose data on areas under the curve (AUC) over given time periods (0-24 hrs and 0-72 hrs) for immediate release product (20 mg bid) and controlled release products (40 mg DR1, 40 mg XR1 and 20 mg/120 mg IR/DR2 combination), including %F, which is a measure of bioequivalence. One can appreciate that at least the DR1 and XR controlled release products provide AUC
data, which are comparable to those obtained with twice daily administered 20 mg immediate release trospium chloride.

Table 5. Single Dose Data for Various Trospium Chloride Formulations Single Dose Formulation AUC 0-24 p.hr/ml AUC 0-72 (pg.hr/ml) %F
20mg bid 23820 39831 100 TOmg DR1 23782 35589 89 40mg XR 24271 36098 91 20/120 mg IR/DR2 33244 52905 38 Example 7- Steady State Human Pharmacokinetic Studies A second human trial was conducted comparing four controlled release formulations described in Table 1 b with 20 mg IR tablet (Spasmo-Lyt , Madaus), which was given twice a day at 12 hour intervals.

Table 6 presents steady state data on AUC (over a 72 hour time period), Cmax, Cmin, and %F obtained from the administration of the various trospium chloride formulations discussed in the preceding paragraph.

Table 6. Steady State Data for Various Trospium Chloride Formulations A B C D E Conc Time 35mg DR1 40mg XR1- 50mg XR1-2 30mg XR1: 20mg IR
1 30mg DR2 BID
Tmax (Hr) 5.39 5.38 5.38 5.95 5.3 Cmax (pg/mL) 3164.9 2819.8 1908.7 2398.2 2978.9 AUClast (Hr*pg/mL) 55025.5 44972.1 42419.8 52060 67068.7 AUCINF_obs 64076.8 53637.4 62784.5 63931.2 74294.4 Hr*pg/mL
Relative BA 94% 67% 51% 52% 100%
(normalized) Example 8 - Enteral administration of a trospium chloride pharmaceutical composition A delayed release formulation, according to the method of the invention, is prepared using a trospium salt, such as a fluoride, chloride, bromide or iodide, using a delayed,release coating, which releases trospium at a pH of about 7Ø For example, an appropriate EUDRAGIT release controlling layer is selected so that the active ingredient is released at approximately neutral pH, which coincides substantially with the pH of the lower GI tract (e.g., lower intestine, colon, or both).
Other release controlling layers may also be selected with the objective of providing a pharmaceutical composition comprising trospium as at least one active ingredient, which releases trospium in sections of the GI tract previously thought not to play a role in the delivery/absorption of significant amounts of trospium. See, e.g., Schroder, S. et al., in International Journal of Clinical Pharmacology and Therapeutics, Vol. 42 - No. 10/2004 (543-549).

Claims (105)

CLAIMS:
1. An oral pharmaceutical composition comprising (1) between 25 mg and 80 mg of trospium chloride and (2) at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH
above 4.5, and an extended release (XR) component comprising from 1% to 40% of a release controlling polymer, wherein at least a portion of said trospium chloride is contained in said at least one component whereby at a once daily dosage the composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
2. The composition of claim 1, which at a once daily dosage at steady state provides minimum blood levels (C min) of trospium of between about 0.5 and about 1.5 ng/ml, and maximum blood levels (C max) of between about 2.0 and about 6.0 ng/ml.
3. The composition of claim 2, wherein the steady state blood levels are between about 1.0 ng/ml and about 5.0 ng/ml.
4. The composition of claim 1, which comprises an immediate release component containing no more than about 20 mg trospium chloride.
5. The composition of claim 1, which comprises an extended release component containing between about 25 and about 60 mg trospium chloride.
6. The composition of claim 1, which comprises a delayed release component containing between about 25 and about 80 mg trospium chloride.
7. The composition of claim 1, comprising a DR
trospium chloride component, in combination with an immediate release (IR) trospium chloride component.
8. The composition of claim 7, wherein the IR component contains no more than about 20 mg trospium chloride and wherein the total dose of trospium chloride is less than about 80 mg.
9. The composition of claim 1, which is a combination of an XR trospium chloride component and a DR trospium chloride component.
10. The composition of claim 9, wherein the XR component contains between about 10 to about 40 mg trospium chloride and the DR component contains between about 10 mg and about 40 mg trospium chloride.
11. The composition of claim 10, wherein the XR component contains about 30 mg trospium chloride and the DR component contains about 30 mg trospium chloride.
12. The composition of claim 1, comprising an XR
trospium chloride component, in combination with an immediate release (IR) trospium chloride component.
13. The composition of claim 12, wherein the IR component contains no more than about 20 mg and the XR component contains about 20 mg to about 60 mg trospium chloride.
14. The composition of claim 13, wherein the XR component contains about 20 mg to about 40 mg trospium chloride.
15. The composition of claim 1, comprising an XR
trospium chloride component and a DR trospium component, in combination with an immediate release (IR) trospium chloride component.
16. The composition of claim 15, wherein the IR component contains no more than about 20 mg trospium chloride, and the XR and DR components combined contain between about 10 mg and 60 mg trospium chloride and are in a ratio of XR:DR of 1:10 to 10:1.
17. The composition of claim 1, which is in the form of a granule, tablet, pellet, beadlet, powder, sachet, capsule, gel, dispersion, solution or suspension.
18. The composition of claim 1, which is in the form of pellets contained within a capsule.
19. The composition of claim 1, which is in the form of pellets that are compressed into a tablet.
20. The composition of any one of claims 7 to 16, which is a layered tablet, wherein each layer contains one of the components.
21. The composition of any one of claims 7 to 16, which is a layered pellet, wherein either or both said DR
and XR portion comprises the core of the pellet, and the IR
portion surrounds the core.
22. Use of the trospium composition as defined in claim 1, in the manufacture of a medicament which provides a daily dose for treating a urinary incontinence disease or condition in a mammal.
23. Use of a daily dose of the trospium composition as defined in claim 1, for treating a urinary incontinence disease or condition in a mammal, for at least a time sufficient to ameliorate the disease or condition.
24. The use of claim 22 or 23, wherein the mammal is a human.
25. The use of claim 22 or 23, wherein about 60 mg trospium chloride is present in the composition.
26. The use of claim 22 or 23, which composition comprises an immediate release component containing no more than about 20 mg trospium chloride.
27. The use of claim 22 or 23, which composition comprises an extended release component containing between about 25 and about 60 mg trospium chloride.
28. The use of claim 22 or 23, which composition comprises a delayed release component containing between about 25 and about 80 mg trospium chloride.
29. The use of claim 22 or 23, which composition comprises a DR trospium chloride component in combination with an immediate release (IR) trospium chloride component.
30. The use of claim 29, wherein the IR component contains no more than about 20 mg trospium chloride and wherein the total dose of trospium chloride is less than about 80 mg.
31. The use of claim 22 or 23, which composition is a combination of an XR trospium chloride component and a DR trospium chloride component.
32. The use of claim 31, wherein the XR component contains between about 10 mg to about 40 mg trospium chloride and the DR component contains between about 10 mg and about 40 mg trospium chloride.
33. The use of claim 32, wherein the XR component contains about 30 mg trospium chloride and the DR component contains about 30 mg trospium chloride.
34. The use of claim 22 or 23, which composition comprises an XR trospium chloride component in combination with an immediate release (IR) trospium chloride component.
35. The use of claim 34, wherein the IR component contains no more than about 20 mg and the XR component contains about 20 mg to about 60 mg trospium chloride.
36. The use of claim 35, wherein the XR component contains about 20 mg to about 40 mg trospium chloride.
37. The use of claim 22 or 23, which composition comprises an XR trospium chloride component and a DR
trospium component in combination with an immediate release (IR) trospium chloride component.
38. The use of claim 37, wherein the IR component contains no more than about 20 mg trospium chloride, and the XR and DR components combined contain between about 10 mg and 60 mg trospium chloride and are in a ratio of XR:DR
of 1:10 to 10:1.
39. The composition of claim 6, wherein about 35 mg of trospium are present, and the DR component comprises a polymeric substance that will dissolve at pH 5.5 - 6Ø
40. The composition of claim 13, wherein the XR component comprises a release controlling coating that gives a 3.5 hour in vivo release profile.
41. The composition of claim 13, wherein the XR component contains about 50 mg trospium chloride, and comprises a release controlling coating that gives a 4.5 hour in vivo release profile.
42. A pharmaceutical composition for a once-a-day administration of trospium comprising (1) a total amount of trospium chloride which is less than 80 mg and (2) at least one component selected from an extended release (XR) component comprised of extended release particulates comprising from 1% to 40% of a release controlling polymer;
and a delayed release (DR) component comprised of delayed release particulates comprising an enteric polymer that dissolves at pH above 4.5, wherein at least a portion of said trospium chloride is contained in the particulates of said at least one component whereby once-a-day administration of said pharmaceutical composition provides steady state blood levels of trospium that are substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg immediate release trospium chloride tablets.
43. The composition of claim 42, in which once-a-day administration of said controlled release pharmaceutical composition provides steady state blood levels of trospium falling in the range of about 0.5 ng/ml to about 6.0 ng/ml.
44. The composition of claim 43, in which once-a-day administration of said controlled release pharmaceutical composition provides steady state blood levels of trospium falling in the range of about 1.0 ng/ml to about 5.0 ng/ml.
45. The composition of claim 43, in which once-a-day administration of said controlled release pharmaceutical composition provides steady state blood C max levels of trospium falling in the range of about 2.5 ng/ml to about 4.5 ng/ml and C min levels of trospium falling in the range of about 0.5 ng/ml to about 1.5 ng/ml.
46. The composition of claim 43, in which once-a-day administration of said controlled release pharmaceutical composition provides steady state areas under the curve (AUCs) falling in the range of about 30 to about 60 ng/ml*hr.
47. The composition of claim 46, in which once-a-day administration of said controlled release pharmaceutical composition provides steady state areas under the curve falling in the range of about 35 to about 45 ng/ml*hr.
48. The composition of claim 42, in which once-a-day administration of said controlled release pharmaceutical composition provides single dose %F values falling in the range of about 80 to about 120.
49. The composition of claim 48, in which once-a-day administration of said controlled release pharmaceutical composition provides single dose %F values falling in the range of about 90 to about 110.
50. The composition of claim 42, additionally comprising an immediate release (IR) component comprised of an immediate release particulate.
51. The composition of claim 42, wherein the whole amount of trospium chloride is in the form of the DR component.
52. The composition of claim 42, wherein the whole amount of trospium chloride is in the form of the XR
component comprised of an extended release particulate that allows release of about 80% trospium chloride at 3.5 hours post ingestion.
53. The composition of claim 42, wherein the whole amount of trospium chloride is in the form of the XR
component comprised of an extended release particulate that allows release of about 80% trospium chloride at 4.5 hours post ingestion.
54. The composition of claim 42, comprising an XR component comprised of an extended release particulate that allows release of about 80% trospium chloride at 3.5 hours post ingestion, and the DR component.
55. The composition of claim 54, wherein the DR component comprises the delayed release particulates that release trospium chloride when they reach the area of the GI tract where the pH is about 7.
56. Use of a once-a-day formulation comprising a therapeutically effective amount of trospium chloride in the form of at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH above 4.5, and an extended release (XR) component comprising from 1% to 40% of a release controlling polymer, which provides steady state blood levels of trospium substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg immediate release trospium chloride tablets and with a lessening of side effects, in the manufacture of a medicament for treating a mammal suffering from overactive bladder that would benefit from a once daily administration of a therapeutically effective amount of trospium chloride.
57. Use of a once-a-day formulation comprising a therapeutically effective amount of trospium chloride in the form of at least one component selected from a delayed release (DR) component comprising an enteric polymer that dissolves at pH above 4.5, and an extended release (XR) component comprising from 1% to 40% of a release controlling polymer, which provides steady state blood levels of trospium substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg immediate release trospium chloride tablets and with a lessening of side effects, for treating a mammal suffering from overactive bladder that would benefit from a once daily administration of a therapeutically effective amount of trospium chloride.
58. Use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride at a pH of about 7.0 for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
59. Use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride in the lower intestine for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
60. Use, for treating overactive bladder, of a pharmaceutical composition comprising a therapeutically effective amount of trospium chloride, in a delayed release formulation of trospium chloride, which releases trospium chloride in the colon for enteral administration, and a carrier, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
61. A pharmaceutical composition comprising from 25 to 80 mg of trospium chloride in which at least a portion of said trospium chloride is contained in a delayed release formulation, which comprises an enteric polymer that dissolves at a pH of about 7.0, releasing trospium chloride, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
62. A pharmaceutical composition comprising from 25 to 80 mg trospium chloride in which at least a portion of said trospium chloride is contained in a delayed release formulation, which comprises an enteric polymer that dissolves in the lower intestine, colon, or both, releasing trospium chloride, wherein the pharmaceutical composition provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml.
63. A process of preparation of a once daily dosage unit of trospium chloride, which will give steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, comprising the steps of: 1. preparing at least one component selected from an extended release (XR) component and a delayed release (DR) component; 2. optionally, combining the result of step 1 with at least one component selected from an XR component, a DR component and an immediate release (IR) component; and 3.
encapsulating the result of step 1 or step 2 into a capsule, wherein the total dose of trospium chloride is less than about 80 mg.
64. The process of claim 63, wherein the XR component is prepared by obtaining a drug layered core or drug loaded granule, and applying to said core or granule a coat of a release controlling polymer.
65. The process of claim 63, wherein the DR component is prepared by obtaining a drug layered core or drug loaded granule, and applying a coat on said core or granule of an enteric material.
66. The process of claim 63, wherein the IR component is prepared by a process comprising the steps of: 1. spraying onto an inert core a drug solution containing trospium chloride and, optionally, a binder and anti-tacking agents, to thereby form a layer of drug on the core; 2. optionally, applying a protective coat of a polymeric composition to the drug-layered core of step 1.
67. The process of claim 63, comprising combining the XR component containing about 20 mg to about 60 mg trospium chloride with the IR component containing no more than about 20 mg trospium chloride into a capsule.
68. The process of claim 67, wherein the XR pellets contain between about 20 mg and about 40 mg trospium chloride.
69. The process of claim 63, comprising combining the DR component with the IR component containing no more than about 20 mg trospium chloride into a capsule, wherein the total dose of the combination is less than about 80 mg.
70. The process of claim 63, comprising combining the XR component containing between about 10 and 40 mg trospium chloride with the DR component containing between about 10 and 40 mg into a capsule.
71. The process of claim 70, wherein each of the XR
and DR components contains about 30 mg trospium chloride.
72. The process of claim 63, wherein the blood levels at steady state are between about 1.0 ng/ml and about 5.0 ng/ml.
73. A once daily dosage form of trospium chloride comprising the composition as defined in any one of claims 1 to 16, 39 to 55, 61 and 62.
74. The dosage form of claim 73, selected from a granule, a tablet, a pellet, a beadlet, a powder, a sachet, a capsule, a gel, a dispersion, a solution, and a suspension.
75. An oral pharmaceutical composition of trospium comprising 25 to 80 mg of trospium chloride and a carrier, wherein at least part of the trospium chloride is contained in an extended release (XR) component or in a delayed release (DR) component, and wherein said composition at a once daily dosage provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.
76. An oral pharmaceutical composition of trospium comprising at least one component selected from a delayed release (DR) component and an extended release (XR) component, and a carrier, wherein said composition contains between 25 mg and 80 mg of trospium chloride and at a once daily dosage provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, while minimizing the occurrence of adverse side effects as compared to an immediate release formulation containing 40 mg of trospium chloride.
77. The composition of claim 75 or 76, wherein use of the composition results in steady state blood levels of trospium substantially equivalent to steady state blood levels of trospium achieved with twice daily administration of 20 mg of immediate release trospium chloride tablets, and in minimizing the occurrence of adverse side effects.
78. The composition of claim 75 or 76, wherein said XR
component comprises at least one release controlling polymer selected from copolymers of acrylic or methacrylic acid esters, ethylcellulose aqueous dispersions, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, polyethylene glycols, and any combinations thereof.
79. The composition of claim 75 or 76, wherein said DR
component comprises at least one enteric polymer selected from cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, polyvinyl acetate phthalate, hydroxypropyl methylcellulose acetate succinate, cellulose acetate trimellitate, hydroxypropyl methylcellulose succinate, cellulose acetate succinate, cellulose acetate hexahydrophthalate, cellulose propionate phthalate, copolymer of methylmethacrylic acid and methyl methacrylate, copolymer of methyl acrylate, methylmethacrylate and methacrylic acid, copolymer of methylvinyl ether and maleic anhydride, ethyl methyacrylate-methylmethacrylate-chlorotrimethylammonium ethyl acrylate copolymer, zein, shellac, copal collophorium, carboxymethyl ethylcellulose, co-polymerized methacrylic acid/methacrylic acid methyl esters, and any combinations thereof.
80. The composition of claim 75 or 76, wherein said composition additionally comprises an immediate release (IR) trospium component.
81. The composition of claim 80, wherein said IR
component contains not more than about 20 mg of trospium chloride.
82. The composition of claim 81, wherein said composition is a combination of said IR trospium chloride component and said DR trospium chloride component.
83. The composition of claim 75 or 76, wherein said composition is a combination of said XR trospium chloride component and said DR trospium chloride component.
84. The composition of claim 83, wherein said DR
component comprises trospium chloride and an enteric polymer, and said XR component comprises trospium chloride and a release controlling polymer.
85. The composition of claim 84, wherein each of said XR component and said DR component is in the form of pellets.
86. The composition of claim 85, wherein said DR
component is composed of DR pellets consisting essentially of 1. trospium chloride, 2. a sugar core, 3. hydroxypropyl methylcellulose, 4. a coating of the enteric polymer that delays release of the trospium chloride from the pellet for a period of time after administration, 5. triethyl citrate, 6. a protective overcoating, and 7. talc; and said XR component is composed of XR pellets consisting essentially of 1. trospium chloride, 2. a sugar core, 3.
hydroxypropyl methylcellulose, 4. a surface coating that controls a release profile of the trospium chloride from the pellet after administration, 5. a protective overcoating, and 6. talc.
87. The composition of claim 86, wherein said XR
component contains about 30 mg trospium chloride, and said DR component contains about 30 mg trospium chloride.
88. The composition of claim 81, wherein said composition is a combination of said IR trospium chloride component and said XR trospium chloride component.
89. The composition of claim 81, wherein said composition is a combination of said IR trospium chloride component, said XR trospium chloride component, and said DR
trospium component.
90. The composition of claim 75 or 76, wherein said DR
component releases trospium chloride at a pH of about 7Ø
91. The composition of claim 75 or 76, wherein said DR
component releases trospium chloride in a lower intestine.
92. The composition of claim 75 or 76, wherein said DR
component releases trospium chloride in a colon.
93. The composition of claim 77, wherein said adverse effects are selected from dry mouth, headache, constipation, dyspepsia, abdominal pain, and any combinations thereof.
94. The composition of claim 75 or 76, wherein said bladder dysfunction is selected from urinary frequency, urgency, nocturia, urge-incontinence due to detrusor instability, urge syndrome, detrusor hyperreflexia, and any combinations thereof.
95. The composition of claim 75 or 76, wherein said composition is in an oral dosage form selected from a granule, tablet, pellet, beadlet, powder, sachet, capsule, gel, dispersion, solution, and suspension.
96. The composition of claim 95, wherein said tablet is a rapidly dispersible tablet.
97. The composition of claim 75 or 76, for use without the intake of high-fat food.
98. The composition of claim 95, wherein said tablet, pellet or beadlet is a layered tablet, pellet or beadlet comprising at least two trospium-containing layers, wherein each layer comprises at least one component selected from said XR component, said DR component or an immediate release (IR) component.
99. An oral pharmaceutical composition of trospium, containing a once daily dose of trospium chloride and comprising a combination of a delayed release (DR) component comprising an enteric polymer that dissolves at pH above 4.5, and an extended release (XR) component comprising from 1% to 40% of a release controlling polymer, wherein said DR
component is composed of enteric coated pellets consisting essentially of: 1. trospium chloride, 2. a sugar core, 3. hydroxypropyl methylcellulose, 4. a coating of the enteric polymer that delays release of the trospium chloride from the pellet for a period of time after administration, 5. triethyl citrate, 6. a protective overcoating, and 7. talc; and said XR component is composed of extended release pellets consisting essentially of: 1.
trospium chloride, 2. a sugar core, 3. hydroxypropyl methylcellulose, 4. a surface coating that controls a release profile of the trospium chloride from the pellet after administration, 5. a protective overcoating, and 6.
talc; and the once daily dose provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.
100. An oral pharmaceutical composition of trospium, containing a once daily dose of trospium chloride and comprising a mixture of enteric coated pellets and extended release pellets in a ratio of 1:1, the enteric coated pellets consisting essentially of trospium chloride, sugar spheres, hydroxypropyl methylcellulose, methacrylic acid copolymer, triethyl citrate, and talc and wherein the extended release pellets consist essentially of trospium chloride, sugar spheres, hydroxypropyl methylcellulose, ethyl cellulose, OPADRY WHITE.TM. and talc; and the once daily dose provides steady state blood levels of trospium of a minimum of about 0.5 ng/ml and a maximum of about 6.0 ng/ml, for use in treating a bladder dysfunction in a mammal.
101. The composition of claim 99 or 100, wherein 60 mg of trospium chloride is in the pharmaceutical composition.
102. The composition of claim 101, wherein the 60 mg of trospium chloride is equally divided between the enteric coated pellets and extended release pellets.
103. The composition of any one of claims 99 to 102, wherein the bladder dysfunction is overactive bladder (OAB) with symptoms of urge urinary incontinence, urgency and urinary frequency and the composition is in a capsule.
104. The composition of any one of claims 1 to 21, 42 to 55, 61 and 62, further comprising a carrier.
105. The use of claim 56 or 57, wherein the formulation further comprises a carrier.
CA002537103A 2003-11-04 2004-11-04 Once daily dosage forms of trospium Expired - Fee Related CA2537103C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US51719803P 2003-11-04 2003-11-04
US60/517,198 2003-11-04
US52396803P 2003-11-21 2003-11-21
US60/523,968 2003-11-21
PCT/US2004/036394 WO2005046684A1 (en) 2003-11-04 2004-11-04 Once daily dosage forms of trospium

Publications (2)

Publication Number Publication Date
CA2537103A1 CA2537103A1 (en) 2005-05-26
CA2537103C true CA2537103C (en) 2010-01-19

Family

ID=34594857

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002537103A Expired - Fee Related CA2537103C (en) 2003-11-04 2004-11-04 Once daily dosage forms of trospium

Country Status (12)

Country Link
US (12) US7410978B2 (en)
EP (2) EP1680110B1 (en)
JP (2) JP5610663B2 (en)
AT (1) ATE493981T1 (en)
AU (1) AU2004289223B2 (en)
CA (1) CA2537103C (en)
DE (1) DE602004030931D1 (en)
DK (1) DK2210605T3 (en)
ES (1) ES2359375T3 (en)
HK (1) HK1088547A1 (en)
PT (1) PT2210605T (en)
WO (1) WO2005046684A1 (en)

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8709476B2 (en) 2003-11-04 2014-04-29 Supernus Pharmaceuticals, Inc. Compositions of quaternary ammonium compounds containing bioavailability enhancers
DE602004030931D1 (en) * 2003-11-04 2011-02-17 Supernus Pharmaceuticals Inc
US20070134315A1 (en) * 2005-12-08 2007-06-14 Viera Michael L Orally administrable extended release pellet and tablet formulations of a highly water soluble compound
US20070134322A1 (en) * 2005-12-14 2007-06-14 Forest Laboratories, Inc. Modified and pulsatile release pharmaceutical formulations of escitalopram
DK1880718T3 (en) * 2006-07-10 2011-11-21 Pfleger R Chem Fab Pharmaceutical composition for oral administration with controlled release of active substance in the small intestine and method of preparation thereof
WO2008027557A2 (en) * 2006-08-31 2008-03-06 Spherics, Inc. Topiramate compositions and methods of enhancing its bioavailability
MX2009001711A (en) 2006-11-17 2009-05-08 Supernus Pharmaceuticals Inc Sustained-release formulations of topiramate.
EP2363113B1 (en) * 2006-12-04 2017-08-02 Supernus Pharmaceuticals, Inc. Enhanced immediate release formulations of topiramate
WO2009130712A2 (en) * 2008-04-22 2009-10-29 Lupin Limited Controlled release pharmaceutical compositions of trospium
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US20110020423A1 (en) 2009-07-22 2011-01-27 Puretech Ventures Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
GB2479213B (en) 2010-04-01 2013-07-10 Theravida Inc Pharmaceutical formulations for the treatment of overactive bladder
WO2011159824A1 (en) * 2010-06-16 2011-12-22 Allergan, Inc. Composition and method for treating overactive bladder
RU2016125229A (en) * 2010-07-09 2018-12-04 БиЭйчВи ФАРМА, ИНК. COMBINED DELIVERY SYSTEM WITH IMMEDIATE / SLOW-DELIVERY FOR MEDICINES WITH A SHORT HALF-HOUR, INCLUDING REMOGLYPHLOSIN
RU2671575C2 (en) 2011-05-10 2018-11-02 Теравида, Инк. Appliance of solifenacin and saliva flow stimulators for the treatment of overactive bladder
SG194807A1 (en) 2011-05-10 2013-12-30 Theravida Inc Combinations of trospium and salivary stimulants for the treatment of overactive bladder
NZ629468A (en) * 2012-03-02 2017-08-25 Rhodes Pharmaceuticals Lp Tamper resistant immediate release formulations
CN103316026B (en) 2012-03-23 2016-05-11 中国人民解放军军事医学科学院毒物药物研究所 Contain joint product of Phentermine and Topiramate and preparation method thereof
WO2013139292A1 (en) * 2012-03-23 2013-09-26 中国人民解放军军事医学科学院毒物药物研究所 Joint product containing synephrine and topiramate
MX358211B (en) * 2012-07-23 2018-08-10 Landsteiner Scient S A De C V New differential-release pharmaceutical composition containing three active principles.
US8962020B2 (en) * 2012-07-26 2015-02-24 Glycadia Inc. Long-acting and controlled release formulations of 2-[(3-chlorophenyl) amino] phenylacetic acid
US10751287B2 (en) * 2013-03-15 2020-08-25 Purdue Pharma L.P. Tamper resistant pharmaceutical formulations
CA2936740C (en) 2014-10-31 2017-10-10 Purdue Pharma Methods and compositions particularly for treatment of attention deficit disorder
CN104739808B (en) * 2015-02-13 2017-09-22 舒泰神(北京)生物制药股份有限公司 Double release capsules of a kind of trospium chloride and preparation method thereof
BR112018014661A2 (en) 2016-01-20 2018-12-11 Theravida Inc composition and composition for use
BR112021005802B1 (en) 2018-09-28 2022-02-15 Karuna Therapeutics, Inc ORAL PHARMACEUTICAL COMPOSITION
US10722473B2 (en) 2018-11-19 2020-07-28 Purdue Pharma L.P. Methods and compositions particularly for treatment of attention deficit disorder

Family Cites Families (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2899357A (en) * 1959-08-11 Oral pharmaceutical composition for
US518777A (en) * 1894-04-24 Bicycle
US652837A (en) * 1899-11-17 1900-07-03 Daniel Mcnamara Wheel for vehicles.
US2095282A (en) 1935-03-15 1937-10-12 Standard Oil Co Capillary viscometer
US2956926A (en) 1958-09-23 1960-10-18 American Cyanamid Co Coated citric acid particles
US3065143A (en) 1960-04-19 1962-11-20 Richardson Merrell Inc Sustained release tablet
DE1194422B (en) 1963-03-05 1965-06-10 Robert Pfleger Chem Fab Dr Process for the production of azonia spironortropane derivatives
US4259314A (en) * 1979-12-10 1981-03-31 Hans Lowey Method and composition for the preparation of controlled long-acting pharmaceuticals
DE3237575A1 (en) * 1982-10-09 1984-04-12 Dr. Karl Thomae Gmbh, 7950 Biberach NEW ORAL MOPIDAMOL SHAPES
US4556552A (en) 1983-09-19 1985-12-03 Colorcon, Inc. Enteric film-coating compositions
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
ATE43789T1 (en) 1985-02-23 1989-06-15 Asta Pharma Ag COMBINATION OF FLUPIRTIN AND ANTICHOLINERGIC SPASMOLYTICS.
GB8613689D0 (en) * 1986-06-05 1986-07-09 Euro Celtique Sa Pharmaceutical composition
US4713243A (en) 1986-06-16 1987-12-15 Johnson & Johnson Products, Inc. Bioadhesive extruded film for intra-oral drug delivery and process
JP2528652B2 (en) 1987-03-30 1996-08-28 エスエス製薬株式会社 Long-acting cefradine formulation
US4786503A (en) 1987-04-06 1988-11-22 Alza Corporation Dosage form comprising parallel lamine
GB8717446D0 (en) * 1987-07-23 1987-08-26 Merck Sharp & Dohme Chemical compounds
US5656286A (en) * 1988-03-04 1997-08-12 Noven Pharmaceuticals, Inc. Solubility parameter based drug delivery system and method for altering drug saturation concentration
US5279660A (en) 1988-05-24 1994-01-18 Berol Nobel Stenungsund Ab Use of viscosity-adjusting agent to counteract viscosity decrease upon temperature increase of a water-based system
US5520932A (en) 1988-06-24 1996-05-28 The Upjohn Company Fine-milled colestipol hydrochloride
US4970081A (en) * 1989-01-03 1990-11-13 Sterling Drug Inc. Controlled-release, low-dose aspirin formulation and method of treating vascular occlusive disease therewith
US5330766A (en) * 1989-01-06 1994-07-19 F. H. Faulding & Co. Limited Sustained release pharmaceutical composition
US5372823A (en) 1989-03-16 1994-12-13 Bristol-Myers Squibb Company Direct compression cholestyramine tablet and solvent-free coating thereof
US5179660A (en) * 1989-05-15 1993-01-12 International Business Machines Incorporated System for reducing communications overhead in distributed database transactions by serializing in order related requests into single transmission message and receiving transmission response
US5158777A (en) * 1990-02-16 1992-10-27 E. R. Squibb & Sons, Inc. Captopril formulation providing increased duration of activity
WO1991016449A1 (en) * 1990-04-16 1991-10-31 The Trustees Of The University Of Pennsylvania Saccharide compositions, methods and apparatus for their synthesis
IT1246382B (en) 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
US5686094A (en) * 1991-04-01 1997-11-11 Theratech, Inc. Controlled release formulations for the treatment of xerostomia
US5095282A (en) * 1990-08-23 1992-03-10 Nvision, Inc. Differential amplifier apparatus
US5203203A (en) 1990-10-10 1993-04-20 Bryan William L Viscometer for in situ monitoring
US5326570A (en) * 1991-07-23 1994-07-05 Pharmavene, Inc. Advanced drug delivery system and method of treating psychiatric, neurological and other disorders with carbamazepine
DE69233093T2 (en) * 1991-09-26 2003-12-18 Us Gov Health & Human Serv SUBSTITUTED PHENSERINE AND PHENYLCARBAMATE OF (-) - ESEROLIN, (-) - N1-NORESEROLIN AND (-) - N1-BENZYLNORESOROLIN AS SPECIFIC ACETYLCHOLINESTERASE INHIBITORS
US5681585A (en) 1991-12-24 1997-10-28 Euro-Celtique, S.A. Stabilized controlled release substrate having a coating derived from an aqueous dispersion of hydrophobic polymer
US5273760A (en) 1991-12-24 1993-12-28 Euroceltigue, S.A. Stabilized controlled release substrate having a coating derived from an aqueous dispersion of hydrophobic polymer
WO1993015724A1 (en) * 1992-02-18 1993-08-19 Nippon Shinyaku Co., Ltd. Fast soluble tablet
US5405648A (en) 1993-05-10 1995-04-11 Hermann; Paul F. Coating particulate material with a polymer film
WO1995005809A1 (en) * 1993-08-20 1995-03-02 Nippon Shinyaku Co., Ltd. Gastric remaining preparation, swollen molding, and production process
EP0716597A1 (en) * 1993-08-30 1996-06-19 Warner-Lambert Company Tablet coating based on a melt-spun mixture of a saccharide and apolymer
US5399359A (en) * 1994-03-04 1995-03-21 Edward Mendell Co., Inc. Controlled release oxybutynin formulations
US6077533A (en) * 1994-05-25 2000-06-20 Purdue Pharma L.P. Powder-layered oral dosage forms
US5520832A (en) * 1994-10-28 1996-05-28 Exxon Research And Engineering Company Tractor hydraulic fluid with wide temperature range (Law180)
JPH08157392A (en) 1994-12-01 1996-06-18 Kanegafuchi Chem Ind Co Ltd Release control type preparation
US5582838A (en) 1994-12-22 1996-12-10 Merck & Co., Inc. Controlled release drug suspension delivery device
FI98343C (en) 1995-03-21 1997-06-10 Orion Yhtymae Oy Oral formulation
US6124355A (en) * 1995-05-22 2000-09-26 Guittard; George V. Oxybutynin therapy
US5811388A (en) * 1995-06-07 1998-09-22 Cibus Pharmaceutical, Inc. Delivery of drugs to the lower GI tract
US5879712A (en) 1995-06-07 1999-03-09 Sri International Method for producing drug-loaded microparticles and an ICAM-1 dosage form so produced
US5779661A (en) * 1995-12-11 1998-07-14 Physion, S.R.L. Method of treating dysfunctional bladder syndromes by electromotive drug administration
JPH09188617A (en) * 1996-01-08 1997-07-22 Pola Chem Ind Inc Medicinal composition of sustained release
JP3929522B2 (en) * 1996-03-14 2007-06-13 塩野義製薬株式会社 Sustained release formulation of poorly water-soluble drugs
US6156340A (en) 1996-03-29 2000-12-05 Duquesne University Of The Holy Ghost Orally administrable time release drug containing products
US20030064108A1 (en) 1996-04-23 2003-04-03 Stefan Lukas Taste masked pharmaceutical compositions
JPH09315969A (en) 1996-05-24 1997-12-09 Taiyo Yakuhin Kogyo Kk Ibudilast-containing sustained release medicinal composition and its production
US6652837B1 (en) 1996-05-24 2003-11-25 Massachusetts Institute Of Technology Preparation of novel particles for inhalation
EP0920313A1 (en) * 1996-07-01 1999-06-09 Sepracor, Inc. Methods and compositions for treating urinary incontinence using enantiomerically enriched (s,s)-glycopyrrolate
US5959196A (en) * 1996-09-10 1999-09-28 Norcross Corporation In-line viscometer
US20010003588A1 (en) 1996-09-12 2001-06-14 Smithkline Beecham Corporation Controlled release dosage form of [R-(Z)]-alpha-(methoxyimino)-alpha-(1-azabicyclo[2.2.2.]oct-3-yl)acetonitrile monohydrochloride
US5972389A (en) 1996-09-19 1999-10-26 Depomed, Inc. Gastric-retentive, oral drug dosage forms for the controlled-release of sparingly soluble drugs and insoluble matter
ES2125696T3 (en) * 1996-11-27 1999-03-01 Pfleger R Chem Fab USE OF TROPSY CHLORIDE FOR THE PRODUCTION OF A PHARMACEUTICAL PREPARATION FOR THE TREATMENT OF VESIC DYSFUNCTIONS.
US5973389A (en) * 1997-04-22 1999-10-26 International Business Machines Corporation Semiconductor chip carrier assembly
US6141624A (en) * 1997-05-13 2000-10-31 International Remote Imaging Systems Fluid sample for analysis controlled by total fluid volume and by total particle counts
DE69818607T2 (en) 1997-05-30 2004-07-29 Osmotica Corp. MULTILAYER OSMOSIS DEVICE
US6141625A (en) * 1997-06-09 2000-10-31 Dickey-John Corporation Viscometer module with crystal resonator-type sensor
DE19724696A1 (en) 1997-06-12 1998-12-24 Hexal Ag Pharmaceutical preparation with three types of pellets
US6197331B1 (en) 1997-07-24 2001-03-06 Perio Products Ltd. Pharmaceutical oral patch for controlled release of pharmaceutical agents in the oral cavity
US6337091B1 (en) 1997-10-27 2002-01-08 Temple University - Of The Commonwealth System Of Higher Education Matrix for controlled delivery of highly soluble pharmaceutical agents
CO5070568A1 (en) 1998-05-22 2001-08-28 Eurand Internatrional Spa LAYER AND APPLIANCE APPLICATION PROCESS FOR EFFECT
WO2000006128A1 (en) 1998-07-28 2000-02-10 Tanabe Seiyaku Co., Ltd. Preparation capable of releasing drug at target site in intestine
JP2000103732A (en) * 1998-07-28 2000-04-11 Tanabe Seiyaku Co Ltd Intestinal right place release preparation
SE9803871D0 (en) 1998-11-11 1998-11-11 Pharmacia & Upjohn Ab Therapeutic method and formulation
EA002720B1 (en) 1998-08-27 2002-08-29 Фармациа Энд Апджон Аб Therapeutic formulation for administering tolterodine with controlled release and method for treating urinary bladder using thereof
US20010055613A1 (en) * 1998-10-21 2001-12-27 Beth A. Burnside Oral pulsed dose drug delivery system
KR20010082251A (en) 1998-10-26 2001-08-29 찌바따 이찌로, 다나까 도시오 Sustained-release particles
US6270805B1 (en) 1998-11-06 2001-08-07 Andrx Pharmaceuticals, Inc. Two pellet controlled release formulation for water soluble drugs which contains an alkaline metal stearate
US6419960B1 (en) * 1998-12-17 2002-07-16 Euro-Celtique S.A. Controlled release formulations having rapid onset and rapid decline of effective plasma drug concentrations
MXPA01007463A (en) 1999-01-29 2002-06-04 Losan Pharma Gmbh Pharmaceutical compositions.
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6576619B2 (en) 1999-05-24 2003-06-10 Cv Therapeutics, Inc. Orally active A1 adenosine receptor agonists
US6395300B1 (en) 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6632451B2 (en) * 1999-06-04 2003-10-14 Dexcel Pharma Technologies Ltd. Delayed total release two pulse gastrointestinal drug delivery system
ES2213949T3 (en) * 1999-07-02 2004-09-01 Cognis Iberia, S.L. MICROCAPSULES I.
CA2383212C (en) 1999-09-02 2008-11-25 Nostrum Pharmaceuticals, Inc. Controlled release oral dosage suitable for oral administration
BR0013720A (en) * 1999-09-02 2002-07-23 Nostrum Pharmaceuticals Inc Controlled release pellet formulation
US20020004065A1 (en) 2000-01-20 2002-01-10 David Kanios Compositions and methods to effect the release profile in the transdermal administration of active agents
EP1252887A4 (en) 2000-01-27 2009-07-29 Mitsubishi Tanabe Pharma Corp Sustained-release preparation and process for producing the same
MXPA02008183A (en) 2000-02-24 2002-11-29 Upjohn Co New drug combinations.
WO2001078725A2 (en) 2000-04-13 2001-10-25 Synthon B.V. Modified release formulations containing a hypnotic agent
US6350471B1 (en) 2000-05-31 2002-02-26 Pharma Pass Llc Tablet comprising a delayed release coating
CA2420597C (en) 2000-08-31 2011-05-17 Rtp Pharma Inc. Milled particles
US6620439B1 (en) * 2000-10-03 2003-09-16 Atul M. Mehta Chrono delivery formulations and method of use thereof
PL201766B1 (en) * 2001-01-31 2009-05-29 Roehm Gmbh Multi-particulate form of medicament, comprising at least two differently coated forms of pellet
ES2284646T3 (en) 2001-02-22 2007-11-16 Jagotec Ag STATIN-FIBRATE COMBINATIONS WITH SECONDARY EFFECTS IN REDUCED FAST-FASTING.
AUPR752201A0 (en) * 2001-09-07 2001-09-27 Schefenacker Vision Systems Australia Pty Ltd Powered telescoping trailer tow mirror
BR0206207A (en) 2001-10-26 2003-12-23 Pharmacia & Up John Company Quaternary ammonium compounds and their use as antimuscarinic agents
US20030185882A1 (en) * 2001-11-06 2003-10-02 Vergez Juan A. Pharmaceutical compositions containing oxybutynin
US6682759B2 (en) * 2002-02-01 2004-01-27 Depomed, Inc. Manufacture of oral dosage forms delivering both immediate-release and sustained-release drugs
US6958161B2 (en) 2002-04-12 2005-10-25 F H Faulding & Co Limited Modified release coated drug preparation
JP2006511449A (en) * 2002-04-29 2006-04-06 シャイア ラボラトリーズ,インコーポレイテッド Pharmaceutical formulations with improved bioavailability
US7588825B2 (en) 2002-10-23 2009-09-15 Boston Scientific Scimed, Inc. Embolic compositions
US6974820B2 (en) 2002-11-06 2005-12-13 Bridge Pharma, Inc. Methods for treating urinary incontinence and other disorders using trospium
WO2004062577A2 (en) * 2003-01-03 2004-07-29 Shire Laboratories Inc. Two or more enteric materials to regulate drug release
US20040138634A1 (en) 2003-01-10 2004-07-15 Litvay John D. Absorbent article having improved softness
BRPI0406861A (en) 2003-01-22 2006-01-03 Pfizer Health Ab Use of antimuscarinic agent
JP2006523703A (en) 2003-04-14 2006-10-19 シャイア ラボラトリーズ,インコーポレイテッド Pharmaceutical compositions that release active agents from buccal or sublingual locations to overcome the absorption window problem
US20050009862A1 (en) 2003-04-25 2005-01-13 Sabounjian Luann Method for promoting uninterrupted sleep by administration of trospium chloride
US6772801B1 (en) 2003-05-14 2004-08-10 Shire Laboratories, Inc. Fluidization of particles for encapsulation in oral dosage pharmaceutical products
DE602004030931D1 (en) * 2003-11-04 2011-02-17 Supernus Pharmaceuticals Inc
WO2006027783A2 (en) * 2004-09-08 2006-03-16 Ramot At Tel Aviv University Ltd. Mri imaging and contrast method
JP2006146117A (en) * 2004-10-20 2006-06-08 Brother Ind Ltd Image forming apparatus

Also Published As

Publication number Publication date
DE602004030931D1 (en) 2011-02-17
JP5610663B2 (en) 2014-10-22
US7759359B2 (en) 2010-07-20
US20100221353A1 (en) 2010-09-02
US20050191351A1 (en) 2005-09-01
US20100222375A1 (en) 2010-09-02
JP2007510656A (en) 2007-04-26
EP2210605A1 (en) 2010-07-28
US7410978B2 (en) 2008-08-12
AU2004289223A1 (en) 2005-05-26
WO2005046684A1 (en) 2005-05-26
EP1680110A4 (en) 2008-02-20
US7763635B2 (en) 2010-07-27
US20080207664A1 (en) 2008-08-28
AU2004289223B2 (en) 2009-11-05
CA2537103A1 (en) 2005-05-26
US7781448B2 (en) 2010-08-24
EP2210605B1 (en) 2017-03-01
EP1680110B1 (en) 2011-01-05
US20100221355A1 (en) 2010-09-02
US7781449B2 (en) 2010-08-24
US20100221352A1 (en) 2010-09-02
JP5783489B2 (en) 2015-09-24
US20100221356A1 (en) 2010-09-02
PT2210605T (en) 2017-04-24
US20080207662A1 (en) 2008-08-28
US20130089607A1 (en) 2013-04-11
DK2210605T3 (en) 2017-05-22
HK1088547A1 (en) 2006-11-10
US20080207661A1 (en) 2008-08-28
US20080207663A1 (en) 2008-08-28
ATE493981T1 (en) 2011-01-15
ES2359375T3 (en) 2011-05-20
US20100221354A1 (en) 2010-09-02
EP1680110A1 (en) 2006-07-19
JP2012144577A (en) 2012-08-02

Similar Documents

Publication Publication Date Title
US7781449B2 (en) Trospium chloride treatment method
EP1123087B1 (en) Oral pulsed dose drug delivery system
US9040086B2 (en) Timed, sustained release systems for propranolol
AU2009223019C1 (en) Drug delivery systems comprising weakly basic drugs and organic acids
PL200816B1 (en) Pharmaceutical dosage forms for controlled release producing at least a timed pulse
US20200261372A1 (en) Extended release compositions comprising trihexyphenidyl
CA2303973C (en) Prolonged release formulation of carbamazepine
CA2380333A1 (en) Pharmaceutical formulations
KR20070044911A (en) Controlled-release formulation containing tamsulosin hydrochloride
ZA200608190B (en) Controleld release dosage for GABA receptor antagonist
CA3064891A1 (en) Delayed sustained release pharmaceutical compositions
MXPA06004017A (en) Once daily dosage forms of trospium
JP2003507416A (en) Pharmaceutical preparations

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20171106