WO2017039833A1 - Delayed-release formulations, methods of making and use thereof - Google Patents

Delayed-release formulations, methods of making and use thereof Download PDF

Info

Publication number
WO2017039833A1
WO2017039833A1 PCT/US2016/041354 US2016041354W WO2017039833A1 WO 2017039833 A1 WO2017039833 A1 WO 2017039833A1 US 2016041354 W US2016041354 W US 2016041354W WO 2017039833 A1 WO2017039833 A1 WO 2017039833A1
Authority
WO
WIPO (PCT)
Prior art keywords
component
coating
release
delayed
pharmaceutical composition
Prior art date
Application number
PCT/US2016/041354
Other languages
French (fr)
Inventor
David A. Dill
Original Assignee
Wellesley Pharmaceuticals, Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US14/842,539 external-priority patent/US10278925B2/en
Application filed by Wellesley Pharmaceuticals, Llc filed Critical Wellesley Pharmaceuticals, Llc
Priority to EP16842483.6A priority Critical patent/EP3344241A4/en
Priority to AU2016317093A priority patent/AU2016317093A1/en
Priority to JP2018530487A priority patent/JP2018526442A/en
Priority to SG11201805812QA priority patent/SG11201805812QA/en
Priority to RU2018111401A priority patent/RU2018111401A/en
Priority to KR1020187009153A priority patent/KR20180054656A/en
Priority to MX2018002449A priority patent/MX2018002449A/en
Priority to CN201680050660.8A priority patent/CN108348487A/en
Publication of WO2017039833A1 publication Critical patent/WO2017039833A1/en
Priority to HK19101249.6A priority patent/HK1259196A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/612Salicylic acid; Derivatives thereof having the hydroxy group in position 2 esterified, e.g. salicylsulfuric acid
    • A61K31/616Salicylic acid; Derivatives thereof having the hydroxy group in position 2 esterified, e.g. salicylsulfuric acid by carboxylic acids, e.g. acetylsalicylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/618Salicylic acid; Derivatives thereof having the carboxyl group in position 1 esterified, e.g. salsalate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • 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
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • A61K9/2081Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets with microcapsules or coated microparticles according to A61K9/50
    • 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
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer
    • 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
    • A61K9/5073Microcapsules 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present application generally relates to methods and compositions for inhibiting the contraction of muscles and, in particular, to methods and compositions for inhibiting the contraction of smooth muscles of the urinary bladder.
  • the detrusor muscle is a layer of the urinary bladder wall made of smooth muscle fibers arranged in spiral, longitudinal, and circular bundles. When the bladder is stretched, this signals the parasympathetic nervous system to contract the detrusor muscle. This encourages the bladder to expel urine through the urethra.
  • the human adult urinary bladder usually holds about 300-350 ml of urine (the working volume), but a full adult bladder may hold up to about 1000 ml (the absolute volume), varying among individuals.
  • the ridges produced by folding of the wall of the bladder rugae
  • the wall of the bladder thins as it stretches, allowing the bladder to store larger amounts of urine without a significant rise in internal pressure.
  • the desire to urinate usually starts when the volume of urine in the bladder reaches around 200 ml. At this stage it is easy for the subject, if desired, to resist the urge to urinate. As the bladder continues to fill, the desire to urinate becomes stronger and harder to ignore. Eventually, the bladder will fill to the point where the urge to urinate becomes overwhelming, and the subject will no longer be able to ignore it. In some individuals, this desire to urinate starts when the bladder is less than 100% full in relation to its working volume. Such increased desire to urinate may interfere with normal activities, including the ability to sleep for sufficient uninterrupted periods of rest. In some cases, this increased desire to urinate may be associated with medical conditions such as benign prostate hyperplasia or prostate cancer in men, or pregnancy in women. However, increased desire to urinate also occurs in individuals, both male and female, who are not affected by another medical condition.
  • compositions and methods for the treatment of male and female subjects who suffer from a desire to urinate when the bladder is less than 100% full of urine in relation to its working volume are needed for the inhibition of muscle contraction in order to allow in said subjects the desire to urinate to start when the volume of urine in the bladder exceeds around 100% of its working volume.
  • One aspect of the present application relates to a method for manufacturing a pharmaceutical composition for reducing the frequency of urination.
  • the method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a first component, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after oral administration; forming a second mixture comprising a second active ingredient comprising one or more analgesic agents; coating the second mixture with a second delayed-release coating to form a second component, wherein the second delayed-release coating releases the second mixture with a lag time of 4-6 hours after oral administration; and combining the first component with the second component to form a third mixture.
  • the method comprises the steps of forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after direct contact with a body fluid; coating the core structure with a second mixture to form a coated core structure, wherein the second mixture comprises a second active ingredient comprising one or more analgesic agents; and coating the coated core structure with a second delayed-release coating to form a double-coated core structure, wherein the second delayed-release coating releases the second mixture with a lag time of 1-4 hours after oral administration.
  • Another aspect of the present application relates to pharmaceutical compositions manufactured using the method of the present application.
  • Figure 1 A and IB are diagrams showing that analgesics regulate expression of co-stimulatory molecules by Raw 264 macrophage cells in the absence ( Figure 1 A) or presence ( Figure IB) of LPS.
  • Cells were cultures for 24 hrs in the presence of analgesic alone or together with Salmonella typhymurium LPS (0.05 ⁇ g/ml). Results are mean relative % of CD40+CD80+ cells.
  • the term "effective amount” means an amount necessary to achieve a selected result.
  • analgesic refers to agents, compounds or drugs used to relieve pain and inclusive of anti-inflammatory compounds.
  • exemplary analgesic and/or anti-inflammatory agents, compounds or drugs include, but are not limited to, the following substances: non-steroidal anti-inflammatory drugs (NSAIDs), salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen
  • NSAIDs non-steroidal
  • coxib and "COX inhibitor” refer to a composition of compounds that is capable of inhibiting the activity or expression of COX2 enzymes or is capable of inhibiting or reducing the severity, including pain and swelling, of a severe inflammatory response.
  • the urinary bladder has two important functions: storage of urine and emptying. Storage of urine occurs at low pressure, which implies that the detrusor muscle relaxes during the filling phase. Emptying of the bladder requires a coordinated contraction of the detrusor muscle and relaxation of the sphincter muscles of the urethra. Disturbances of the storage function may result in lower urinary tract symptoms, such as urgency, frequency, and urge incontinence, the components of the overactive bladder syndrome.
  • the overactive bladder syndrome which may be due to involuntary contractions of the smooth muscle of the bladder (detrusor) during the storage phase, is a common and underreported problem, the prevalence of which has only recently been assessed.
  • One aspect of the present application relates to a method for reducing the frequency of urination by administering to a person in need thereof a pharmaceutical
  • composition formulated in a delayed-release formulation.
  • the pharmaceutical composition comprises one or more analgesic agents and, optionally, one or more antimuscarinic agents.
  • the term “delayed-release” refers to a medication that does not immediately disintegrate and release the active ingredient(s) into the body.
  • the term “delayed-release” is used with reference to a drug formulation having a release profile in which there is a predetermined delay in the release of the drug following administration.
  • the delayed-release formulation includes an enteric coating, which is a barrier applied to oral medication that prevents release of medication before it reaches the small intestine. Delayed-release formulations, such as enteric coatings, prevent drugs having an irritant effect on the stomach, such as aspirin, from dissolving in the stomach. Such coatings are also used to protect acid-unstable drugs from the stomach's acidic exposure, delivering them instead to a basic pH environment (intestine's pH 5.5 and above) where they do not degrade, and give their desired action.
  • pulsatile release is a type of delayed-release, which is used herein with reference to a drug formulation that provides rapid and transient release of the drug within a short time period immediately after a predetermined lag period, thereby producing a "pulsed" plasma profile of the drug after drug administration.
  • Formulations may be designed to provide a single pulsatile release or multiple pulsatile releases at predetermined time intervals following administration.
  • enteric coatings work by presenting a surface that is stable at the highly acidic pH found in the stomach, but breaks down rapidly at a less acidic (relatively more basic) pH. Therefore, an enteric coated pill will not dissolve in the acidic juices of the stomach (pH ⁇ 3), but they will in the alkaline (pH 7-9) environment present in the small intestine.
  • enteric coating materials include, but are not limited to, methyl aciylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), methyl methacrylate-methacrylic acid copolymers, sodium alginate and stearic acid.
  • enteric coating materials include, but are not limited to, methyl aciylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), methyl methacrylate-methacrylic acid copolymers, sodium alginate and stearic acid.
  • PVAP polyvinyl acetate phthalate
  • the pharmaceutical composition is orally administered from a variety of drug formulations designed to provide delayed-release.
  • Delayed oral dosage forms include, for example, tablets, capsules, caplets, and may also comprise a plurality of granules, beads, powders or pellets that may or may not be encapsulated. Tablets and capsules represent the most convenient oral dosage forms, in which case solid pharmaceutical carriers are employed.
  • one or more barrier coatings may be applied to pellets, tablets, or capsules to facilitate slow dissolution and concomitant release of drugs into the intestine.
  • the barrier coating contains one or more polymers encasing, surrounding, or forming a layer, or membrane around the therapeutic composition or active core.
  • the active agents are delivered in a formulation to provide delayed-release at a pre-determined time following administration.
  • the delay may be up to about 10 minutes, about 20 minutes, about 30 minutes, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, or longer.
  • a delayed-release composition may comprise 100% of the total dosage of a given active agent administered in a single unit dose.
  • a delayed-release composition may be included as a component in a combined release profile formulation may provide about 30-95%) of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation.
  • the immediate-release component may provide about 5-70%, or about 50%) of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation.
  • the delayed-release component provides about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% of the total dosage of the active agent(s) to be delivered by the formulation.
  • a delayed-release formulation typically comprises a barrier coating that delays the release of the active ingredient(s).
  • the barrier coating may consist of a variety of different materials, depending on the objective.
  • a formulation may comprise a plurality of barrier coatings to facilitate release in a temporal manner.
  • the coating may be a sugar coating, a film coating (e.g., based on hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone), or a coating based on methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and/or ethylcellulose.
  • a film coating e.g., based on hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone
  • methacrylic acid copolymer cellulose acetate phthalate
  • the formulation may additionally include a time delay material such as, for example, glyceryl monostearate or glyceryl distearate.
  • the delayed-release formulation includes an enteric coating comprised one or more polymers facilitating release of active agents in proximal or distal regions of the gastrointestinal tract.
  • enteric polymer coating a coating comprising of one or more polymers having a pH dependent or pH-independent release profile. Typically the coating resists dissolution in the acidic medium of the stomach, but dissolves or erodes in more distal regions of the gastrointestinal tract, such as the small intestine or colon.
  • An enteric polymer coating typically resists releases of the active agents until sometime after a gastric emptying lag period of about 3-4 hours after administration.
  • pH dependent enteric coatings comprising one or more pH-dependent or pH-sensitive polymers that maintain their structural integrity at low pH, as in the stomach, but dissolve in higher pH environments in more distal regions of the gastrointestinal tract, such as the small intestine, where the drug contents are released.
  • pH dependent is defined as having characteristics (e.g., dissolution) which vary according to environmental pH.
  • Exemplary pH-dependent polymers include, but are not limited to, methacarylic acid copolymers, methacrylic acid-methyl methacrylate copolymers (e.g.,
  • EUDRAGIT® LI 00 Type A
  • EUDRAGIT® S I 00 Type B
  • methacrylic acid-ethyl acrylate copolymers e.g., EUDRAGIT® LI 00-55 (Type C) and
  • EUDRAGIT® L30D-55 copolymer dispersion Rohm GmbH, Germany
  • copolymers of methacrylic acid-methyl methacrylate and methyl methacrylate (EUDRAGIT® FS); terpolymers of methacrylic acid, methacrylate, and ethyl acrylate; cellulose acetate phthalates (CAP);
  • HPMCP hydroxypropyl methylcellulose phthalate
  • PVAP polyvinyl acetate phthalates
  • COATERIC® COATERIC®, OPADRY® enteric white OY-P-7171
  • CAS cellulose acetate succinates
  • HPMCAS hydroxypropyl methylcellulose acetate succinate
  • shellac e.g., MarcoatTM 125 & MarcoatTM 125N
  • carboxymethyl ethylcellulose CMEC, Freund Corporation, Japan
  • CAP cellulose acetate trimellitates
  • pH-dependent polymers typically exhibit a characteristic pH optimum for dissolution.
  • the pH-dependent polymer exhibits a pH optimum between about 5.0 and 5.5, between about 5.5 and 6.0, between about 6.0 and 6.5, or between about 6.5 and 7.0.
  • the pH-dependent polymer exhibits a pH optimum of >5.0, of >5.5, of >6.0, of >6.5, or of >7.0.
  • the enteric coating may comprise one or more
  • pH-independent polymers These polymers provide for release of the drug after a certain time, independent of the pH.
  • pH independent is defined as having characteristics (e.g., dissolution) which are substantially unaffected by pH. pH independent polymers are often referred to in the context of "time-controlled" or
  • a pH independent polymer may be water-insoluble or water soluble.
  • Exemplary water insoluble pH independent polymers include, but are not limited to, neutral methacrylic acid esters with a small portion of trimethylammonioethyl methacrylate chloride (e.g., EUDRAGIT® RS and EUDRAGIT® RL; neutral ester dispersions without any functional groups (e.g., EUDRAGIT® E30D and EUDRAGIT® E30); cellulosic polymers, such as ethylcellulose, hydroxyl ethyl cellulose, cellulose acetate or mixtures and other pH independent coating products.
  • Exemplary water soluble pH independent polymers include OPADRY®amb.
  • the pH independent polymers contain one or more polysaccharides that are resistant to erosion in both the stomach and intestine. Such polymers can be only degraded in the colon, which contains a large microflora containing biodegradable enzymes breaking down, for example, the polysaccharide coatings to release the drug contents in a controlled, time-dependent manner.
  • the coating methodology employs the blending of one or more pH-dependent and one or more pH-independent polymers.
  • pH-dependent and pH-independent polymers can reduce the release rate of active ingredients once the soluble polymer has reached its optimum pH of solubilization.
  • a "time-controlled" or “time-dependent” release profile can be obtained using a water insoluble capsule body containing one or more active agents, wherein the capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug.
  • the plug Upon contact with gastrointestinal fluid or dissolution medium, the plug swells, pushing itself out of the capsule and releasing the drugs after a pre-determined lag time, which can be controlled by e.g., the position and dimensions of the plug.
  • the capsule body may be further coated with an outer pH-dependent enteric coating keeping the capsule intact until it reaches the small intestine.
  • Suitable plug materials include, for example, polymethacrylates, erodible compressed polymers (e.g., HPMC, polyvinyl alcohol), congealed melted polymer (e.g., glyceryl mono oleate) and enzymatically controlled erodible polymers (e.g., polysaccharides, such as amylose, arabinogalactan, chitosan, chondroitin sulfate, cyclodextrin, dextran, guar gum, pectin and xylan).
  • erodible compressed polymers e.g., HPMC, polyvinyl alcohol
  • congealed melted polymer e.g., glyceryl mono oleate
  • enzymatically controlled erodible polymers e.g., polysaccharides, such as amylose, arabinogalactan, chitosan, chondroitin sulfate, cyclodextrin
  • capsules or bilayered tablets may be formulated to contain a drug-containing core, covered by a swelling layer, and an outer insoluble, but semi-permeable polymer coating or membrane.
  • the lag time prior to rupture can be controlled by the permeation and mechanical properties of the polymer coating and the swelling behavior of the swelling layer.
  • the swelling layer comprises one or more swelling agents, such as swellable hydrophilic polymers that swell and retain water in their structures.
  • Exemplary water swellable materials include, but are not limited to, polyethylene oxide (having e.g., an average molecular weight between 1,000,000 to 7,000,000, such as POLYOX ® ), methylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose;
  • polyalkylene oxides having a weight average molecular weight of 100,000 to 6,000,000, including but not limited to poly(methylene oxide), poly(butylene oxide); poly(hydroxy alkyl methacrylate) having a molecular weight of from 25,000 to 5,000,000; poly(vinyl)alcohol, having a low acetal residue, which is cross-linked with glyoxal, formaldehyde or glutaraldehyde and having a degree of polymerization of from 200 to 30,000; mixtures of methyl cellulose, cross-linked agar and carboxymethyl cellulose; hydrogel forming copolymers produced by forming a dispersion of a finely divided copolymer of maleic anhydride with styrene, ethylene, propylene, butylene or isobutylene cross-linked with from 0.001 to 0.5 moles of saturated cross-linking agent per mole of maleic anyhydride in the copolymer; CARBOPOL ® acidic carboxy polymers having
  • AQUA-KEEPS ® acrylate polymer polysaccharides composed of condensed glucose units such as diester cross-linked polyglucan; carbomers having a viscosity of 3,000 to 60,000 mPa as a 0.5%-l% w/v aqueous solution; cellulose ethers such as hydroxypropylcellulose having a viscosity of about 1000-7000 mPa s as a 1% w/w aqueous solution (25° C); hydroxypropyl methylcellulose having a viscosity of about 1000 or higher, preferably 2,500 or higher to a maximum of 25,000 mPa as a 2% w/v aqueous solution; polyvinylpyrrolidone having a viscosity of about 300-700 mPa s as a 10% w/v aqueous solution at 20° C; and combinations thereof.
  • the enteric layer may further comprise anti-tackiness agents, such as talc or glyceryl monostearate and/or plasticizers such as.
  • the enteric layer may further comprise one or more plasticizers including, but not limited to, triethyl citrate, acetyl triethyl citrate, acetyltributyl citrate, polyethylene glycol acetylated monoglycerides, glycerin, triacetin, propylene glycol, phthalate esters (e.g., diethyl phthalate, dibutyl phthalate), titanium dioxide, ferric oxides, castor oil, sorbitol and dibutyl sebacate.
  • plasticizers including, but not limited to, triethyl citrate, acetyl triethyl citrate, acetyltributyl citrate, polyethylene glycol acetylated monoglycerides, glycerin, triacetin, propylene
  • the delay release formulation employs a water-permeable but insoluble film coating to enclose the active ingredient and an osmotic agent utilizing a enclosing. As water from the gut slowly diffuses through the film into the core, the core swells until the film bursts, thereby releasing the active ingredients.
  • the film coating may be adjusted to permit various rates of water permeation or release time.
  • the release time of the drugs can be controlled by a disintegration lag time depending on the balance between the tolerability and thickness of a water insoluble polymer membrane (such as ethyl cellulose, EC) containing predefined micropores at the bottom of the body and the amount of a swellable excipient, such as low substituted hydroxypropyl cellulose (L-HPC) and sodium glycolate.
  • a water insoluble polymer membrane such as ethyl cellulose, EC
  • L-HPC low substituted hydroxypropyl cellulose
  • GI fluids permeate through the micropores, causing swelling of the swellable excipients, which produces an inner pressure disengaging the capsular components, including a first capsule body containing the swellable materials, a second capsule body containing the drugs, and an outer cap attached to the first capsule body.
  • the drugs may be released by an osmotic mechanism.
  • a capsule may be formulated with a single osmotic unit or it may incorporate 2, 3, 4, 5, or 6 push-pull units encapsulated within a hard gelatin capsule, whereby each bilayer push pull unit contains an osmotic push layer and a drug layer, both surrounded by a
  • semi-permeable membrane One or more orifices are drilled through the membrane next to the drug layer.
  • This membrane may be additionally covered with a pH-dependent enteric coating to prevent release until after gastric emptying.
  • the gelatin capsule dissolves immediately after ingestion.
  • the enteric coating breaks down, which then allows fluid to flow through the semi-permeable membrane, swelling the osmotic push compartment to force drugs out through the orifice(s) at a rate precisely controlled by the rate of water transport through the semi-permeable membrane. Release of drugs can occur over a constant rate for up to 24 hours or more.
  • the osmotic push layer comprises one or more osmotic agents creating the driving force for transport of water through the semi-permeable membrane into the core of the delivery vehicle.
  • osmotic agents include water-swellable hydrophilic polymers, also referred to as “osmopolymers” and “hydrogels,” including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium cros
  • osmogens which are capable of imbibing water to effect an osmotic pressure gradient across the semi-permeable membrane.
  • exemplary osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic
  • Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are
  • water-permeable and water-insoluble at physiologically relevant pHs or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking.
  • the delay release formulation employs a
  • the pharmaceutical composition is in a tablet or capsule form containing a single coating layer. In other embodiments, the pharmaceutical composition is in a tablet or capsule form containing multiple coating layers.
  • the pharmaceutical composition comprises a plurality of active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.
  • active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.
  • spasmolytics include, but are not limited to, carisoprodol, benzodiazepines, baclofen, cyclobenzaprine, metaxalone, methocarbamol, clonidine, clonidine analog, and dantrolene.
  • the pharmaceutical composition comprises one or more analgesics.
  • the pharmaceutical composition comprises (1) one or more analgesics, and (2) one or more other active ingredients selected from the group consisting of antimuscarinic agents, antidiuretics and spasmolytics.
  • the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two antimuscarinic agents.
  • the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two antidiuretics.
  • the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two spasmolytics.
  • the pharmaceutical composition comprises (1) one or two analgesics, (2) one or two antimuscarinic agents, and (3) one or two antidiuretics.
  • the plurality of active ingredients are formulated for immediate-release. In other embodiment, the plurality of active ingredients are formulated for delayed-release. In other embodiment, the plurality of active ingredients are formulated for both immediate-release and delayed-release (e.g., a first portion of each active ingredient is formulated for immediate-release and a second portion of each active ingredient is formulated for delayed-release). In yet other embodiment, some of the plurality of active ingredients are formulated for immediate-release and some of the plurality of active ingredients are formulated for delayed-release (e.g., active ingredients A, B, C are formulated for immediate-release and active ingredients C and D are formulated for delayed-release).
  • the pharmaceutical composition comprises an immediate-release component and a delayed-release component.
  • the immediate-release component may comprise one or more active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.
  • the delayed-release component may comprise one or more active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.
  • the immediate-release component and the delayed-release component have exactly the same active ingredients.
  • the immediate-release component and the delayed-release component have different active ingredients.
  • the immediate-release component and the delayed-release component have one or more common active ingredients.
  • the pharmaceutical composition comprises two active ingredients (e.g., two analgesic agents, or a mixture of one analgesic agent and one
  • the pharmaceutical composition comprises two active ingredients, formulated for delayed-release at about the same time.
  • the pharmaceutical composition comprises two active ingredients formulated as two delayed-release components, each providing a different delayed-release profile. For example, a first delayed-release component releases a first active ingredient at a first time point and a second delayed-release component releases a second active ingredient at a second time point.
  • the pharmaceutical composition comprises two active ingredients, one is formulated for immediate-release and the other is formulated for delayed-release.
  • the pharmaceutical composition comprises two active ingredients (e.g., two analgesic agents, or a mixture of one analgesic agent and one
  • the pharmaceutical composition comprises three active ingredients formulated for immediate-release, and (2) three active ingredients formulated for delayed-release.
  • the pharmaceutical composition comprises four active ingredients formulated for immediate-release, and (2) four active ingredients formulated for delayed-release.
  • the active ingredient(s) in the immediate-release component can be the same as, or different from, the active ingredient(s) in the delayed-release component.
  • immediate-release is used herein with reference to a drug formulation that does not contain a dissolution rate controlling material. There is substantially no delay in the release of the active agents following administration of an immediate-release formulation.
  • An immediate-release coating may include suitable materials immediately dissolving following administration so as to release the drug contents therein.
  • Exemplary immediate-release coating materials include gelatin, polyvinyl alcohol polyethylene glycol (PVA-PEG) copolymers (e.g., KOLLICOAT ® ) and various others materials known to those skilled in the art.
  • An immediate-release composition may comprise 100% of the total dosage of a given active agent administered in a single unit dose.
  • an immediate-release component may be included as a component in a combined release profile formulation that may provide about 1% to about 50% of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation.
  • the immediate-release component may provide at least about 5%, or about 10% to about 30%, or about 45% to about 50% of the total dosage of the active agent(s) to be delivered by the formulation.
  • the rest of the active agent(s) may be delivered in a delayed-release formulation.
  • the immediate-release component provides about 10, 15, 20, 25, 30, 35, 40, 45 or 50% of the total dosage of the active agent(s) to be delivered by the formulation.
  • the delayed-release component provides about 90, 85, 80, 75, 70, 65, 60, 55 or 50% of the total dosage of the active agent(s) to be delivered by the formulation.
  • the immediate-release or delayed-release formulation comprises an active core comprised of one or more inert particles, each in the form of a bead, pellet, pill, granular particle, microcapsule, microsphere, microgranule, nanocapsule, or nanosphere coated on its surfaces with drugs in the form of e.g. , a drug-containing film-forming composition using, for example, fluid bed techniques or other methodologies known to those of skill in the art.
  • the inert particle can be of various sizes, so long as it is large enough to remain poorly dissolved.
  • the active core may be prepared by granulating and milling and/or by extrusion and spheronization of a polymer composition containing the drug substance.
  • the amount of drug in the core will depend on the dose that is required, and typically varies from about 5 to 90 weight %.
  • the polymeric coating on the active core will be from about 1 to 50% based on the weight of the coated particle, depending on the lag time and type of release profile required and/or the polymers and coating solvents chosen.
  • the inactive core may be a sugar sphere or a buffer crystal or an encapsulated buffer crystal such as calcium carbonate, sodium bicarbonate, fumaric acid, tartaric acid, etc., which alters the
  • the delayed-release formulation is formed by coating a water soluble/dispersible drug-containing particle, such as a bead, with a mixture of a water insoluble polymer and an enteric polymer, wherein the water insoluble polymer and the enteric polymer may be present at a weight ratio of from 4: 1 to 1 : 1, and the total weight of the coatings is 10 to 60 weight % based on the total weight of the coated beads.
  • the drug layered beads may optionally include an inner dissolution rate controlling membrane of ethylcellulose.
  • the composition of the outer layer, as well as the individual weights of the inner and outer layers of the polymeric membrane are optimized for achieving desired circadian rhythm release profiles for a given active, which are predicted based on in vitro/in vivo correlations.
  • the formulations comprise a mixture of immediate-release drug-containing particles without a dissolution rate controlling polymer membrane and delayed-release beads exhibiting, for example, a lag time of 2-4 hours following oral
  • the formulations comprise a mixture of two types of delayed-release beads: a first type that exhibits a lag time of 1-3 hours and a second type that exhibits a lag time of 4-6 hours.
  • the active core is coated with one or more layers of dissolution rate-controlling polymers to obtain desired release profiles with or without a lag time.
  • An inner layer membrane can largely control the rate of drug release following imbibition of water or body fluids into the core, while the outer layer membrane can provide for a desired lag time (the period of no or little drug release following imbibition of water or body fluids into the core).
  • the inner layer membrane may comprise a water insoluble polymer, or a mixture of water insoluble and water soluble polymers.
  • the polymers suitable for the outer membrane, which largely controls the lag time of up to 6 hours may comprise an enteric polymer, as described above, and a water insoluble polymer at 10 to 50 weight %.
  • the ratio of water insoluble polymer to enteric polymer may vary from 4: 1 to 1 :2, preferably the polymers are present at a ratio of about 1 : 1.
  • the water insoluble polymer typically used is ethylcellulose.
  • Exemplary water insoluble polymers include ethylcellulose, polyvinyl acetate (Kollicoat SR#0D from BASF), neutral copolymers based on ethyl acrylate and
  • methylmethacrylate copolymers of acrylic and methacrylic acid esters with quaternary ammonium groups such as EUDRAGIT ® E, RS and RS30D, RL or RL30D and the like.
  • Exemplary water soluble polymers include low molecular weight HPMC, HPC, methylcellulose, polyethylene glycol (PEG of molecular weight>3000) at a thickness ranging from 1 weight % up to 10 weight % depending on the solubility of the active in water and the solvent or latex suspension based coating formulation used.
  • the water insoluble polymer to water soluble polymer may typically vary from 95:5 to 60:40, preferably from 80:20 to 65:35.
  • the formulations are designed with release profiles to limit interference with restful sleep, wherein the formulation releases the medicine when the individual would normally be awakened by an urge to urinate. For example, consider an individual who begins sleeping at 11 PM and is normally awakened at 12:30 AM, 3 :00 AM, and 6:00 AM to urinate. A delayed-release vehicle could deliver the medicine at 12: 15 AM, thereby delaying the need to urinate for perhaps 2-3 hours.
  • the pharmaceutical composition may be administered daily or administered on an as needed basis.
  • the pharmaceutical composition is administered to the subject prior to bedtime.
  • the pharmaceutical composition is administered immediately before bedtime.
  • the pharmaceutical composition is administered within about two hours before bedtime, preferably within about one hour before bedtime.
  • the pharmaceutical composition is administered about two hours before bedtime.
  • the pharmaceutical composition is administered at least two hours before bedtime.
  • the pharmaceutical composition is administered about one hour before bedtime.
  • the pharmaceutical composition is administered at least one hour before bedtime.
  • the pharmaceutical composition is administered less than one hour before bedtime.
  • the pharmaceutical composition is administered immediately before bedtime.
  • the pharmaceutical composition is administered orally.
  • the appropriate dosage ("therapeutically effective amount") of the active agent(s) in the immediate-component or delayed-release component will depend, for example, the severity and course of the condition, the mode of administration, the bioavailability of the particular agent(s), the age and weight of the patient, the patient's clinical history and response to the active agent(s), discretion of the physician, etc.
  • the therapeutically effective amount of the active agent(s) in the immediate-release component or the delayed-release component is administered in the range of about 100 ⁇ g/kg body weight/day to about 100 mg/kg body weight/day whether by one or more administrations.
  • the range of each active agent administered daily is from about 100 ⁇ g/kg body weight/day to about 50 mg/kg body weight/day, 100 ⁇ g/kg body weight/day to about 10 mg/kg body weight/day, 100 ⁇ g/kg body weight/day to about 1 mg/kg body weight/day, 100 ⁇ g/kg body weight/day to about 10 mg/kg body weight/day, 500 ⁇ g/kg body weight/day to about 100 mg/kg body weight/day, 500 ⁇ g/kg body weight/day to about 50 mg/kg body weight/day, 500 ⁇ g/kg body weight/ day to about 5 mg/kg body weight/ day, 1 mg/kg body weight/day to about 100 mg/kg body weight/day, 1 mg/kg body weight/day to about 50 mg/kg body weight/ day, 1 mg/kg body weight/day to about 10 mg/kg body weight/day, 5 mg/kg body weight/dose to about 100 mg/kg body weight/day, 5 mg/kg body weight/dose to about 50 mg/kg body weight/day, 5
  • the active agent(s) described herein may be included in an immediate-release component or a delayed-release component or combinations thereof for daily oral administration at a single dose or combined dose range of 1 mg to 2000 mg, 5 mg to 2000 mg, 10 mg to 2000 mg, 50 mg to 2000 mg, 100 mg to 2000 mg, 200 mg to 2000 mg, 500 mg to 2000 mg, 5 mg to 1800 mg, 10 mg to 1600 mg, 50 mg to 1600 mg, 100 mg to 1500 mg, 150 mg to 1200 mg, 200 mg to 1000 mg, 300 mg to 800 mg, 325 mg to 500 mg, 1 mg to 1000 mg, 1 mg to 500 mg, 1 mg to 200 mg, 5 mg to 1000 mg, 5 mg to 500 mg, 5 mg to 200 mg, 10 mg to 1000 mg, 10 mg to 500 mg, 10 mg to 200 mg, 50 mg to 1000 mg, 50 mg to 500 mg, 50 mg to 200 mg, 100 mg to 250 mg, 100 mg to 500 mg, 250 mg to 1000 mg, 250 mg to 500 mg, 500 mg to 1000 mg, 500 mg to 2000 mg. As expected, the dosage will be dependent on
  • the pharmaceutical composition comprises a single analgesic agent.
  • the single analgesic agent is aspirin.
  • the single analgesic agent is ibuprofen.
  • the single analgesic agent is naproxen sodium.
  • the single analgesic agent is indomethacin.
  • the single analgesic agent is nabumetone.
  • the single analgesic agent is acetaminophen.
  • the single analgesic agent is given at a daily dose of 1 mg to 2000 mg, 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg.
  • the pharmaceutical composition comprises acetylsalicylic acid, ibuprofen, naproxen sodium, indomethancin, nabumetone or acetaminophen as a single analgesic agent and the analgesic agent is administered orally at a daily dose in the range of 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg.
  • a second analgesic agent is given at a daily dose of 1 mg to 2000 mg, 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg.
  • the pharmaceutical composition comprises a pair of analgesic agents.
  • paired analgesic agents include, but are not limited to, acetylsalicylic acid and ibuprofen, acetylsalicylic acid and naproxen sodium, acetylsalicylic acid and nabumetone, acetylsalicylic acid and acetaminophen, acetylsalicylic acid and indomethancin, ibuprofen and naproxen sodium, ibuprofen and nabumetone, ibuprofen and acetaminophen, ibuprofen and indomethancin, naproxen sodium and nabumetone, naproxen sodium and acetaminophen, naproxen sodium and indomethancin, nabumetone and acetaminophen, naproxen sodium and indomethancin, nabumetone and acetaminophen,
  • the paired analgesic agents are mixed at a weight ratio in the range of 0.1 : 1 to 10: 1, 0.2: 1 to 5: 1 or 0.3 : 1 to 3 : 1, with a combined dose in the range of 5 mg to 2000 mg, 20 mg to 2000 mg, 100 mg to 2000 mg, 200 mg to 2000 mg, 500 mg to 2000 mg, 5 mg to 1500 mg, 20 mg to 1500 mg, 100 mg to 1500 mg, 200 mg to 1500 mg, 500 mg to 1500 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 100 mg to 1000 mg, 250 mg to 500 mg, 250 mg to 1000 mg, 250 mg to 1500 mg, 500 mg to 1000 mg, 500 mg to 1500 mg, 1000 mg to 1500 mg, and 1000 mg to 2000 mg.
  • the paired analgesic agents are mixed at a weight ratio of 1 : 1.
  • the pharmaceutical composition of the present application further comprises one or more antimuscarinic agents. Examples of the
  • antimuscarinic agents include, but are not limited to, oxybutynin, solifenacin, darifenacin, fesoterodine, tolterodine, trospium and atropine.
  • the daily dose of antimuscarinic agent is in the range of 0.01 mg to 100 mg, 0.1 mg to 100 mg, 1 mg to 100 mg, 10 mg to 100 mg, 0.01 mg to 25 mg, 0.1 mg to 25 mg, 1 mg to 25 mg, 10 mg to 25 mg, 0.01 mg to 10 mg, 0.1 mg to 10 mg, 1 mg to 10 mg, 10 mg to 100 mg and 10 mg to 25 mg.
  • Another aspect of the present application relates to a method for reducing the frequency of urination by administering to a person in need thereof a pharmaceutical
  • composition formulated in an immediate-release formulation.
  • the pharmaceutical composition comprises one or more analgesic agents and one or more
  • the pharmaceutical composition comprises one or more analgesic agents and one or more antidiuretic agents. In yet other embodiments, the pharmaceutical composition comprises one or more analgesic agents, one or more antimuscarinic agents, and one or more antidiuretic agents.
  • the pharmaceutical composition of the present application further comprises one or more spasmolytics.
  • spasmolytics include, but are not limited to, carisoprodol, benzodiazepines, baclofen, cyclobenzaprine, metaxalone,
  • the spasmolytics is used at a daily dose of 1 mg to 1000 mg, 1 mg to 100 mg, 10 mg to 1000 mg, 10 mg to 100 mg, 20 mg to 1000 mg, 20 mg to 800 mg, 20 mg to 500 mg, 20 mg to 200 mg, 50 mg to 1000 mg, 50 mg to 800 mg, 50 mg to 200 mg, 100 mg to 800 mg, 100 mg to 500 mg, 200 mg to 800 mg, and 200 mg to 500 mg.
  • the spasmolytics may be formulated, alone or together with other active ingredient(s) in the pharmaceutical composition, for immediate-release, delayed-release or combinations thereof.
  • the pharmaceutical composition comprises two or more analgesic agents.
  • the pharmaceutical composition comprises one or more analgesic agents and one or more antimuscarinic agents and/or antidiuretic agents.
  • the pharmaceutical composition may be formulated into a tablet, capsule, dragee, powder, granulate, liquid, gel or emulsion form. Said liquid, gel or emulsion may be ingested by the subject in naked form or contained within a capsule.
  • the analgesic agent is selected from the group consisting of salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin; enolic acids, oxicam derivatives, piroxicam, meloxicam, tenoxicam, ampiroxicam, droxicam, pivoxicam, pyrazolon
  • the antimuscarinic agent is selected from the group consisting of oxybutynin, solifenacin, darifenacin and atropine.
  • the pharmaceutical composition comprises a single analgesic agent and a single antimuscarinic agent.
  • the single analgesic agent is aspirin.
  • the single analgesic agent is ibuprofen.
  • the single analgesic agent is naproxen sodium.
  • the single analgesic agent is indomethacin.
  • the single analgesic agent is nabumetone.
  • the single analgesic agent is acetaminophen.
  • the analgesic agent and anti-muscarinic agent may be given at doses in the ranges described above.
  • Another aspect of the present application relates to a method for treating nocturia by administering to a subject in need thereof (1) one or more analgesic agents and (2) one or more antidiuretic agents.
  • the antidiuretic agent(s) act to: (1) increase vasopressin secretion; (2) increase vasopressin receptor activation; (3) reduce secretion of atrial natriuretic peptide (ANP) or C-type natriuretic peptide (C P); or (4) reduce A P and/or C P receptor activation.
  • antidiuretic agents include, but are not limited to, antidiuretic hormone (ADH), angiotensin II, aldosterone, vasopressin, vasopressin analogs (e.g., ADH), angiotensin II, aldosterone, vasopressin, vasopressin analogs (e.g.
  • vasopressin receptor agonists atrial natriuretic peptide (ANP) and C-type natriuretic peptide (CNP) receptor (i.e., NPR1, NPR2, NPR3) antagonists (e.g., HS-142-1, isatin, [Asu7,23']b-ANP-(7-28)], anantin, a cyclic peptide from Streptomyces coerulescens, and 3G12 monoclonal antibody); somatostatin type 2 receptor antagonists (e.g., somatostatin), and pharmaceutically-acceptable derivatives, analogs, salts, hydrates, and solvates thereof.
  • NPR1, NPR2, NPR3 antagonists e.g., HS-142-1, isatin, [Asu7,23']b-ANP-(7-28)], anantin, a cyclic peptide from Streptomyces coerulescens, and 3G12 mono
  • the one or more analgesic agent and one or more antidiuretic agents are formulated for delayed-release.
  • Another aspect of the present application relates to a method for treating nocturia by administering to a person in need thereof a first pharmaceutical composition comprising a diuretic, followed with a second pharmaceutical composition comprising one or more analgesic agents.
  • the first pharmaceutical composition is dosed and formulated to have a diuretic effect within 6 hours of administration and is administered at least 8 hours prior to bedtime.
  • the second pharmaceutical composition is administered within 2 hours prior to bedtime.
  • diuretics include, but are not limited to, acidifying salts, such as CaCl 2 and NH 4 C1; arginine vasopressin receptor 2 antagonists, such as amphotericin B and lithium citrate; aquaretics, such as Goldenrod and Junipe; Na-H exchanger antagonists, such as dopamine; carbonic anhydrase inhibitors, such as acetazolamide and dorzolamide; loop diuretics, such as bumetanide, ethacrynic acid, furosemide and torsemide; osmotic diuretics, such as glucose and mannitol; potassium-sparing diuretics, such as amiloride, spironolactone,
  • triamterene, potassium canrenoate triamterene, potassium canrenoate
  • thiazides such as bendroflumethiazide
  • hydrochlorothiazide such as caffeine, theophylline and theobromine.
  • the second pharmaceutical composition further comprises one or more antimuscarinic agents.
  • antimuscarinic agents include, but are not limited to, oxybutynin, solifenacin, darifenacin, fesoterodine, tolterodine, trospium and atropine.
  • the second pharmaceutical composition may be formulated in immediate-release formulation or delayed-release formulation.
  • the first pharmaceutical composition is formulated for immediate-release and the second pharmaceutical composition is formulated for delayed-release.
  • the second pharmaceutical composition further comprises one or more antidiuretic agents.
  • a pharmaceutical composition comprising a plurality of active ingredients and a pharmaceutically acceptable carrier.
  • the plurality of active ingredients comprise two or more analgesics.
  • the plurality of active ingredients comprise one or more analgesics and one or more antimuscarinic agents.
  • the plurality of active ingredients comprise one or more analgesics and one or more antidiuretic agents.
  • the plurality of active ingredients comprise one or more analgesics, one or more antidiuretic agents, and one or more antimuscarinic agents.
  • at least one of said plurality of active ingredients is formulated for delayed-release.
  • the pharmaceutical composition comprises two analgesics selected from the group consisting of cetylsalicylic acid, ibuprofen, naproxen sodium, nabumetone, acetaminophen and indomethancin. In other embodiments, the pharmaceutical composition comprises one or more analgesics selected from the group consisting of
  • cetylsalicylic acid ibuprofen, naproxen sodium, nabumetone, acetaminophen and indomethancin; and an antimuscarinic agent selected from the group consisting of oxybutynin, solifenacin, darifenacin and atropine.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, sweeteners and the like.
  • the pharmaceutically acceptable carriers may be prepared from a wide range of materials including, but not limited to, flavoring agents, sweetening agents and miscellaneous materials such as buffers and absorbents that may be needed in order to prepare a particular therapeutic composition.
  • flavoring agents, sweetening agents and miscellaneous materials such as buffers and absorbents that may be needed in order to prepare a particular therapeutic composition.
  • the use of such media and agents with pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
  • Another aspect of the present application relates to a method for reducing the frequency of urination by administering to a subject in need thereof, two or more analgesic agents alternatively to prevent the development of drug resistance.
  • the method comprises administering a first analgesic agent for a first period of time and then administering a second analgesic agent for a second period of time.
  • the method further comprises administering a third analgesic agent for a third period of time.
  • the first, second and third analgesic agents are different from each other and may be selected from the group consisting of: salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin; enolic acids, oxicam derivatives, piroxicam, meloxicam, tenoxicam, ampiroxicam, droxicam, pivoxicam,
  • the first analgesic agent is acetaminophen
  • the second analgesic agent is ibuprofen
  • the third analgesic agent is naproxen sodium.
  • the length of each period may vary depending on the subject's response to each analgesic agent. In some embodiments, each period lasts from 3 days to three weeks. In another embodiment, one or more of the first, second and third analgesic is formulated for delayed-release.
  • Another aspect of the present application relates to a pharmaceutical composition
  • a pharmaceutical composition comprising (1) a first component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 1-4 hours after oral
  • a second component comprising one or more analgesic agents, wherein the second component is formulated for delayed-release with a lag time of 4-6 hours after oral administration.
  • the one or more analgesic agents in the first component or the second component are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the second component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
  • analgesic agent such as acetaminophen
  • the pharmaceutical composition is coated with an enteric coating.
  • the first component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the second components further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • both the first and second components further comprise an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the second component is coated with a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
  • WIP water insoluble polymer
  • EP enteric polymer
  • the second component is coated with multiple coating layers.
  • the second component is covered by a swelling layer and an outer insoluble but semi-permeable polymer coating.
  • the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
  • the second component formulation comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
  • compositions comprising (1) a first component comprising an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg, wherein the first component is formulated for immediate-release; and (2) a second component comprising one or more analgesic agents, wherein the second component is formulated for delay ed-release with a lag time of 1-4 hours after oral administration; and (3) a third component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 4-6 hours after oral administration.
  • an analgesic agent such as acetaminophen
  • the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the second component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
  • analgesic agent such as acetaminophen
  • the second component is coated with an enteric coating.
  • the third component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
  • the first component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the second component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the third component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first and the second components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first and the third components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and
  • the second and the third components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first, the second, and the second components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the second component is coated with an enteric coating.
  • the second component or the third component is coated with a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
  • WIP water insoluble polymer
  • EP enteric polymer
  • the third component is coated with multiple coating layers.
  • composition pharmaceutical composition for reducing the frequency of urination, comprising: (1) a first component comprising acetaminophen in an amount of 5-2000 mg, wherein the first component is formulated for immediate-release; and (2) a second component comprising one or more analgesic agents, wherein the second component is formulated for delayed-release with a lag time of 1-4 hours after oral administration; and (3) a third component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 4-6 hours after oral administration, wherein the pharmaceutical composition reduces the frequency of urination in patients in need thereof.
  • the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a first component, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after oral administration; forming a second mixture comprising a second active ingredient comprising one or more analgesic agents; coating the second mixture with a second delayed-release coating to form a second component, wherein the second delayed-release coating releases the second mixture with a lag time of 4-6 hours after oral administration; and combining the first component with the second component to form a third mixture.
  • the first, second and/or third mixture comprises a pharmaceutically acceptable carrier, such as an excipient.
  • the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • At least one of the first and the second active ingredients contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen.
  • an analgesic agent such as acetaminophen.
  • the first active ingredient contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen.
  • the second active ingredient contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen.
  • the first and the second active ingredients each contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen.
  • At least one of the first and the second active ingredients further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the second active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first and the second active ingredients each contains an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first delay ed-release coating is an enteric coating.
  • the second delayed-release coating comprises a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
  • the second delayed-release coating comprises multiple coating layers.
  • the second delayed-release coating comprises a swelling layer and an outer insoluble but semi-permeable polymer coating.
  • the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
  • the second delayed-release coating comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
  • the method further comprises the step of pressing or molding the third mixture into a tablet form.
  • the third mixture is formed by combining the first component and the second component with a third component, wherein the third component comprises a third active ingredient comprising one or more analgesic agents.
  • the one or more analgesic agents are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the first, second and third active ingredients each comprises one or more analgesic agents.
  • the one or more analgesic agents are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the first, second and third active ingredients each comprises 5-2000 mg acetaminophen.
  • one of the first, second and third active ingredients comprises 5-2000 mg acetaminophen.
  • two of the first, second and third active ingredients comprise 5-2000 mg acetaminophen.
  • the method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a core structure, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after direct contact with a body fluid; coating the core structure with a second mixture to form a coated core structure, wherein the second mixture comprises a second active ingredient comprising one or more analgesic agents; and coating the coated core structure with a second delayed-release coating to form a double-coated core structure, wherein the second delayed-release coating releases the second mixture with a lag time of 1-4 hours after oral administration.
  • the first mixture does not have direct contact with a body fluid until the second mixture is released.
  • the one or more analgesic agents in the first active ingredient or the second active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the second delayed-release coating is an enteric coating.
  • the first active ingredient or the second active ingredient or both further comprise an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first active ingredient or the second active ingredient or both comprise acetaminophen.
  • the method further comprises the step of coating the double-coated core structure with a third coating, wherein the third coating comprises a third active ingredient comprising one or more analgesic agents.
  • the one or more analgesic agents in the third active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
  • the third active ingredient comprises 5 mg to 2000 mg analgesic agent.
  • the third active ingredient comprises 5 mg to 2000 mg acetaminophen.
  • the third active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
  • the first, second and third active ingredients each comprises acetaminophen.
  • the first delayed-release coating comprises pH
  • the first delayed-release coating, or the second delayed-release coating, or both comprise a water-permeable but insoluble film coating to enclose the active ingredient and an osmotic agent in the core structure or the coated core structure. As water from the gut slowly diffuses through the insoluble film into the core structure, the core swells until the film bursts, thereby releasing the active ingredients.
  • the film coating may be adjusted to permit various rates of water permeation or release time.
  • the release time of the drugs can be controlled by a disintegration lag time depending on the balance between the tolerability and thickness of a water insoluble polymer membrane (such as ethyl cellulose, EC) containing predefined micropores at the bottom of the body and the amount of a swellable excipient, such as low substituted hydroxypropyl cellulose (L-HPC) and sodium glycolate.
  • a water insoluble polymer membrane such as ethyl cellulose, EC
  • L-HPC low substituted hydroxypropyl cellulose
  • the drugs are released by an osmotic mechanism.
  • the first delayed-release coating, or the second delayed-release coating, or both comprise a semi-permeable membrane.
  • the semi-permeable membrane is additionally covered with a pH-dependent enteric coating to prevent release until after gastric emptying.
  • EXAMPLE 2 EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON MACROPHAGE RESPONSES TO INFLAMMATORY
  • This study is designed to determine the dose and in vitro efficacy of analgesics and antimuscarinic agents in controlling macrophage response to inflammatory and
  • non-inflammatory stimuli mediated by Cox-2 and prostaglandins (PGE, PGH, etc.). It establishes baseline (dose and kinetic) responses to inflammatory and non-inflammatory effectors in bladder cells. Briefly, cultured cells are exposed to analgesic agents and/or antimuscarinic agents in the absence or presence of various effectors.
  • the effectors include: lipopolysaccharide (LPS), an inflammatory agent and COX2 inducer, as inflammatory stimuli; carbachol or acetylcholine, a stimulator of smooth muscle contraction, as non-inflammatory stimuli; botulinum neurotoxin A, a known inhibitor of acetylcholine release, as positive control; and arachidonic acid (AA), gamma linolenic acid (DGLA) or eicosapentaenoic acid (EPA) as precursors of prostaglandins, which are produced following the sequential oxidation of AA, DGLA or EPA inside the cell by cyclooxygenases (COX1 and COX2) and terminal prostaglandin synthases.
  • LPS lipopolysaccharide
  • COX2 COX2 inducer
  • the analgesic agents include: Salicylates such as aspirin,
  • iso-butyl-propanoic-phenolic acid derivative such as Advil, Motrin, Nuprin, and Medipren
  • naproxen sodium such as Aleve, Anaprox, Antalgin, Feminax Ultra, Flanax, Inza, Midol Extended Relief, Nalgesin, Naposin, Naprelan, Naprogesic, Naprosyn, Naprosyn suspension, EC-Naprosyn, Narocin, Proxen, Synflex and Xenobid
  • acetic acid derivative such as indomethacin (Indocin),l-naphthaleneacetic acid derivative such as nabumetone or relafen
  • APAP N-acetyl-para-aminophenol
  • the antimuscarinic agents include: oxybutynin, solifenacin, darifenacin and atropine.
  • Macrophages are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
  • Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
  • Botulinum neurotoxin A at various doses in the presence of carbachol or
  • Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
  • Thromboxane IL- ⁇ , IL-6, TNF- ⁇ , the COX2 activity, the production of cAMP and cGMP, the production of IL- ⁇ , IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
  • Murine RAW264.7 or J774 macrophage cells were used in this study. Cells were maintained in a culture medium containing RPMI 1640 supplemented with 10 % fetal bovine serum (FBS), 15 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, and 100 ⁇ g / ml of streptomycin. Cells were cultured at 37° C in a 5 % C0 2 atmosphere and split (passages) once a week.
  • FBS fetal bovine serum
  • HEPES 15 fetal bovine serum
  • 2 mM L-glutamine 100 U/ml penicillin
  • streptomycin 100 ⁇ g / ml of streptomycin
  • RAW264.7 macrophage cells were seeded in 96-well plates at a cell density of 1.5xl0 5 cells per well in 100 ⁇ of the culture medium.
  • the cells were treated with (1) various concentrations of analgesic (acetaminophen, aspirin, ibuprophen or naproxen), (2) various concentrations of lipopolysaccharide (LPS), which is an effector of inflammatory stimuli to macrophage cells, (3) various concentrations of carbachol or acetylcholine, which are effectors of non-inflammatory stimuli, (4) analgesic and LPS or (5) analgesic and carbachol or acetylcholine.
  • analgesic acetaminophen, aspirin, ibuprophen or naproxen
  • LPS lipopolysaccharide
  • carbachol or acetylcholine which are effectors of non-inflammatory stimuli
  • analgesic and LPS or (5) analgesic and carbachol or
  • the analgesics were dissolved in FBS-free culture medium ⁇ i.e., RPMI 1640 supplemented with 15 mM HEPES, 2 mM L-glutamine, 100 U / ml penicillin, and 100 ⁇ g / ml of streptomycin), and diluted to desired concentrations by serial dilution with the same medium.
  • FBS-free culture medium ⁇ i.e., RPMI 1640 supplemented with 15 mM HEPES, 2 mM L-glutamine, 100 U / ml penicillin, and 100 ⁇ g / ml of streptomycin
  • 50 ⁇ of analgesic solution and 50 ⁇ of FBS-free culture medium were added to each well.
  • 50 ⁇ of analgesic solution and 50 ⁇ of LPS from Salmonella
  • FACS buffer phosphate buffered saline (PBS) with 2% bovine serum albumin (BSA) and 0.01% NaN 3
  • FACS buffer phosphate buffered saline
  • BSA bovine serum albumin
  • I-A d anti MHC class II
  • Immunoplates (Nunc) coated overnight with 100 ⁇ of anti-mouse IL-6, TNF-oc mAbs (BD Biosciences) or IL- ⁇ mAb (R&D Systems) in 0.1 M sodium bicarbonate buffer (pH 9.5).
  • COX2 activity in the cultured macrophages is determined by COX2 activity assay.
  • the production of cAMP and cGMP is determined by the cAMP assay and cGMP assay. These assays are performed routinely in the art.
  • Table 1 summarizes the experiments performed with Raw 264 macrophage cell line and main findings in terms of the effects of analgesics on cell surface expression of costimulatory molecules CD40 and CD80. Expression of these molecules is stimulated by COX2 and inflammatory signals and thus, was evaluated to determine functional consequences of inhibition of COX2.
  • acetaminophen, aspirin, ibuprophen and naproxen inhibit basal expression of co-stimulatory molecules CD40 and CD80 by macrophages at all the tested doses (i.e., 5x 10 5 nM, 5x 10 4 nM, 5x 10 3 nM, 5x 10 2 nM, 50 nM and 5 nM), except for the highest dose (i.e., 5x 10 6 nM), which appears to enhance, rather than inhibit, expression of the co-stimulatory molecules.
  • Table 3 summarizes the results of several studies that measured serum levels of analgesic after oral therapeutic doses in adult humans. As shown in Table 3, the maximum serum levels of analgesic after an oral therapeutic dose are in the range of 10 4 to 10 5 nM. Therefore, the doses of analgesic tested in vitro in Table 2 cover the range of concentrations achievable in vivo in humans.
  • EXAMPLE 3 EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON-INFLAMMATORY STIMULI
  • Example 2 This study is designed to characterize how the optimal doses of analgesic determined in Example 2 affect bladder smooth muscle cells in cell culture or tissue cultures, and to address whether different classes of analgesics can synergize to more efficiently inhibit COX2 and PGE2 responses.
  • Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
  • Botulinum neurotoxin A at various doses in the presence of carbachol or
  • Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
  • Thromboxane IL- ⁇ , IL-6, TNF-a, the COX2 activity, the production of cAMP and cGMP, the production of IL- ⁇ , IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
  • Bladder cells were removed from euthanized animals C57BL/6 mice (8-12 weeks old) and cells were isolated by enzymatic digestion followed by purification on a Percoll gradient. Briefly, bladders from 10 mice were minced with scissors to fine slurry in 10 ml of digestion buffer (RPMI 1640, 2% fetal bovine serum, 0.5 mg/ml collagenase, 30 ⁇ g/ml DNase). Bladder slurries were enzymatically digested for 30 minutes at 37°C. Undigested fragments were further dispersed through a cell-trainer. The cell suspension was pelleted and added to a discontinue 20%, 40% and 75% Percoll gradient for purification on mononuclear cells. Each experiment used 50-60 bladders.
  • digestion buffer RPMI 1640, 2% fetal bovine serum, 0.5 mg/ml collagenase, 30 ⁇ g/ml DNase.
  • Bladder slurries were enzymatically digested for 30
  • bladder cells were resuspended RPMI 1640 supplemented with 10 % fetal bovine serum, 15 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, and 100 ⁇ g / ml of streptomycin and seeded in clear-bottom black 96-well cell culture microculture plates at a cell density of 3xl0 4 cells per well in 100 ⁇ . Cells were cultured at 37° C in a 5 % C0 2 atmosphere.
  • Bladder cells were treated with analgesic solutions (50 ⁇ / well) either alone or together with carbachol (10-Molar, 50 ⁇ / well), as an example of non-inflammatory stimuli, or lipopolysaccharide (LPS) of Salmonella typhimurium (1 ⁇ g/ml, 50 ⁇ / well), as an example of non-inflammatory stimuli.
  • analgesic solutions 50 ⁇ / well
  • carbachol 10-Molar, 50 ⁇ / well
  • LPS lipopolysaccharide
  • Salmonella typhimurium 1 ⁇ g/ml, 50 ⁇ / well
  • COX2 responses were analyzed by a Cell-Based ELISA using Human/mouse total COX2 immunoassay (R&D Systems), following the instructions of the manufacturer. Briefly, after cells fixation and permeabilization, a mouse anti-total COX2 and a rabbit anti-total GAPDH were added to the wells of the clear-bottom black 96-well cell culture microculture plates. After incubation and washes, an HRP-conjugated anti-mouse IgG and an AP-conjugated anti-rabbit IgG were added to the wells. Following another incubation and set of washes, the HRP- and AP-fluorogenic substrates were added.
  • R&D Systems Human/mouse total COX2 immunoassay
  • Prostaglandin E2 responses were analyzed by a sequential competitive ELISA (R&D Systems). More specifically, culture supernatants or PGE2 standards were added to the wells of a 96-well polystyrene microplate coated with a goat anti-mouse polyclonal antibody. After one hour incubation on a microplate shaker, an HRP-conjugated PGE2 was added and plates incubated for an additional two hours at room temperature. The plates were then washed and HRP substrate solution added to each well. The color was allowed to develop for 30 min and the reaction stopped by the addition of sulfuric acid before reading the plate at 450 nm with wavelength correction at 570 nm. Results are expressed as mean pg/ml of PGE2.
  • Analgesics inhibit COX2 responses of murine bladder cells to an inflammatory stimuli
  • Analgesics inhibit PGE2 responses of murine bladder cells to an inflammatory stimuli
  • EXAMPLE 4 EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON BLADDER SMOOTH MUSCLE CELL CONTRACTION. Experimental Design
  • Botulinum neurotoxin A at various doses in the presence of carbachol or
  • Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
  • EXAMPLE 5 EFFECT OF ORAL ANALGESIC AGENTS AND ANTIMUSCARINIC AGENTS ON COX2 AND PGE2 RESPONSES OF BLADDER SMOOTH MUSCLE CELLS.
  • mice and mice with over active bladder syndrome are given oral doses of aspirin, naproxen sodium, Ibuprofen, Indocin, nabumetone, Tylenol, Celecoxib, oxybutynin, solifenacin, darifenacin, atropine and combinations thereof.
  • Control groups include untreated normal mice and untreated OAB mice without over active bladder syndrome.
  • the bladders are collected and stimulated ex vivo with carbachol or acetylcholine.
  • the bladders are treated with botulinum neurotoxin A before stimulation with carbachol. Animals are maintained in metabolic cages and frequency (and volume) of urination are evaluated.
  • Bladder outputs are determined by monitoring water intake and cage litter weight. Serum PGH 2 , PGE, PGE 2 , Prostacydin, Thromboxane, IL- ⁇ , IL-6, TNF-a, cAMP, and cGMP levels are determined by ELISA. CD80, CD86, MHC class II expression in whole blood cells are determined by flow cytometry.
  • EXAMPLE 6 EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON HUMAN BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON-INFLAMMATORY STIMULI Experimental Design
  • Human bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
  • Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
  • Botulinum neurotoxin A at various doses in the presence of carbachol or
  • Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
  • Thromboxane IL- ⁇ , IL-6, T F- ⁇ , the COX2 activity, the production of cAMP and cGMP, the production of IL- ⁇ , IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
  • EXAMPLE 7 EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTPMUSCARINIC AGENTS ON HUMAN BLADDER SMOOTH MUSCLE CELL CONTRACTION.
  • Human bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
  • Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
  • Botulinum neurotoxin A at various doses in the presence of carbachol or
  • Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
  • EXAMPLE 8 EFFECT OF ANALGESIC AGENTS ON NORMAL HUMAN BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON INFLAMMATORY SIGNALS EXPERIMENTAL DESIGN
  • Bladder smooth muscle cells trypsinized and seeded in microculture plates at a cell density of 3xl0 4 cells per well in 100 ⁇ were treated with analgesic solutions (50 ⁇ / well) either alone or together carbachol (10-Molar, 50 ⁇ / well), as an example of non-inflammatory stimuli, or lipopolysaccharide (LPS) of Salmonella typhimurium (1 ⁇ g/ml, 50 ⁇ / well), as an example of non-inflammatory stimuli.
  • analgesic solutions 50 ⁇ / well
  • carbachol (10-Molar, 50 ⁇ / well) as an example of non-inflammatory stimuli
  • LPS lipopolysaccharide
  • Salmonella typhimurium 1 ⁇ g/ml, 50 ⁇ / well
  • Analgesics inhibit COX2 responses of normal human bladder smooth muscle cells to inflammatory and non- inflammatory stimuli - Analysis of cells and culture supernatants after 24 hours of cultures showed that none of the analgesics tested alone induced COX2 responses in normal human bladder smooth muscle cells. However, as summarized in Table 6, carbachol induced low, but significant COX2 responses in normal human bladder smooth muscle cells. On the other hand, LPS treatment resulted in higher levels of COX2 responses in normal human bladder smooth muscle cells. Acetaminophen, aspirin, ibuprofen and naproxen could all suppress the effect of carbachol and LPS on COX2 levels. The suppressive effect of the analgesics was seen on LPS-induced responses when these drugs were tested at either 5 ⁇ or 50 ⁇ .
  • Analgesics inhibit PGE2 responses of normal human bladder smooth muscle cells to inflammatory and non- inflammatory stimuli - Consistent with the induction of COX2 responses described above, both carbachol and LPS induced production of PGE2 by normal human bladder smooth muscle cells. Acetaminophen, aspirin, ibuprofen and naproxen were also found to suppress the LPS-induced PGE2 responses at either 5 ⁇ or 50 ⁇ (Table 7). Table 7. PGE2 secretion by normal human bladder smooth muscle cells after in vitro stimulation with inflammatory and non- inflammatory stimuli and treatment with analgesic
  • Analgesics inhibit cytokine responses of normal human bladder cells to an inflammatory stimuli - Analysis of cells and culture supernatants after 24 hours of cultures showed that none the analgesics tested alone induced IL-6 or TNFa secretion in normal human bladder smooth muscle cells. As shown in Tables 8 and 9, the dose of carbachol tested induced low, but significant TNFa and IL-6 responses in normal human bladder smooth muscle cells. On the other hand, LPS treatment resulted in massive induction of these proinflammatory cytokines. Acetaminophen, aspirin, ibuprofen and naproxen suppress the effect of carbachol and LPS on TNFa and IL-6 responses.

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compositions for reducing the frequency of urination and methods of manufacture of the compositions are disclosed. One method of manufacture include the steps of forming a first mixture comprising a first active ingredient comprising one or more analgesic, coating the first mixture with a first delayed-release coating to form a first component, forming a second mixture comprising a second active ingredient comprising one or more analgesic agents, coating the second mixture with a second delayed-release coating to form a second component, and combining the first component with the second component to form a third mixture.

Description

TITLE
DELAYED-RELEASE FORMULATIONS, METHODS OF MAKING
AND USE THEREOF
[0001] This application claims priority from U.S. Application Serial No. 14/842,539, filed September 1, 2015. The entirety of the aforementioned application is incorporated herein by reference.
FIELD
[0002] The present application generally relates to methods and compositions for inhibiting the contraction of muscles and, in particular, to methods and compositions for inhibiting the contraction of smooth muscles of the urinary bladder.
BACKGROUND
[0003] The detrusor muscle is a layer of the urinary bladder wall made of smooth muscle fibers arranged in spiral, longitudinal, and circular bundles. When the bladder is stretched, this signals the parasympathetic nervous system to contract the detrusor muscle. This encourages the bladder to expel urine through the urethra.
[0004] For the urine to exit the bladder, both the autonomically controlled internal sphincter and the voluntarily controlled external sphincter must be opened. Problems with these muscles can lead to incontinence. If the amount of urine reaches 100% of the urinary bladder's absolute capacity, the voluntary sphincter becomes involuntary and the urine will be ejected instantly.
[0005] The human adult urinary bladder usually holds about 300-350 ml of urine (the working volume), but a full adult bladder may hold up to about 1000 ml (the absolute volume), varying among individuals. As urine accumulates, the ridges produced by folding of the wall of the bladder (rugae) flatten and the wall of the bladder thins as it stretches, allowing the bladder to store larger amounts of urine without a significant rise in internal pressure.
[0006] In most individuals, the desire to urinate usually starts when the volume of urine in the bladder reaches around 200 ml. At this stage it is easy for the subject, if desired, to resist the urge to urinate. As the bladder continues to fill, the desire to urinate becomes stronger and harder to ignore. Eventually, the bladder will fill to the point where the urge to urinate becomes overwhelming, and the subject will no longer be able to ignore it. In some individuals, this desire to urinate starts when the bladder is less than 100% full in relation to its working volume. Such increased desire to urinate may interfere with normal activities, including the ability to sleep for sufficient uninterrupted periods of rest. In some cases, this increased desire to urinate may be associated with medical conditions such as benign prostate hyperplasia or prostate cancer in men, or pregnancy in women. However, increased desire to urinate also occurs in individuals, both male and female, who are not affected by another medical condition.
[0007] Accordingly, there exists a need for compositions and methods for the treatment of male and female subjects who suffer from a desire to urinate when the bladder is less than 100% full of urine in relation to its working volume. Said compositions and methods are needed for the inhibition of muscle contraction in order to allow in said subjects the desire to urinate to start when the volume of urine in the bladder exceeds around 100% of its working volume.
SUMMARY
[0008] One aspect of the present application relates to a method for manufacturing a pharmaceutical composition for reducing the frequency of urination. The method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a first component, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after oral administration; forming a second mixture comprising a second active ingredient comprising one or more analgesic agents; coating the second mixture with a second delayed-release coating to form a second component, wherein the second delayed-release coating releases the second mixture with a lag time of 4-6 hours after oral administration; and combining the first component with the second component to form a third mixture.
[0009] In another embodiment, the method comprises the steps of forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after direct contact with a body fluid; coating the core structure with a second mixture to form a coated core structure, wherein the second mixture comprises a second active ingredient comprising one or more analgesic agents; and coating the coated core structure with a second delayed-release coating to form a double-coated core structure, wherein the second delayed-release coating releases the second mixture with a lag time of 1-4 hours after oral administration.
[0010] Another aspect of the present application relates to pharmaceutical compositions manufactured using the method of the present application.
BRIEF DESCRIPTION OF DRAWINGS
[0011] Figure 1 A and IB are diagrams showing that analgesics regulate expression of co-stimulatory molecules by Raw 264 macrophage cells in the absence (Figure 1 A) or presence (Figure IB) of LPS. Cells were cultures for 24 hrs in the presence of analgesic alone or together with Salmonella typhymurium LPS (0.05 μg/ml). Results are mean relative % of CD40+CD80+ cells. DETAILED DESCRIPTION
[0012] The following detailed description is presented to enable any person skilled in the art to make and use the invention. For purposes of explanation, specific nomenclature is set forth to provide a thorough understanding of the present invention. However, it will be apparent to one skilled in the art that these specific details are not required to practice the invention. Descriptions of specific applications are provided only as representative examples. The present invention is not intended to be limited to the embodiments shown, but is to be accorded the broadest possible scope consistent with the principles and features disclosed herein.
[0013] As used herein, the term "effective amount" means an amount necessary to achieve a selected result.
[0014] As used herein, the term "analgesic" refers to agents, compounds or drugs used to relieve pain and inclusive of anti-inflammatory compounds. Exemplary analgesic and/or anti-inflammatory agents, compounds or drugs include, but are not limited to, the following substances: non-steroidal anti-inflammatory drugs (NSAIDs), salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin; enolic acids, oxicam derivatives, piroxicam, meloxicam, tenoxicam, ampiroxicam, droxicam, pivoxicam, pyrazolon derivatives, phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, dipyrone, coxibs, celecoxib, rofecoxib, nabumetone, apazone, indomethacin, sulindac, etodolac, isobutylphenyl propionic acid, lumiracoxib, etoricoxib, parecoxib, valdecoxib, tiracoxib, etodolac, darbufelone, dexketoprofen, aceclofenac, licofelone, bromfenac, pranoprofen, loxoprofen, piroxicam, nimesulide, cizolirine,
3-formylamino-7-methylsulfonylamino-6-phenoxy-4H-l-benzopyran-4-one, meloxicam, lornoxicam, d-indobufen, mofezolac, amtolmetin, pranoprofen, tolfenamic acid, flurbiprofen, suprofen, oxaprozin, zaltoprofen, alminoprofen, tiaprofenic acid, pharmacological salts thereof, hydrates thereof, and solvates thereof.
[0015] As used herein, the terms "coxib" and "COX inhibitor" refer to a composition of compounds that is capable of inhibiting the activity or expression of COX2 enzymes or is capable of inhibiting or reducing the severity, including pain and swelling, of a severe inflammatory response.
[0016] The urinary bladder has two important functions: storage of urine and emptying. Storage of urine occurs at low pressure, which implies that the detrusor muscle relaxes during the filling phase. Emptying of the bladder requires a coordinated contraction of the detrusor muscle and relaxation of the sphincter muscles of the urethra. Disturbances of the storage function may result in lower urinary tract symptoms, such as urgency, frequency, and urge incontinence, the components of the overactive bladder syndrome. The overactive bladder syndrome, which may be due to involuntary contractions of the smooth muscle of the bladder (detrusor) during the storage phase, is a common and underreported problem, the prevalence of which has only recently been assessed.
[0017] One aspect of the present application relates to a method for reducing the frequency of urination by administering to a person in need thereof a pharmaceutical
composition formulated in a delayed-release formulation. The pharmaceutical composition comprises one or more analgesic agents and, optionally, one or more antimuscarinic agents.
[0018] As used herein, the term "delayed-release" refers to a medication that does not immediately disintegrate and release the active ingredient(s) into the body. In some embodiments, the term "delayed-release" is used with reference to a drug formulation having a release profile in which there is a predetermined delay in the release of the drug following administration. In some embodiments, the delayed-release formulation includes an enteric coating, which is a barrier applied to oral medication that prevents release of medication before it reaches the small intestine. Delayed-release formulations, such as enteric coatings, prevent drugs having an irritant effect on the stomach, such as aspirin, from dissolving in the stomach. Such coatings are also used to protect acid-unstable drugs from the stomach's acidic exposure, delivering them instead to a basic pH environment (intestine's pH 5.5 and above) where they do not degrade, and give their desired action.
[0019] The term "pulsatile release" is a type of delayed-release, which is used herein with reference to a drug formulation that provides rapid and transient release of the drug within a short time period immediately after a predetermined lag period, thereby producing a "pulsed" plasma profile of the drug after drug administration. Formulations may be designed to provide a single pulsatile release or multiple pulsatile releases at predetermined time intervals following administration.
[0020] Most enteric coatings work by presenting a surface that is stable at the highly acidic pH found in the stomach, but breaks down rapidly at a less acidic (relatively more basic) pH. Therefore, an enteric coated pill will not dissolve in the acidic juices of the stomach (pH ~3), but they will in the alkaline (pH 7-9) environment present in the small intestine. Examples of enteric coating materials include, but are not limited to, methyl aciylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), methyl methacrylate-methacrylic acid copolymers, sodium alginate and stearic acid.
[0021] In some embodiments, the pharmaceutical composition is orally administered from a variety of drug formulations designed to provide delayed-release. Delayed oral dosage forms include, for example, tablets, capsules, caplets, and may also comprise a plurality of granules, beads, powders or pellets that may or may not be encapsulated. Tablets and capsules represent the most convenient oral dosage forms, in which case solid pharmaceutical carriers are employed.
[0022] In a delayed-release formulation, one or more barrier coatings may be applied to pellets, tablets, or capsules to facilitate slow dissolution and concomitant release of drugs into the intestine. Typically, the barrier coating contains one or more polymers encasing, surrounding, or forming a layer, or membrane around the therapeutic composition or active core.
[0023] In some embodiments, the active agents are delivered in a formulation to provide delayed-release at a pre-determined time following administration. The delay may be up to about 10 minutes, about 20 minutes, about 30 minutes, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, or longer.
[0024] A delayed-release composition may comprise 100% of the total dosage of a given active agent administered in a single unit dose. Alternatively, a delayed-release composition may be included as a component in a combined release profile formulation may provide about 30-95%) of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation. For example, the immediate-release component may provide about 5-70%, or about 50%) of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation. In alternate embodiments, the delayed-release component provides about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% of the total dosage of the active agent(s) to be delivered by the formulation.
[0025] A delayed-release formulation typically comprises a barrier coating that delays the release of the active ingredient(s). The barrier coating may consist of a variety of different materials, depending on the objective. In addition, a formulation may comprise a plurality of barrier coatings to facilitate release in a temporal manner. The coating may be a sugar coating, a film coating (e.g., based on hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone), or a coating based on methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and/or ethylcellulose. Furthermore, the formulation may additionally include a time delay material such as, for example, glyceryl monostearate or glyceryl distearate. [0026] In some embodiments, the delayed-release formulation includes an enteric coating comprised one or more polymers facilitating release of active agents in proximal or distal regions of the gastrointestinal tract. As used herein, the term "enteric polymer coating" a coating comprising of one or more polymers having a pH dependent or pH-independent release profile. Typically the coating resists dissolution in the acidic medium of the stomach, but dissolves or erodes in more distal regions of the gastrointestinal tract, such as the small intestine or colon. An enteric polymer coating typically resists releases of the active agents until sometime after a gastric emptying lag period of about 3-4 hours after administration.
[0027] pH dependent enteric coatings comprising one or more pH-dependent or pH-sensitive polymers that maintain their structural integrity at low pH, as in the stomach, but dissolve in higher pH environments in more distal regions of the gastrointestinal tract, such as the small intestine, where the drug contents are released. For purposes of the present invention, "pH dependent" is defined as having characteristics (e.g., dissolution) which vary according to environmental pH. Exemplary pH-dependent polymers include, but are not limited to, methacarylic acid copolymers, methacrylic acid-methyl methacrylate copolymers (e.g.,
EUDRAGIT® LI 00 (Type A), EUDRAGIT® S I 00 (Type B), Rohm GmbH, Germany;
methacrylic acid-ethyl acrylate copolymers (e.g., EUDRAGIT® LI 00-55 (Type C) and
EUDRAGIT® L30D-55 copolymer dispersion, Rohm GmbH, Germany); copolymers of methacrylic acid-methyl methacrylate and methyl methacrylate (EUDRAGIT® FS); terpolymers of methacrylic acid, methacrylate, and ethyl acrylate; cellulose acetate phthalates (CAP);
hydroxypropyl methylcellulose phthalate (HPMCP) (e.g., HP-55, HP-50, HP-55S, Shinetsu Chemical, Japan); polyvinyl acetate phthalates (PVAP) (e.g., COATERIC®, OPADRY® enteric white OY-P-7171); cellulose acetate succinates (CAS); hydroxypropyl methylcellulose acetate succinate (HPMCAS), e.g., HPMCAS LF Grade, MF Grade, HF Grade, including AQOAT® LF and AQOAT® MF (Shin-Etsu Chemical, Japan); Shinetsu Chemical, Japan); shellac (e.g., Marcoat™ 125 & Marcoat™ 125N); carboxymethyl ethylcellulose (CMEC, Freund Corporation, Japan), cellulose acetate phthalates (CAP)(e.g., AQUATERIC®); cellulose acetate trimellitates (CAT); and mixtures of two or more thereof at weight ratios between about 2: 1 to about 5 : 1, such as, for instance, a mixture of EUDRAGIT® L 100-55 and EUDRAGIT® S 100 at a weight ratio of about 3 : 1 to about 2: 1, or a mixture of EUDRAGIT® L 30 D-55 and EUDRAGIT® FS at a weight ratio of about 3 : 1 to about 5 : 1.
[0028] pH-dependent polymers typically exhibit a characteristic pH optimum for dissolution. In some embodiments, the pH-dependent polymer exhibits a pH optimum between about 5.0 and 5.5, between about 5.5 and 6.0, between about 6.0 and 6.5, or between about 6.5 and 7.0. In other embodiments, the pH-dependent polymer exhibits a pH optimum of >5.0, of >5.5, of >6.0, of >6.5, or of >7.0.
[0029] In other embodiments, the enteric coating may comprise one or more
pH-independent polymers. These polymers provide for release of the drug after a certain time, independent of the pH. For purposes of the present invention, "pH independent" is defined as having characteristics (e.g., dissolution) which are substantially unaffected by pH. pH independent polymers are often referred to in the context of "time-controlled" or
"time-dependent" release profiles.
[0030] A pH independent polymer may be water-insoluble or water soluble.
Exemplary water insoluble pH independent polymers include, but are not limited to, neutral methacrylic acid esters with a small portion of trimethylammonioethyl methacrylate chloride (e.g., EUDRAGIT® RS and EUDRAGIT® RL; neutral ester dispersions without any functional groups (e.g., EUDRAGIT® E30D and EUDRAGIT® E30); cellulosic polymers, such as ethylcellulose, hydroxyl ethyl cellulose, cellulose acetate or mixtures and other pH independent coating products. Exemplary water soluble pH independent polymers include OPADRY®amb.
[0031] In some embodiments, the pH independent polymers contain one or more polysaccharides that are resistant to erosion in both the stomach and intestine. Such polymers can be only degraded in the colon, which contains a large microflora containing biodegradable enzymes breaking down, for example, the polysaccharide coatings to release the drug contents in a controlled, time-dependent manner.
[0032] In certain embodiment, the coating methodology employs the blending of one or more pH-dependent and one or more pH-independent polymers. The blending of
pH-dependent and pH-independent polymers can reduce the release rate of active ingredients once the soluble polymer has reached its optimum pH of solubilization.
[0033] In some embodiments, a "time-controlled" or "time-dependent" release profile can be obtained using a water insoluble capsule body containing one or more active agents, wherein the capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug. Upon contact with gastrointestinal fluid or dissolution medium, the plug swells, pushing itself out of the capsule and releasing the drugs after a pre-determined lag time, which can be controlled by e.g., the position and dimensions of the plug. The capsule body may be further coated with an outer pH-dependent enteric coating keeping the capsule intact until it reaches the small intestine. Suitable plug materials include, for example, polymethacrylates, erodible compressed polymers (e.g., HPMC, polyvinyl alcohol), congealed melted polymer (e.g., glyceryl mono oleate) and enzymatically controlled erodible polymers (e.g., polysaccharides, such as amylose, arabinogalactan, chitosan, chondroitin sulfate, cyclodextrin, dextran, guar gum, pectin and xylan). [0034] In other embodiments, capsules or bilayered tablets may be formulated to contain a drug-containing core, covered by a swelling layer, and an outer insoluble, but semi-permeable polymer coating or membrane. The lag time prior to rupture can be controlled by the permeation and mechanical properties of the polymer coating and the swelling behavior of the swelling layer. Typically, the swelling layer comprises one or more swelling agents, such as swellable hydrophilic polymers that swell and retain water in their structures.
[0035] Exemplary water swellable materials include, but are not limited to, polyethylene oxide (having e.g., an average molecular weight between 1,000,000 to 7,000,000, such as POLYOX®), methylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose;
polyalkylene oxides having a weight average molecular weight of 100,000 to 6,000,000, including but not limited to poly(methylene oxide), poly(butylene oxide); poly(hydroxy alkyl methacrylate) having a molecular weight of from 25,000 to 5,000,000; poly(vinyl)alcohol, having a low acetal residue, which is cross-linked with glyoxal, formaldehyde or glutaraldehyde and having a degree of polymerization of from 200 to 30,000; mixtures of methyl cellulose, cross-linked agar and carboxymethyl cellulose; hydrogel forming copolymers produced by forming a dispersion of a finely divided copolymer of maleic anhydride with styrene, ethylene, propylene, butylene or isobutylene cross-linked with from 0.001 to 0.5 moles of saturated cross-linking agent per mole of maleic anyhydride in the copolymer; CARBOPOL® acidic carboxy polymers having a molecular weight of 450,000 to 4,000,000; CYANAMER® polyacrylamides; cross-linked water swellable indenemaleicanhydride polymers; GOODRITE® polyacrylic acid having a molecular weight of 80,000 to 200,000; starch graft copolymers;
AQUA-KEEPS® acrylate polymer polysaccharides composed of condensed glucose units such as diester cross-linked polyglucan; carbomers having a viscosity of 3,000 to 60,000 mPa as a 0.5%-l% w/v aqueous solution; cellulose ethers such as hydroxypropylcellulose having a viscosity of about 1000-7000 mPa s as a 1% w/w aqueous solution (25° C); hydroxypropyl methylcellulose having a viscosity of about 1000 or higher, preferably 2,500 or higher to a maximum of 25,000 mPa as a 2% w/v aqueous solution; polyvinylpyrrolidone having a viscosity of about 300-700 mPa s as a 10% w/v aqueous solution at 20° C; and combinations thereof.
[0036] The enteric layer may further comprise anti-tackiness agents, such as talc or glyceryl monostearate and/or plasticizers such as. The enteric layer may further comprise one or more plasticizers including, but not limited to, triethyl citrate, acetyl triethyl citrate, acetyltributyl citrate, polyethylene glycol acetylated monoglycerides, glycerin, triacetin, propylene glycol, phthalate esters (e.g., diethyl phthalate, dibutyl phthalate), titanium dioxide, ferric oxides, castor oil, sorbitol and dibutyl sebacate.
[0037] In another embodiment, the delay release formulation employs a water-permeable but insoluble film coating to enclose the active ingredient and an osmotic agent utilizing a enclosing. As water from the gut slowly diffuses through the film into the core, the core swells until the film bursts, thereby releasing the active ingredients. The film coating may be adjusted to permit various rates of water permeation or release time.
[0038] Alternatively, the release time of the drugs can be controlled by a disintegration lag time depending on the balance between the tolerability and thickness of a water insoluble polymer membrane (such as ethyl cellulose, EC) containing predefined micropores at the bottom of the body and the amount of a swellable excipient, such as low substituted hydroxypropyl cellulose (L-HPC) and sodium glycolate. After oral administration, GI fluids permeate through the micropores, causing swelling of the swellable excipients, which produces an inner pressure disengaging the capsular components, including a first capsule body containing the swellable materials, a second capsule body containing the drugs, and an outer cap attached to the first capsule body.
[0039] In other embodiments, the drugs may be released by an osmotic mechanism. By way of example, a capsule may be formulated with a single osmotic unit or it may incorporate 2, 3, 4, 5, or 6 push-pull units encapsulated within a hard gelatin capsule, whereby each bilayer push pull unit contains an osmotic push layer and a drug layer, both surrounded by a
semi-permeable membrane. One or more orifices are drilled through the membrane next to the drug layer. This membrane may be additionally covered with a pH-dependent enteric coating to prevent release until after gastric emptying. The gelatin capsule dissolves immediately after ingestion. As the push pull unit(s) enter the small intestine, the enteric coating breaks down, which then allows fluid to flow through the semi-permeable membrane, swelling the osmotic push compartment to force drugs out through the orifice(s) at a rate precisely controlled by the rate of water transport through the semi-permeable membrane. Release of drugs can occur over a constant rate for up to 24 hours or more.
[0040] The osmotic push layer comprises one or more osmotic agents creating the driving force for transport of water through the semi-permeable membrane into the core of the delivery vehicle. One class of osmotic agents includes water-swellable hydrophilic polymers, also referred to as "osmopolymers" and "hydrogels," including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.
[0041] Another class of osmotic agents includes osmogens, which are capable of imbibing water to effect an osmotic pressure gradient across the semi-permeable membrane. Exemplary osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-toluenesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.
[0042] Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are
water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking.
[0043] In another embodiment, the delay release formulation employs a
water-impermeable tablet coating whereby water enters through a controlled aperture in the coating until the core bursts. When the tablet bursts, the drug contents are released
immediately or over a longer period of time. These and other techniques may be modified to allow for a pre-determined lag period before release of drugs is initiated.
[0044] Various coating techniques may be applied to granules, beads, powders or pellets, tablets, capsules or combinations thereof containing active agents to produce different and distinct release profiles. In some embodiments, the pharmaceutical composition is in a tablet or capsule form containing a single coating layer. In other embodiments, the pharmaceutical composition is in a tablet or capsule form containing multiple coating layers.
[0045] In some embodiments, the pharmaceutical composition comprises a plurality of active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics. Examples of spasmolytics include, but are not limited to, carisoprodol, benzodiazepines, baclofen, cyclobenzaprine, metaxalone, methocarbamol, clonidine, clonidine analog, and dantrolene. In some embodiments, the pharmaceutical composition comprises one or more analgesics. In other embodiments, the pharmaceutical composition comprises (1) one or more analgesics, and (2) one or more other active ingredients selected from the group consisting of antimuscarinic agents, antidiuretics and spasmolytics. In another embodiment, the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two antimuscarinic agents. In another embodiment, the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two antidiuretics. In another embodiment, the pharmaceutical composition comprises (1) one or two analgesics and (2) one or two spasmolytics. In yet another embodiment, the pharmaceutical composition comprises (1) one or two analgesics, (2) one or two antimuscarinic agents, and (3) one or two antidiuretics.
[0046] In one embodiment, the plurality of active ingredients are formulated for immediate-release. In other embodiment, the plurality of active ingredients are formulated for delayed-release. In other embodiment, the plurality of active ingredients are formulated for both immediate-release and delayed-release (e.g., a first portion of each active ingredient is formulated for immediate-release and a second portion of each active ingredient is formulated for delayed-release). In yet other embodiment, some of the plurality of active ingredients are formulated for immediate-release and some of the plurality of active ingredients are formulated for delayed-release (e.g., active ingredients A, B, C are formulated for immediate-release and active ingredients C and D are formulated for delayed-release).
[0047] In certain embodiments, the pharmaceutical composition comprises an immediate-release component and a delayed-release component. The immediate-release component may comprise one or more active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.. The delayed-release component may comprise one or more active ingredients selected from the group consisting of analgesics, antimuscarinic agents, antidiuretics and spasmolytics.. In some embodiments, the immediate-release component and the delayed-release component have exactly the same active ingredients. In other embodiments, the immediate-release component and the delayed-release component have different active ingredients. In yet other embodiments, the immediate-release component and the delayed-release component have one or more common active ingredients.
[0048] In one embodiment, the pharmaceutical composition comprises two active ingredients (e.g., two analgesic agents, or a mixture of one analgesic agent and one
antimuscarinic agent or antidiuretic or spasmolytic), formulated for immediate-release at about the same time. In another embodiment, the pharmaceutical composition comprises two active ingredients, formulated for delayed-release at about the same time. In another embodiment, the pharmaceutical composition comprises two active ingredients formulated as two delayed-release components, each providing a different delayed-release profile. For example, a first delayed-release component releases a first active ingredient at a first time point and a second delayed-release component releases a second active ingredient at a second time point. In another embodiment, the pharmaceutical composition comprises two active ingredients, one is formulated for immediate-release and the other is formulated for delayed-release.
[0049] In other embodiments, the pharmaceutical composition comprises two active ingredients (e.g., two analgesic agents, or a mixture of one analgesic agent and one
antimuscarinic agent or antidiuretic or spasmolytic) formulated for immediate-release, and (2) two active ingredients (e.g., two analgesic agents, or a mixture of one analgesic agent and one antimuscarinic agent or antidiuretic or spasmolytic) formulated for delayed-release. In other embodiments, the pharmaceutical composition comprises three active ingredients formulated for immediate-release, and (2) three active ingredients formulated for delayed-release. In other embodiments, the pharmaceutical composition comprises four active ingredients formulated for immediate-release, and (2) four active ingredients formulated for delayed-release. In these embodiments, the active ingredient(s) in the immediate-release component can be the same as, or different from, the active ingredient(s) in the delayed-release component.
[0050] The term "immediate-release" is used herein with reference to a drug formulation that does not contain a dissolution rate controlling material. There is substantially no delay in the release of the active agents following administration of an immediate-release formulation. An immediate-release coating may include suitable materials immediately dissolving following administration so as to release the drug contents therein. Exemplary immediate-release coating materials include gelatin, polyvinyl alcohol polyethylene glycol (PVA-PEG) copolymers (e.g., KOLLICOAT®) and various others materials known to those skilled in the art.
[0051] An immediate-release composition may comprise 100% of the total dosage of a given active agent administered in a single unit dose. Alternatively, an immediate-release component may be included as a component in a combined release profile formulation that may provide about 1% to about 50% of the total dosage of the active agent(s) to be delivered by the pharmaceutical formulation. For example, the immediate-release component may provide at least about 5%, or about 10% to about 30%, or about 45% to about 50% of the total dosage of the active agent(s) to be delivered by the formulation. The rest of the active agent(s) may be delivered in a delayed-release formulation. In alternate embodiments, the immediate-release component provides about 10, 15, 20, 25, 30, 35, 40, 45 or 50% of the total dosage of the active agent(s) to be delivered by the formulation. The delayed-release component provides about 90, 85, 80, 75, 70, 65, 60, 55 or 50% of the total dosage of the active agent(s) to be delivered by the formulation.
[0052] In some embodiments, the immediate-release or delayed-release formulation comprises an active core comprised of one or more inert particles, each in the form of a bead, pellet, pill, granular particle, microcapsule, microsphere, microgranule, nanocapsule, or nanosphere coated on its surfaces with drugs in the form of e.g. , a drug-containing film-forming composition using, for example, fluid bed techniques or other methodologies known to those of skill in the art. The inert particle can be of various sizes, so long as it is large enough to remain poorly dissolved. Alternatively, the active core may be prepared by granulating and milling and/or by extrusion and spheronization of a polymer composition containing the drug substance.
[0053] The amount of drug in the core will depend on the dose that is required, and typically varies from about 5 to 90 weight %. Generally, the polymeric coating on the active core will be from about 1 to 50% based on the weight of the coated particle, depending on the lag time and type of release profile required and/or the polymers and coating solvents chosen. Those skilled in the art will be able to select an appropriate amount of drug for coating onto or incorporating into the core to achieve the desired dosage. In one embodiment, the inactive core may be a sugar sphere or a buffer crystal or an encapsulated buffer crystal such as calcium carbonate, sodium bicarbonate, fumaric acid, tartaric acid, etc., which alters the
microenvironment of the drug to facilitate its release.
[0054] In some embodiments, the delayed-release formulation is formed by coating a water soluble/dispersible drug-containing particle, such as a bead, with a mixture of a water insoluble polymer and an enteric polymer, wherein the water insoluble polymer and the enteric polymer may be present at a weight ratio of from 4: 1 to 1 : 1, and the total weight of the coatings is 10 to 60 weight % based on the total weight of the coated beads. The drug layered beads may optionally include an inner dissolution rate controlling membrane of ethylcellulose. The composition of the outer layer, as well as the individual weights of the inner and outer layers of the polymeric membrane are optimized for achieving desired circadian rhythm release profiles for a given active, which are predicted based on in vitro/in vivo correlations.
[0055] In other embodiments the formulations comprise a mixture of immediate-release drug-containing particles without a dissolution rate controlling polymer membrane and delayed-release beads exhibiting, for example, a lag time of 2-4 hours following oral
administration, thus providing a two-pulse release profile. In yet other embodiments the formulations comprise a mixture of two types of delayed-release beads: a first type that exhibits a lag time of 1-3 hours and a second type that exhibits a lag time of 4-6 hours.
[0056] In some embodiments, the active core is coated with one or more layers of dissolution rate-controlling polymers to obtain desired release profiles with or without a lag time. An inner layer membrane can largely control the rate of drug release following imbibition of water or body fluids into the core, while the outer layer membrane can provide for a desired lag time (the period of no or little drug release following imbibition of water or body fluids into the core). The inner layer membrane may comprise a water insoluble polymer, or a mixture of water insoluble and water soluble polymers. [0057] The polymers suitable for the outer membrane, which largely controls the lag time of up to 6 hours may comprise an enteric polymer, as described above, and a water insoluble polymer at 10 to 50 weight %. The ratio of water insoluble polymer to enteric polymer may vary from 4: 1 to 1 :2, preferably the polymers are present at a ratio of about 1 : 1. The water insoluble polymer typically used is ethylcellulose.
[0058] Exemplary water insoluble polymers include ethylcellulose, polyvinyl acetate (Kollicoat SR#0D from BASF), neutral copolymers based on ethyl acrylate and
methylmethacrylate, copolymers of acrylic and methacrylic acid esters with quaternary ammonium groups such as EUDRAGIT® E, RS and RS30D, RL or RL30D and the like.
Exemplary water soluble polymers include low molecular weight HPMC, HPC, methylcellulose, polyethylene glycol (PEG of molecular weight>3000) at a thickness ranging from 1 weight % up to 10 weight % depending on the solubility of the active in water and the solvent or latex suspension based coating formulation used. The water insoluble polymer to water soluble polymer may typically vary from 95:5 to 60:40, preferably from 80:20 to 65:35.
[0059] Preferably, the formulations are designed with release profiles to limit interference with restful sleep, wherein the formulation releases the medicine when the individual would normally be awakened by an urge to urinate. For example, consider an individual who begins sleeping at 11 PM and is normally awakened at 12:30 AM, 3 :00 AM, and 6:00 AM to urinate. A delayed-release vehicle could deliver the medicine at 12: 15 AM, thereby delaying the need to urinate for perhaps 2-3 hours.
[0060] The pharmaceutical composition may be administered daily or administered on an as needed basis. In certain embodiments, the pharmaceutical composition is administered to the subject prior to bedtime. In some embodiments, the pharmaceutical composition is administered immediately before bedtime. In some embodiments, the pharmaceutical composition is administered within about two hours before bedtime, preferably within about one hour before bedtime. In another embodiment, the pharmaceutical composition is administered about two hours before bedtime. In a further embodiment, the pharmaceutical composition is administered at least two hours before bedtime. In another embodiment, the pharmaceutical composition is administered about one hour before bedtime. In a further embodiment, the pharmaceutical composition is administered at least one hour before bedtime. In a still further embodiment, the pharmaceutical composition is administered less than one hour before bedtime. In still another embodiment, the pharmaceutical composition is administered immediately before bedtime. Preferably, the pharmaceutical composition is administered orally.
[0061] The appropriate dosage ("therapeutically effective amount") of the active agent(s) in the immediate-component or delayed-release component will depend, for example, the severity and course of the condition, the mode of administration, the bioavailability of the particular agent(s), the age and weight of the patient, the patient's clinical history and response to the active agent(s), discretion of the physician, etc.
[0062] As a general proposition, the therapeutically effective amount of the active agent(s) in the immediate-release component or the delayed-release component is administered in the range of about 100 μg/kg body weight/day to about 100 mg/kg body weight/day whether by one or more administrations. In some embodiments, the range of each active agent administered daily is from about 100 μg/kg body weight/day to about 50 mg/kg body weight/day, 100 μg/kg body weight/day to about 10 mg/kg body weight/day, 100 μg/kg body weight/day to about 1 mg/kg body weight/day, 100 μg/kg body weight/day to about 10 mg/kg body weight/day, 500 μg/kg body weight/day to about 100 mg/kg body weight/day, 500 μg/kg body weight/day to about 50 mg/kg body weight/day, 500 μg/kg body weight/ day to about 5 mg/kg body weight/ day, 1 mg/kg body weight/day to about 100 mg/kg body weight/day, 1 mg/kg body weight/day to about 50 mg/kg body weight/ day, 1 mg/kg body weight/day to about 10 mg/kg body weight/day, 5 mg/kg body weight/dose to about 100 mg/kg body weight/day, 5 mg/kg body weight/dose to about 50 mg/kg body weight/day, 10 mg/kg body weight/day to about 100 mg/kg body weight/day, and 10 mg/kg body weight/day to about 50 mg/kg body weight/day.
[0063] The active agent(s) described herein may be included in an immediate-release component or a delayed-release component or combinations thereof for daily oral administration at a single dose or combined dose range of 1 mg to 2000 mg, 5 mg to 2000 mg, 10 mg to 2000 mg, 50 mg to 2000 mg, 100 mg to 2000 mg, 200 mg to 2000 mg, 500 mg to 2000 mg, 5 mg to 1800 mg, 10 mg to 1600 mg, 50 mg to 1600 mg, 100 mg to 1500 mg, 150 mg to 1200 mg, 200 mg to 1000 mg, 300 mg to 800 mg, 325 mg to 500 mg, 1 mg to 1000 mg, 1 mg to 500 mg, 1 mg to 200 mg, 5 mg to 1000 mg, 5 mg to 500 mg, 5 mg to 200 mg, 10 mg to 1000 mg, 10 mg to 500 mg, 10 mg to 200 mg, 50 mg to 1000 mg, 50 mg to 500 mg, 50 mg to 200 mg, 100 mg to 250 mg, 100 mg to 500 mg, 250 mg to 1000 mg, 250 mg to 500 mg, 500 mg to 1000 mg, 500 mg to 2000 mg. As expected, the dosage will be dependent on the condition, size, age and condition of the patient.
[0064] In some embodiments, the pharmaceutical composition comprises a single analgesic agent. In one embodiment, the single analgesic agent is aspirin. In another embodiment, the single analgesic agent is ibuprofen. In another embodiment, the single analgesic agent is naproxen sodium. In another embodiment, the single analgesic agent is indomethacin. In another embodiment, the single analgesic agent is nabumetone. In another embodiment, the single analgesic agent is acetaminophen.
[0065] In some embodiments, the single analgesic agent is given at a daily dose of 1 mg to 2000 mg, 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg. In certain embodiments, the pharmaceutical composition comprises acetylsalicylic acid, ibuprofen, naproxen sodium, indomethancin, nabumetone or acetaminophen as a single analgesic agent and the analgesic agent is administered orally at a daily dose in the range of 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg. In some embodiments, a second analgesic agent is given at a daily dose of 1 mg to 2000 mg, 5 mg to 2000 mg, 20 mg to 2000 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 50 mg to 500 mg, 100 mg to 500 mg, 250 mg to 500 mg, 250 mg to 1000 mg or 500 mg to 1000 mg.
[0066] In other embodiments, the pharmaceutical composition comprises a pair of analgesic agents. Examples of such paired analgesic agents include, but are not limited to, acetylsalicylic acid and ibuprofen, acetylsalicylic acid and naproxen sodium, acetylsalicylic acid and nabumetone, acetylsalicylic acid and acetaminophen, acetylsalicylic acid and indomethancin, ibuprofen and naproxen sodium, ibuprofen and nabumetone, ibuprofen and acetaminophen, ibuprofen and indomethancin, naproxen sodium and nabumetone, naproxen sodium and acetaminophen, naproxen sodium and indomethancin, nabumetone and acetaminophen, nabumetone and indomethancin, and acetaminophen and indomethancin. The paired analgesic agents are mixed at a weight ratio in the range of 0.1 : 1 to 10: 1, 0.2: 1 to 5: 1 or 0.3 : 1 to 3 : 1, with a combined dose in the range of 5 mg to 2000 mg, 20 mg to 2000 mg, 100 mg to 2000 mg, 200 mg to 2000 mg, 500 mg to 2000 mg, 5 mg to 1500 mg, 20 mg to 1500 mg, 100 mg to 1500 mg, 200 mg to 1500 mg, 500 mg to 1500 mg, 5 mg to 1000 mg, 20 mg to 1000 mg, 100 mg to 1000 mg, 250 mg to 500 mg, 250 mg to 1000 mg, 250 mg to 1500 mg, 500 mg to 1000 mg, 500 mg to 1500 mg, 1000 mg to 1500 mg, and 1000 mg to 2000 mg. In one embodiment, the paired analgesic agents are mixed at a weight ratio of 1 : 1.
[0067] In some other embodiments, the pharmaceutical composition of the present application further comprises one or more antimuscarinic agents. Examples of the
antimuscarinic agents include, but are not limited to, oxybutynin, solifenacin, darifenacin, fesoterodine, tolterodine, trospium and atropine. The daily dose of antimuscarinic agent is in the range of 0.01 mg to 100 mg, 0.1 mg to 100 mg, 1 mg to 100 mg, 10 mg to 100 mg, 0.01 mg to 25 mg, 0.1 mg to 25 mg, 1 mg to 25 mg, 10 mg to 25 mg, 0.01 mg to 10 mg, 0.1 mg to 10 mg, 1 mg to 10 mg, 10 mg to 100 mg and 10 mg to 25 mg.
[0068] Another aspect of the present application relates to a method for reducing the frequency of urination by administering to a person in need thereof a pharmaceutical
composition formulated in an immediate-release formulation. In some embodiments, the pharmaceutical composition comprises one or more analgesic agents and one or more
antimuscarinic agents. In other embodiments, the pharmaceutical composition comprises one or more analgesic agents and one or more antidiuretic agents. In yet other embodiments, the pharmaceutical composition comprises one or more analgesic agents, one or more antimuscarinic agents, and one or more antidiuretic agents.
[0069] In other embodiments, the pharmaceutical composition of the present application further comprises one or more spasmolytics. Examples of spasmolytics include, but are not limited to, carisoprodol, benzodiazepines, baclofen, cyclobenzaprine, metaxalone,
methocarbamol, clonidine, clonidine analog, and dantrolene. In some embodiments, the spasmolytics is used at a daily dose of 1 mg to 1000 mg, 1 mg to 100 mg, 10 mg to 1000 mg, 10 mg to 100 mg, 20 mg to 1000 mg, 20 mg to 800 mg, 20 mg to 500 mg, 20 mg to 200 mg, 50 mg to 1000 mg, 50 mg to 800 mg, 50 mg to 200 mg, 100 mg to 800 mg, 100 mg to 500 mg, 200 mg to 800 mg, and 200 mg to 500 mg. The spasmolytics may be formulated, alone or together with other active ingredient(s) in the pharmaceutical composition, for immediate-release, delayed-release or combinations thereof.
[0070] In certain embodiments, the pharmaceutical composition comprises two or more analgesic agents. In other embodiments, the pharmaceutical composition comprises one or more analgesic agents and one or more antimuscarinic agents and/or antidiuretic agents. The pharmaceutical composition may be formulated into a tablet, capsule, dragee, powder, granulate, liquid, gel or emulsion form. Said liquid, gel or emulsion may be ingested by the subject in naked form or contained within a capsule.
[0071] In certain embodiments, the analgesic agent is selected from the group consisting of salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin; enolic acids, oxicam derivatives, piroxicam, meloxicam, tenoxicam, ampiroxicam, droxicam, pivoxicam, pyrazolon derivatives, phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, dipyrone, coxibs, celecoxib, rofecoxib, nabumetone, apazone, nimesulide, indomethacin, sulindac, etodolac, and
isobutylphenyl propionic acid. The antimuscarinic agent is selected from the group consisting of oxybutynin, solifenacin, darifenacin and atropine.
[0072] In some embodiments, the pharmaceutical composition comprises a single analgesic agent and a single antimuscarinic agent. In one embodiment, the single analgesic agent is aspirin. In another embodiment, the single analgesic agent is ibuprofen. In another embodiment, the single analgesic agent is naproxen sodium. In another embodiment, the single analgesic agent is indomethacin. In another embodiment, the single analgesic agent is nabumetone. In another embodiment, the single analgesic agent is acetaminophen. The analgesic agent and anti-muscarinic agent may be given at doses in the ranges described above.
[0073] Another aspect of the present application relates to a method for treating nocturia by administering to a subject in need thereof (1) one or more analgesic agents and (2) one or more antidiuretic agents. In certain embodiments, the antidiuretic agent(s) act to: (1) increase vasopressin secretion; (2) increase vasopressin receptor activation; (3) reduce secretion of atrial natriuretic peptide (ANP) or C-type natriuretic peptide (C P); or (4) reduce A P and/or C P receptor activation.
[0074] Exemplary antidiuretic agents include, but are not limited to, antidiuretic hormone (ADH), angiotensin II, aldosterone, vasopressin, vasopressin analogs (e.g.,
desmopressin argipressin, lypressin, felypressin, ornipressin, terlipressin); vasopressin receptor agonists, atrial natriuretic peptide (ANP) and C-type natriuretic peptide (CNP) receptor (i.e., NPR1, NPR2, NPR3) antagonists (e.g., HS-142-1, isatin, [Asu7,23']b-ANP-(7-28)], anantin, a cyclic peptide from Streptomyces coerulescens, and 3G12 monoclonal antibody); somatostatin type 2 receptor antagonists (e.g., somatostatin), and pharmaceutically-acceptable derivatives, analogs, salts, hydrates, and solvates thereof.
[0075] In certain embodiments, the one or more analgesic agent and one or more antidiuretic agents are formulated for delayed-release.
[0076] Another aspect of the present application relates to a method for treating nocturia by administering to a person in need thereof a first pharmaceutical composition comprising a diuretic, followed with a second pharmaceutical composition comprising one or more analgesic agents. The first pharmaceutical composition is dosed and formulated to have a diuretic effect within 6 hours of administration and is administered at least 8 hours prior to bedtime. The second pharmaceutical composition is administered within 2 hours prior to bedtime.
[0077] Examples of diuretics include, but are not limited to, acidifying salts, such as CaCl2 and NH4C1; arginine vasopressin receptor 2 antagonists, such as amphotericin B and lithium citrate; aquaretics, such as Goldenrod and Junipe; Na-H exchanger antagonists, such as dopamine; carbonic anhydrase inhibitors, such as acetazolamide and dorzolamide; loop diuretics, such as bumetanide, ethacrynic acid, furosemide and torsemide; osmotic diuretics, such as glucose and mannitol; potassium-sparing diuretics, such as amiloride, spironolactone,
triamterene, potassium canrenoate; thiazides, such as bendroflumethiazide and
hydrochlorothiazide; and xanthines, such as caffeine, theophylline and theobromine.
[0078] In some embodiments, the second pharmaceutical composition further comprises one or more antimuscarinic agents. Examples of the antimuscarinic agents include, but are not limited to, oxybutynin, solifenacin, darifenacin, fesoterodine, tolterodine, trospium and atropine. The second pharmaceutical composition may be formulated in immediate-release formulation or delayed-release formulation. In one embodiment, the first pharmaceutical composition is formulated for immediate-release and the second pharmaceutical composition is formulated for delayed-release.
[0079] In some other embodiments, the second pharmaceutical composition further comprises one or more antidiuretic agents.
[0080] Another aspect of the present application relates to a pharmaceutical composition comprising a plurality of active ingredients and a pharmaceutically acceptable carrier. In some embodiments, the plurality of active ingredients comprise two or more analgesics. In other embodiments, the plurality of active ingredients comprise one or more analgesics and one or more antimuscarinic agents. In other embodiments, the plurality of active ingredients comprise one or more analgesics and one or more antidiuretic agents. In yet other embodiments, the plurality of active ingredients comprise one or more analgesics, one or more antidiuretic agents, and one or more antimuscarinic agents. In other embodiments, at least one of said plurality of active ingredients is formulated for delayed-release.
[0081] In some embodiments, the pharmaceutical composition comprises two analgesics selected from the group consisting of cetylsalicylic acid, ibuprofen, naproxen sodium, nabumetone, acetaminophen and indomethancin. In other embodiments, the pharmaceutical composition comprises one or more analgesics selected from the group consisting of
cetylsalicylic acid, ibuprofen, naproxen sodium, nabumetone, acetaminophen and indomethancin; and an antimuscarinic agent selected from the group consisting of oxybutynin, solifenacin, darifenacin and atropine.
[0082] As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, sweeteners and the like. The pharmaceutically acceptable carriers may be prepared from a wide range of materials including, but not limited to, flavoring agents, sweetening agents and miscellaneous materials such as buffers and absorbents that may be needed in order to prepare a particular therapeutic composition. The use of such media and agents with pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
[0083] Another aspect of the present application relates to a method for reducing the frequency of urination by administering to a subject in need thereof, two or more analgesic agents alternatively to prevent the development of drug resistance. In one embodiment, the method comprises administering a first analgesic agent for a first period of time and then administering a second analgesic agent for a second period of time. In another embodiment, the method further comprises administering a third analgesic agent for a third period of time. The first, second and third analgesic agents are different from each other and may be selected from the group consisting of: salicylates, aspirin, salicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, para-aminophenol derivatives, acetanilide, acetaminophen, phenacetin, fenamates, mefenamic acid, meclofenamate, sodium meclofenamate, heteroaryl acetic acid derivatives, tolmetin, ketorolac, diclofenac, propionic acid derivatives, ibuprofen, naproxen sodium, naproxen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin; enolic acids, oxicam derivatives, piroxicam, meloxicam, tenoxicam, ampiroxicam, droxicam, pivoxicam, pyrazolon derivatives, phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, dipyrone, coxibs, celecoxib, rofecoxib, nabumetone, apazone, nimesulide, indomethacin, sulindac, etodolac, and isobutylphenyl propionic acid.
[0084] In one embodiment, the first analgesic agent is acetaminophen, the second analgesic agent is ibuprofen and the third analgesic agent is naproxen sodium. The length of each period may vary depending on the subject's response to each analgesic agent. In some embodiments, each period lasts from 3 days to three weeks. In another embodiment, one or more of the first, second and third analgesic is formulated for delayed-release.
[0085] Another aspect of the present application relates to a pharmaceutical composition comprising (1) a first component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 1-4 hours after oral
administration; and (2) a second component comprising one or more analgesic agents, wherein the second component is formulated for delayed-release with a lag time of 4-6 hours after oral administration.
[0086] In some embodiments, the one or more analgesic agents in the first component or the second component are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
[0087] In some embodiments, the second component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
[0088] In some embodiments, the pharmaceutical composition is coated with an enteric coating.
[0089] In some embodiments, the first component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0090] In some embodiments, the second components further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0091] In some embodiments, both the first and second components further comprise an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0092] In some embodiments, the second component is coated with a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
[0093] In some embodiments, the second component is coated with multiple coating layers.
[0094] In some embodiments, the second component is covered by a swelling layer and an outer insoluble but semi-permeable polymer coating. In some related embodiments, the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
[0095] In some embodiments, the second component formulation comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
[0096] Another aspect of the present application relates to pharmaceutical composition comprising (1) a first component comprising an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg, wherein the first component is formulated for immediate-release; and (2) a second component comprising one or more analgesic agents, wherein the second component is formulated for delay ed-release with a lag time of 1-4 hours after oral administration; and (3) a third component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 4-6 hours after oral administration.
[0097] In some embodiments, the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
[0098] In some embodiments, the second component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
[0099] In some embodiments, the second component is coated with an enteric coating. [0100] In some embodiments, the third component comprises an analgesic agent, such as acetaminophen, in an amount of 5-2000 mg.
[0101] In some embodiments, the first component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0102] In some embodiments, the second component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0103] In some embodiments, the third component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0104] In some embodiments, the first and the second components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0105] In some embodiments, the first and the third components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and
spasmolytics.
[0106] In some embodiments, the second and the third components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0107] In some embodiments, the first, the second, and the second components each comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0108] In some embodiments, the second component is coated with an enteric coating.
[0109] In some embodiments, the second component or the third component is coated with a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
[0110] In some embodiments, the third component is coated with multiple coating layers.
[0111] Another aspect of the present application relates to pharmaceutical composition pharmaceutical composition for reducing the frequency of urination, comprising: (1) a first component comprising acetaminophen in an amount of 5-2000 mg, wherein the first component is formulated for immediate-release; and (2) a second component comprising one or more analgesic agents, wherein the second component is formulated for delayed-release with a lag time of 1-4 hours after oral administration; and (3) a third component comprising one or more analgesic agents, wherein the first component is formulated for delayed-release with a lag time of 4-6 hours after oral administration, wherein the pharmaceutical composition reduces the frequency of urination in patients in need thereof. [0112] In some embodiments, the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
Method of manufacture
[0113] Another aspect of the present application relates to methods for manufacturing delayed-release pharmaceutical compositions for reducing the frequency of urination. In some embodiments, the method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a first component, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after oral administration; forming a second mixture comprising a second active ingredient comprising one or more analgesic agents; coating the second mixture with a second delayed-release coating to form a second component, wherein the second delayed-release coating releases the second mixture with a lag time of 4-6 hours after oral administration; and combining the first component with the second component to form a third mixture.
[0114] In some embodiments, the first, second and/or third mixture comprises a pharmaceutically acceptable carrier, such as an excipient.
[0115] In some embodiments, the one or more analgesic agents in the second component or the third component or both are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
[0116] In some embodiments, at least one of the first and the second active ingredients contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen. In some
embodiments, the first active ingredient contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen. In other embodiments, the second active ingredient contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen. In yet other embodiments, the first and the second active ingredients each contains 5 mg to 2000 mg of an analgesic agent, such as acetaminophen.
[0117] In some embodiments, at least one of the first and the second active ingredients further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics. In some embodiments, the first active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics. In other embodiments, the second active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics. In yet other embodiments, the first and the second active ingredients each contains an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0118] In some embodiments, the first delay ed-release coating is an enteric coating. In some embodiments, the second delayed-release coating comprises a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2. In some embodiments, the second delayed-release coating comprises multiple coating layers.
[0119] In some embodiments, the second delayed-release coating comprises a swelling layer and an outer insoluble but semi-permeable polymer coating. In a related embodiment, the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
[0120] In some embodiments, the second delayed-release coating comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
[0121] In some embodiments, the method further comprises the step of pressing or molding the third mixture into a tablet form.
[0122] In some embodiments, the third mixture is formed by combining the first component and the second component with a third component, wherein the third component comprises a third active ingredient comprising one or more analgesic agents. In some embodiments, the one or more analgesic agents are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
[0123] In some embodiments, the first, second and third active ingredients each comprises one or more analgesic agents. In some embodiments, the one or more analgesic agents are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen. In some embodiments, the first, second and third active ingredients each comprises 5-2000 mg acetaminophen. In some embodiments, one of the first, second and third active ingredients comprises 5-2000 mg acetaminophen. In some embodiments, two of the first, second and third active ingredients comprise 5-2000 mg acetaminophen. [0124] In other embodiments, the method comprises the steps of: forming a first mixture comprising a first active ingredient comprising one or more analgesic agents; coating the first mixture with a first delayed-release coating to form a core structure, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after direct contact with a body fluid; coating the core structure with a second mixture to form a coated core structure, wherein the second mixture comprises a second active ingredient comprising one or more analgesic agents; and coating the coated core structure with a second delayed-release coating to form a double-coated core structure, wherein the second delayed-release coating releases the second mixture with a lag time of 1-4 hours after oral administration. In some embodiments, the first mixture does not have direct contact with a body fluid until the second mixture is released.
[0125] In some embodiments, the one or more analgesic agents in the first active ingredient or the second active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
[0126] In some embodiments, the second delayed-release coating is an enteric coating.
[0127] In some embodiments, the first active ingredient or the second active ingredient or both further comprise an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
[0128] In some embodiments, the first active ingredient or the second active ingredient or both comprise acetaminophen.
[0129] In some embodiments, the method further comprises the step of coating the double-coated core structure with a third coating, wherein the third coating comprises a third active ingredient comprising one or more analgesic agents. In some embodiments, the one or more analgesic agents in the third active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen. In some embodiments, the third active ingredient comprises 5 mg to 2000 mg analgesic agent. In some embodiments, the third active ingredient comprises 5 mg to 2000 mg acetaminophen.
[0130] In some related embodiments, the third active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics. In other related embodiments, the first, second and third active ingredients each comprises acetaminophen.
[0131] In some embodiments, the first delayed-release coating comprises pH
independent polymers containing one or more polysaccharides, wherein the pH independent polymers are resistant to erosion in both the stomach and intestine, but can be degraded in the colon. [0132] In other embodiments, the first delayed-release coating, or the second delayed-release coating, or both, comprise a water-permeable but insoluble film coating to enclose the active ingredient and an osmotic agent in the core structure or the coated core structure. As water from the gut slowly diffuses through the insoluble film into the core structure, the core swells until the film bursts, thereby releasing the active ingredients. The film coating may be adjusted to permit various rates of water permeation or release time. As noted earlier, the release time of the drugs can be controlled by a disintegration lag time depending on the balance between the tolerability and thickness of a water insoluble polymer membrane (such as ethyl cellulose, EC) containing predefined micropores at the bottom of the body and the amount of a swellable excipient, such as low substituted hydroxypropyl cellulose (L-HPC) and sodium glycolate. After oral administration, GI fluids permeate through the micropores, causing swelling of the swellable excipients, which produces an inner pressure disengaging the capsular components.
[0133] In other embodiments, the drugs are released by an osmotic mechanism. In some embodiments, the first delayed-release coating, or the second delayed-release coating, or both comprise a semi-permeable membrane. In some embodiments, the semi-permeable membrane is additionally covered with a pH-dependent enteric coating to prevent release until after gastric emptying.
[0134] The present invention is further illustrated by the following example which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are incorporated herein by reference.
EXAMPLE 1 : INHIBITION OF THE URGE TO URINATE
[0135] Twenty volunteer subjects, both male and female were enrolled, each of which experienced premature urge or desire to urinate, interfering with their ability to sleep for a sufficient period of time to feel adequately rested. Each subject ingested 400-800 mg of ibuprofen as a single dose prior to bedtime. At least 14 subjects reported that they were able to rest better because they were not being awakened as frequently by the urge to urinate.
[0136] Several subjects reported that after several weeks of nightly use of ibuprofen, the benefit of less frequent urges to urinate was no longer being realized. However, all of these subjects further reported the return of the benefit after several days of abstaining from taking the dosages.
EXAMPLE 2: EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON MACROPHAGE RESPONSES TO INFLAMMATORY
AND NON-INFLAMMATORY STIMULI Experimental Design
[0137] This study is designed to determine the dose and in vitro efficacy of analgesics and antimuscarinic agents in controlling macrophage response to inflammatory and
non-inflammatory stimuli mediated by Cox-2 and prostaglandins (PGE, PGH, etc.). It establishes baseline (dose and kinetic) responses to inflammatory and non-inflammatory effectors in bladder cells. Briefly, cultured cells are exposed to analgesic agents and/or antimuscarinic agents in the absence or presence of various effectors.
[0138] The effectors include: lipopolysaccharide (LPS), an inflammatory agent and COX2 inducer, as inflammatory stimuli; carbachol or acetylcholine, a stimulator of smooth muscle contraction, as non-inflammatory stimuli; botulinum neurotoxin A, a known inhibitor of acetylcholine release, as positive control; and arachidonic acid (AA), gamma linolenic acid (DGLA) or eicosapentaenoic acid (EPA) as precursors of prostaglandins, which are produced following the sequential oxidation of AA, DGLA or EPA inside the cell by cyclooxygenases (COX1 and COX2) and terminal prostaglandin synthases.
[0139] The analgesic agents include: Salicylates such as aspirin,
iso-butyl-propanoic-phenolic acid derivative (ibuprofen) such as Advil, Motrin, Nuprin, and Medipren, naproxen sodium such as Aleve, Anaprox, Antalgin, Feminax Ultra, Flanax, Inza, Midol Extended Relief, Nalgesin, Naposin, Naprelan, Naprogesic, Naprosyn, Naprosyn suspension, EC-Naprosyn, Narocin, Proxen, Synflex and Xenobid, acetic acid derivative such as indomethacin (Indocin),l-naphthaleneacetic acid derivative such as nabumetone or relafen, N-acetyl-para-aminophenol (APAP) derivative such as acetaminophen or paracetamol (Tylenol) and Celecoxib.
[0140] The antimuscarinic agents include: oxybutynin, solifenacin, darifenacin and atropine.
[0141] Macrophages are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
1) Each analgesic agent alone at various doses.
(2) Each analgesic agent at various doses in the presence of LPS.
(3) Each analgesic agent at various doses in the presence of carbachol or acetylcholine.
(4) Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
(5) Botulinum neurotoxin A alone at various doses.
(6) Botulinum neurotoxin A at various doses in the presence of LPS.
(7) Botulinum neurotoxin A at various doses in the presence of carbachol or
acetylcholine.
(8) Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA. (9) Each antimuscarinic agent alone at various doses.
(10) Each antimuscarinic agent at various doses in the presence of LPS.
(11) Each antimuscarinic agent at various doses in the presence of carbachol or acetylcholine.
(12) Each antimuscarinic agent at various doses in the presence of AA, DGLA, or EPA.
[0142] The cells are then analyzed for the release of PGH2, PGE, PGE2, Prostacydin,
Thromboxane, IL-Ιβ, IL-6, TNF-α, the COX2 activity, the production of cAMP and cGMP, the production of IL-Ιβ, IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
Materials and Methods
Macrophage cells
[0143] Murine RAW264.7 or J774 macrophage cells (obtained from ATCC) were used in this study. Cells were maintained in a culture medium containing RPMI 1640 supplemented with 10 % fetal bovine serum (FBS), 15 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, and 100 μg / ml of streptomycin. Cells were cultured at 37° C in a 5 % C02 atmosphere and split (passages) once a week.
In vitro treatment of macrophage cells with analgesics
[0144] RAW264.7 macrophage cells were seeded in 96-well plates at a cell density of 1.5xl05 cells per well in 100 μΐ of the culture medium. The cells were treated with (1) various concentrations of analgesic (acetaminophen, aspirin, ibuprophen or naproxen), (2) various concentrations of lipopolysaccharide (LPS), which is an effector of inflammatory stimuli to macrophage cells, (3) various concentrations of carbachol or acetylcholine, which are effectors of non-inflammatory stimuli, (4) analgesic and LPS or (5) analgesic and carbachol or acetylcholine. Briefly, the analgesics were dissolved in FBS-free culture medium {i.e., RPMI 1640 supplemented with 15 mM HEPES, 2 mM L-glutamine, 100 U / ml penicillin, and 100 μg / ml of streptomycin), and diluted to desired concentrations by serial dilution with the same medium. For cells treated with analgesic in the absence of LPS, 50 μΐ of analgesic solution and 50 μΐ of FBS-free culture medium were added to each well. For cells treated with analgesic in the presence of LPS, 50 μΐ of analgesic solution and 50 μΐ of LPS (from Salmonella
typhimurium) in FBS-free culture medium were added to each well. All conditions were tested in duplicates.
[0145] After 24 or 48 hours of culture, 150 μΐ of culture supernatants were collected, spun down for 2 min at 8,000 rpm at 4°C to remove cells and debris and stored at -70°C for analysis of cytokine responses by ELISA. The cells were collected and washed by
centrifugation (5 min at 1,500 rpm at 4°C) in 500 μΐ of Phosphate buffer (PBS). Half of the cells were then snap frozen in liquid nitrogen and stored at -70°C. The remaining cells were stained with fluorescent monoclonal antibodies and analyzed by flow cytometry.
Flow cytometry analysis of co-stimulatory molecule expression
[0146] For flow cytometry analysis, macrophages were diluted in 100 μΐ of F ACS buffer (phosphate buffered saline (PBS) with 2% bovine serum albumin (BSA) and 0.01% NaN3) and stained 30 min at 4°C by addition of FITC-conjugated anti-CD40, PE-conjugated anti-CD80, PE-conjugated anti-CD86 antibody, anti MHC class II (I-Ad ) PE (BD Bioscience). Cells were then washed by centrifugation (5 min at 1,500 rpm at 4°C) in 300 μΐ of FACS buffer. After a second wash, cells were re-suspended in 200 μΐ of FACS buffer and the percentage of cells expressing a given marker (single positive), or a combination of markers (double positive) were analyzed with the aid of an Accuri C6 flow cytometer (BD Biosciences).
Analysis of cytokine responses by ELISA
[0147] Culture supernatants were subjected to cytokine-specific ELISA to determine IL-Ιβ, IL-6 and T F-oc responses in cultures of macrophages treated with analgesic, LPS alone or a combination of LPS and analgesic. The assays were performed on Nunc MaxiSorp
Immunoplates (Nunc) coated overnight with 100 μΐ of anti-mouse IL-6, TNF-oc mAbs (BD Biosciences) or IL-Ιβ mAb (R&D Systems) in 0.1 M sodium bicarbonate buffer (pH 9.5).
After two washes with PBS (200 μΐ per well), 200 μΐ of PBS 3% BSA were added in each well (blocking) and the plates incubated for 2 hours at room temperature. Plates were washed again two times by addition of 200 μΐ per well, 100 μΐ of cytokine standards and serial dilutions of culture supernatants were added in duplicate and the plates were incubated overnight at 4°C. Finally, the plates were washed twice and incubated with 100 μΐ of secondary biotinylated anti-mouse IL-6, TNFoc mAbs (BD Biosciences) or IL-Ιβ (R&D Systems) followed by peroxidase-labelled goat anti-biotin mAb (Vector Laboratories). The colorimetric reaction was developed by the addition of 2,2'-azino-bis (3)-ethylbenzylthiazoline-6-sulfonic acid (ABTS) substrate and H202 (Sigma) and the absorbance measured at 415 nm with a Victor® V multilabel plate reader (PerkinElmer).
Determination of COX2 activity and the production of cAMP and cGMP
[0148] The COX2 activity in the cultured macrophages is determined by COX2 activity assay. The production of cAMP and cGMP is determined by the cAMP assay and cGMP assay. These assays are performed routinely in the art.
Results
[0149] Table 1 summarizes the experiments performed with Raw 264 macrophage cell line and main findings in terms of the effects of analgesics on cell surface expression of costimulatory molecules CD40 and CD80. Expression of these molecules is stimulated by COX2 and inflammatory signals and thus, was evaluated to determine functional consequences of inhibition of COX2.
Table 1. Summary of experiments
Figure imgf000031_0001
[0150] As shown in Table 2, acetaminophen, aspirin, ibuprophen and naproxen inhibit basal expression of co-stimulatory molecules CD40 and CD80 by macrophages at all the tested doses (i.e., 5x 105 nM, 5x 104 nM, 5x 103 nM, 5x 102 nM, 50 nM and 5 nM), except for the highest dose (i.e., 5x 106 nM), which appears to enhance, rather than inhibit, expression of the co-stimulatory molecules. As shown in Figures 1 A and IB, such inhibitory effect on CD40 and CD50 expression was observed at analgesic doses as low as 0.05 nM (i.e., 0.00005 μΜ). This finding supports the notion that a controlled release of small doses of analgesic may be preferable to acute delivery of large doses. The experiment also revealed that acetaminophen, aspirin, ibuprophen and naproxen have a similar inhibitory effect on LPS induced expression of CD40 and CD80. Table 2. Summary of main findings
Figure imgf000032_0001
* ND: not done (toxicity)
[0151] Table 3 summarizes the results of several studies that measured serum levels of analgesic after oral therapeutic doses in adult humans. As shown in Table 3, the maximum serum levels of analgesic after an oral therapeutic dose are in the range of 104 to 105 nM. Therefore, the doses of analgesic tested in vitro in Table 2 cover the range of concentrations achievable in vivo in humans.
Table 3. Serum levels of analgesic in human blood after oral therapeutic doses
Figure imgf000032_0002
EXAMPLE 3 : EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON-INFLAMMATORY STIMULI
Experimental Design
[0152] This study is designed to characterize how the optimal doses of analgesic determined in Example 2 affect bladder smooth muscle cells in cell culture or tissue cultures, and to address whether different classes of analgesics can synergize to more efficiently inhibit COX2 and PGE2 responses.
[0153] The effectors, analgesic agents and antimuscarinic agents are described in Example 2.
[0154] Primary culture of mouse bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
(1) Each analgesic agent alone at various doses.
(2) Each analgesic agent at various doses in the presence of LPS.
(3) Each analgesic agent at various doses in the presence of carbachol or acetylcholine.
(4) Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
(5) Botulinum neurotoxin A alone at various doses.
(6) Botulinum neurotoxin A at various doses in the presence of LPS.
(7) Botulinum neurotoxin A at various doses in the presence of carbachol or
acetylcholine.
(8) Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
(9) Each antimuscarinic agent alone at various doses.
(10) Each antimuscarinic agent at various doses in the presence of LPS.
(11) Each antimuscarinic agent at various doses in the presence of carbachol or acetylcholine.
(12) Each antimuscarinic agent at various doses in the presence of AA, DGLA, or EPA.
[0155] The cells are then analyzed for the release of PGH2, PGE, PGE2, Prostacydin,
Thromboxane, IL-Ιβ, IL-6, TNF-a, the COX2 activity, the production of cAMP and cGMP, the production of IL-Ιβ, IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
Materials and Methods
Isolation and purification of murine bladder cells
[0156] Bladder cells were removed from euthanized animals C57BL/6 mice (8-12 weeks old) and cells were isolated by enzymatic digestion followed by purification on a Percoll gradient. Briefly, bladders from 10 mice were minced with scissors to fine slurry in 10 ml of digestion buffer (RPMI 1640, 2% fetal bovine serum, 0.5 mg/ml collagenase, 30 μg/ml DNase). Bladder slurries were enzymatically digested for 30 minutes at 37°C. Undigested fragments were further dispersed through a cell-trainer. The cell suspension was pelleted and added to a discontinue 20%, 40% and 75% Percoll gradient for purification on mononuclear cells. Each experiment used 50-60 bladders.
[0157] After washes in RPMI 1640, bladder cells were resuspended RPMI 1640 supplemented with 10 % fetal bovine serum, 15 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, and 100 μg / ml of streptomycin and seeded in clear-bottom black 96-well cell culture microculture plates at a cell density of 3xl04 cells per well in 100 μΐ. Cells were cultured at 37° C in a 5 % C02 atmosphere.
In vitro treatment of cells with analgesics
[0158] Bladder cells were treated with analgesic solutions (50 μΐ/ well) either alone or together with carbachol (10-Molar, 50 μΐ/ well), as an example of non-inflammatory stimuli, or lipopolysaccharide (LPS) of Salmonella typhimurium (1 μg/ml, 50 μΐ/ well), as an example of non-inflammatory stimuli. When no other effectors were added to the cells, 50 μΐ of RPMI 1640 without fetal bovine serum were added to the wells to adjust the final volume to 200 μΐ.
[0159] After 24 hours of culture, 150 μΐ of culture supernatants were collected, spun down for 2 min at 8,000 rpm at 4°C to remove cells and debris and stored at -70°C for analysis of Prostaglandin E2 (PGE2) responses by ELISA. Cells were fixed, permeabilized and blocked for detection of Cyclooxygenase-2 (COX2) using a fluorogenic substrate. In selected experiment cells were stimulated 12 hours in vitro for analysis of COX2 responses.
Analysis of COX2 responses
[0160] COX2 responses were analyzed by a Cell-Based ELISA using Human/mouse total COX2 immunoassay (R&D Systems), following the instructions of the manufacturer. Briefly, after cells fixation and permeabilization, a mouse anti-total COX2 and a rabbit anti-total GAPDH were added to the wells of the clear-bottom black 96-well cell culture microculture plates. After incubation and washes, an HRP-conjugated anti-mouse IgG and an AP-conjugated anti-rabbit IgG were added to the wells. Following another incubation and set of washes, the HRP- and AP-fluorogenic substrates were added. Finally, a Victor® V multilabel plate reader (PerkinElmer) was used to read the fluorescence emitted at 600 nm (COX2 fluorescence) and 450 nm (GAPDH fluorescence). Results are expressed as relative levels of total COX2 as determined by relative fluorescence unit (RFUs) and normalized to the housekeeping protein GAPDH. Analysis of PGE2 responses
[0161] Prostaglandin E2 responses were analyzed by a sequential competitive ELISA (R&D Systems). More specifically, culture supernatants or PGE2 standards were added to the wells of a 96-well polystyrene microplate coated with a goat anti-mouse polyclonal antibody. After one hour incubation on a microplate shaker, an HRP-conjugated PGE2 was added and plates incubated for an additional two hours at room temperature. The plates were then washed and HRP substrate solution added to each well. The color was allowed to develop for 30 min and the reaction stopped by the addition of sulfuric acid before reading the plate at 450 nm with wavelength correction at 570 nm. Results are expressed as mean pg/ml of PGE2.
Other assays
[0162] The release of PGH2, PGE, Prostacydin, Thromboxane, IL-Ιβ, IL-6, and T F-a, the production of cAMP and cGMP, the production of IL-Ιβ, IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules are determined as described in Example 2.
Results
Analgesics inhibit COX2 responses of murine bladder cells to an inflammatory stimuli
[0163] Several analgesics (acetaminophen, aspirin, ibuprofen and naproxen) were tested on mouse bladder cells at the concentration of 5 μΜ or 50 μΜ to determine whether the analgesics could induce COX2 responses. Analysis of 24-hour cultures showed that none of the analgesics tested induced COX2 responses in murine bladder cells in vitro.
[0164] The effect of these analgesics on the COX2 responses of murine bladder cells to carbachol or LPS stimulation in vitro was also tested. As indicated in Table 1, the dose of carbachol tested has no significant effect on COX2 levels in murine bladder cells. On the other hand, LPS significantly increased total COX2 levels. Interestingly, acetaminophen, aspirin, ibuprofen and naproxen could all suppress the effect of LPS on COX2 levels. The suppressive effect of the analgesic was seen when these drugs were tested at either 5 μΜ or 50 μΜ (Table 4).
Table 4. COX2 expression by murine bladder cells after in vitro stimulation and treatment with analgesics
Figure imgf000036_0001
Analgesics inhibit PGE2 responses of murine bladder cells to an inflammatory stimuli
[0165] The secretion of PGE2 in culture supernatants of murine bladder cells was measured to determine the biological significance of the alteration of murine bladder cell COX2 levels by analgesics. As shown in Table 5, PGE2 was not detected in the culture supernatants of unstimulated bladder cells or bladder cells cultured in the presence of carbachol. Consistent with COX2 responses described above, stimulation of murine bladder cells with LPS induced the secretion of high levels of PGE2. Addition of the analgesics acetaminophen, aspirin, ibuprofen and naproxen suppressed the effect of LPS on PGE2 secretion and no difference was seen between the responses of cells treated with the 5 or 50 μΜ dose of analgesic.
Table 5. PGE2 secretion by murine bladder cells after in vitro stimulation and treatment with analgesics
Figure imgf000037_0001
[0166] In summary, these data show that the analgesics alone at 5 μΜ or 50 μΜ do not induce COX2 and PGE2 responses in murine bladder cells. The analgesics at 5 μΜ or 50 μΜ, however, significantly inhibit COX2 and PGE2 responses of murine bladder cells stimulated in vitro with LPS (1 μg/ml). No significant effect of analgesics was observed on COX2 and PGE2 responses of murine bladder cells stimulated with carbachol (1 mM).
EXAMPLE 4: EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON BLADDER SMOOTH MUSCLE CELL CONTRACTION. Experimental Design
[0167] Cultured mouse or rat bladder smooth muscle cells and mouse or rat bladder smooth muscle tissue are exposed to inflammatory stimuli and non-inflammatory stimuli in the presence of analgesic agent and/or antimuscarinic agent at various concentrations. The stimuli-induced muscle contraction is measured to evaluate the inhibitory effect of the analgesic agent and/or antimuscarinic agent.
[0168] The effectors, analgesic agents and antimuscarinic agents are described in Example 2.
[0169] Primary culture of mouse bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
(1) Each analgesic agent alone at various doses.
(2) Each analgesic agent at various doses in the presence of LPS.
(3) Each analgesic agent at various doses in the presence of carbachol or acetylcholine. (4) Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
(5) Botulinum neurotoxin A alone at various doses.
(6) Botulinum neurotoxin A at various doses in the presence of LPS.
(7) Botulinum neurotoxin A at various doses in the presence of carbachol or
acetylcholine.
(8) Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
(9) Each antimuscarinic agent alone at various doses.
(10) Each antimuscarinic agent at various doses in the presence of LPS.
(11) Each antimuscarinic agent at various doses in the presence of carbachol or acetylcholine.
(12) Each antimuscarinic agent at various doses in the presence of AA, DGLA, or EPA. Materials and Methods
[0170] Primary mouse bladder cells are isolated as described in Example 3. In selected experiments, cultures of bladder tissue are used. Bladder smooth muscle cell contractions are recorded with a Grass polygraph (Quincy Mass, USA).
EXAMPLE 5: EFFECT OF ORAL ANALGESIC AGENTS AND ANTIMUSCARINIC AGENTS ON COX2 AND PGE2 RESPONSES OF BLADDER SMOOTH MUSCLE CELLS. Experimental design:
[0171] Normal mice and mice with over active bladder syndrome are given oral doses of aspirin, naproxen sodium, Ibuprofen, Indocin, nabumetone, Tylenol, Celecoxib, oxybutynin, solifenacin, darifenacin, atropine and combinations thereof. Control groups include untreated normal mice and untreated OAB mice without over active bladder syndrome. Thirty (30) min after the last doses, the bladders are collected and stimulated ex vivo with carbachol or acetylcholine. In selected experiments the bladders are treated with botulinum neurotoxin A before stimulation with carbachol. Animals are maintained in metabolic cages and frequency (and volume) of urination are evaluated. Bladder outputs are determined by monitoring water intake and cage litter weight. Serum PGH2, PGE, PGE2, Prostacydin, Thromboxane, IL-Ιβ, IL-6, TNF-a, cAMP, and cGMP levels are determined by ELISA. CD80, CD86, MHC class II expression in whole blood cells are determined by flow cytometry.
[0172] At the end of the experiment, animals are euthanized and ex vivo bladder contractions are recorded with a Grass polygraph. Portions of bladders are fixed in formalin, and COX2 responses are analyzed by immunohistochemistry.
EXAMPLE 6: EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTFMUSCARINIC AGENTS ON HUMAN BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON-INFLAMMATORY STIMULI Experimental Design
[0173] This study is designed to characterize how the optimal doses of analgesics determined in Examples 1-5 affect human bladder smooth muscle cells in cell culture or tissue cultures, and to address whether different classes of analgesics can synergize to more efficiently inhibit COX2 and PGE2 responses.
[0174] The effectors, analgesic agents and antimuscarinic agents are described in Example 2.
[0175] Human bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
(1) Each analgesic agent alone at various doses.
(2) Each analgesic agent at various doses in the presence of LPS.
(3) Each analgesic agent at various doses in the presence of carbachol or acetylcholine.
(4) Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
(5) Botulinum neurotoxin A alone at various doses.
(6) Botulinum neurotoxin A at various doses in the presence of LPS.
(7) Botulinum neurotoxin A at various doses in the presence of carbachol or
acetylcholine.
(8) Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
(9) Each antimuscarinic agent alone at various doses.
(10) Each antimuscarinic agent at various doses in the presence of LPS.
(11) Each antimuscarinic agent at various doses in the presence of carbachol or acetylcholine.
(12) Each antimuscarinic agent at various doses in the presence of AA, DGLA, or EPA.
[0176] The cells are then analyzed for the release of PGH2, PGE, PGE2, Prostacydin,
Thromboxane, IL-Ιβ, IL-6, T F-α, the COX2 activity, the production of cAMP and cGMP, the production of IL-Ιβ, IL-6, TNF-a and COX2 mRNA, and surface expression of CD80, CD86 and MHC class II molecules.
EXAMPLE 7: EFFECT OF ANALGESIC AGENTS. BOTULINUM NEUROTOXIN AND ANTPMUSCARINIC AGENTS ON HUMAN BLADDER SMOOTH MUSCLE CELL CONTRACTION.
Experimental Design
[0177] Cultured human bladder smooth muscle cells are exposed to inflammatory stimuli and non-inflammatory stimuli in the presence of analgesic agent and/or antimuscarinic agent at various concentrations. The stimuli-induced muscle contraction is measured to evaluate the inhibitory effect of the analgesic agent and/or antimuscarinic agent. [0178] The effectors, analgesic agents and antimuscarinic agents are described in Example 2.
[0179] Human bladder smooth muscle cells are subjected to short term (1-2 hrs) or long term (24-48 hrs) stimulation of with:
(1) Each analgesic agent alone at various doses.
(2) Each analgesic agent at various doses in the presence of LPS.
(3) Each analgesic agent at various doses in the presence of carbachol or acetylcholine.
(4) Each analgesic agent at various doses in the presence of AA, DGLA, or EPA.
(5) Botulinum neurotoxin A alone at various doses.
(6) Botulinum neurotoxin A at various doses in the presence of LPS.
(7) Botulinum neurotoxin A at various doses in the presence of carbachol or
acetylcholine.
(8) Botulinum neurotoxin A at various doses in the presence of AA, DGLA, or EPA.
(9) Each antimuscarinic agent alone at various doses.
(10) Each antimuscarinic agent at various doses in the presence of LPS.
(11) Each antimuscarinic agent at various doses in the presence of carbachol or acetylcholine.
(12) Each antimuscarinic agent at various doses in the presence of AA, DGLA, or EPA.
[0180] Bladder smooth muscle cell contractions are recorded with a Grass polygraph
(Quincy Mass, USA).
EXAMPLE 8: EFFECT OF ANALGESIC AGENTS ON NORMAL HUMAN BLADDER SMOOTH MUSCLE CELL RESPONSES TO INFLAMMATORY AND NON INFLAMMATORY SIGNALS EXPERIMENTAL DESIGN
Culture of normal human bladder smooth muscle cells
[0181] Normal human bladder smooth muscle cells were isolated by enzymatic digestion from macroscopically normal pieces of human bladder. Cells were expended in vitro by culture at 37° C in a 5 % C02 atmosphere in RPMI 1640 supplemented with 10 % fetal bovine serum, 15 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin, and 100 mg / ml of streptomycin and passage once a week by treatment with trypsin to detach cells followed by reseeding in a new culture flask. The first week of culture, the culture medium was supplemented with 0.5 ng/ml epidermal growth factor, 2 ng/ml fibroblast growth factor, and 5 μg/ml insulin.
Treatment of normal human bladder smooth muscle cells with analgesics in vitro
[0182] Bladder smooth muscle cells trypsinized and seeded in microculture plates at a cell density of 3xl04 cells per well in 100 μΐ were treated with analgesic solutions (50 μΐ/ well) either alone or together carbachol (10-Molar, 50 μΐ/ well), as an example of non-inflammatory stimuli, or lipopolysaccharide (LPS) of Salmonella typhimurium (1 μg/ml, 50 μΐ/ well), as an example of non-inflammatory stimuli. When no other effectors were added to the cells, 50 μΐ of RPMI 1640 without fetal bovine serum were added to the wells to adjust the final volume to 200 μΐ.
[0183] After 24 hours of culture, 150 μΐ of culture supernatants were collected, spun down for 2 min at 8,000 rpm at 4°C to remove cells and debris and stored at -70°C for analysis of Prostaglandin E2 (PGE2) responses by ELISA. Cells were fixed, permeabilized and blocked for detection of COX2 using a fluorogenic substrate. In selected experiments cells were stimulated 12 hours in vitro for analysis of COX2, PGE2 and cytokine responses.
Analysis of COX2, PGE2 and cytokine responses
[0184] COX2 and PGE2 responses were analyzed as described in Example 3. Cytokine responses were analyzed as described in Example 2.
RESULTS
[0185] Analgesics inhibit COX2 responses of normal human bladder smooth muscle cells to inflammatory and non- inflammatory stimuli - Analysis of cells and culture supernatants after 24 hours of cultures showed that none of the analgesics tested alone induced COX2 responses in normal human bladder smooth muscle cells. However, as summarized in Table 6, carbachol induced low, but significant COX2 responses in normal human bladder smooth muscle cells. On the other hand, LPS treatment resulted in higher levels of COX2 responses in normal human bladder smooth muscle cells. Acetaminophen, aspirin, ibuprofen and naproxen could all suppress the effect of carbachol and LPS on COX2 levels. The suppressive effect of the analgesics was seen on LPS-induced responses when these drugs were tested at either 5 μΜ or 50 μΜ.
Table 6. COX2 expression by normal human bladder smooth muscle cells after in vitro stimulation with inflammatory and non- inflammatory stimuli and treatment with analgesic
Figure imgf000042_0001
#Data are expressed as mean of duplicates
[0186] Analgesics inhibit PGE2 responses of normal human bladder smooth muscle cells to inflammatory and non- inflammatory stimuli - Consistent with the induction of COX2 responses described above, both carbachol and LPS induced production of PGE2 by normal human bladder smooth muscle cells. Acetaminophen, aspirin, ibuprofen and naproxen were also found to suppress the LPS-induced PGE2 responses at either 5 μΜ or 50 μΜ (Table 7). Table 7. PGE2 secretion by normal human bladder smooth muscle cells after in vitro stimulation with inflammatory and non- inflammatory stimuli and treatment with analgesic
Figure imgf000043_0001
[0187] Analgesics inhibit cytokine responses of normal human bladder cells to an inflammatory stimuli - Analysis of cells and culture supernatants after 24 hours of cultures showed that none the analgesics tested alone induced IL-6 or TNFa secretion in normal human bladder smooth muscle cells. As shown in Tables 8 and 9, the dose of carbachol tested induced low, but significant TNFa and IL-6 responses in normal human bladder smooth muscle cells. On the other hand, LPS treatment resulted in massive induction of these proinflammatory cytokines. Acetaminophen, aspirin, ibuprofen and naproxen suppress the effect of carbachol and LPS on TNFa and IL-6 responses. The suppressive effect of the analgesic on LPS-induced responses was seen when these drugs were tested at either 5 μΜ or 50 μΜ. Table 8. TNFa secretion by normal human bladder smooth muscle cells after in vitro stimulation with inflammatory and non- inflammatory stimuli and treatment with analgesic
Figure imgf000044_0001
Table 9. IL-6 secretion by normal human bladder smooth muscle cells after in vitro stimulation with inflammatory and non- inflammatory stimuli and treatment with analgesic
Figure imgf000045_0001
[0188] Primary normal human bladder smooth muscle cells were isolated, cultured and evaluated for their responses to analgesics in the presence of non-inflammatory (carbachol) and inflammatory (LPS) stimuli. The goal of this study was to determine whether or not normal human bladder smooth muscle cells recapitulate the observations previously made with murine bladder cells.
[0189] The above-described experiment will be repeated with analgesic agents and/or antimuscarinic agents in delayed-release, or extended-release formulation or
delayed-and-extended-release formulations.
[0190] The above description is for the purpose of teaching the person of ordinary skill in the art how to practice the present invention, and it is not intended to detail all those obvious modifications and variations of it which will become apparent to the skilled worker upon reading the description. It is intended, however, that all such obvious modifications and variations be included within the scope of the present invention, which is defined by the following claims. The claims are intended to cover the claimed components and steps in any sequence which is effective to meet the objectives there intended, unless the context specifically indicates the contrary.

Claims

WHAT IS CLAIMED IS:
1. A method for manufacturing a pharmaceutical composition for reducing the frequency of urination, comprising the steps of:
forming a first mixture comprising a first active ingredient comprising one or more analgesic agents;
coating the first mixture with a first delayed-release coating to form a first component, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after oral administration;
forming a second mixture comprising a second active ingredient comprising one or more analgesic agents;
coating the second mixture with a second delayed-release coating to form a second component, wherein the second delayed-release coating releases the second mixture with a lag time of 4-6 hours after oral administration; and
combining the first component with the second component to form a third mixture.
2. The method of Claim 1, wherein the one or more analgesic agents in the first active ingredient and the second active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
3. The method of Claim 1, wherein the first delayed-release coating is an enteric coating.
4. The method of Claim 1, wherein the first active ingredient, or the second active ingredient, or both further comprise an agent selected from the group consisting of
antimuscarinic agents, antidiuretic agents and spasmolytics.
5. The method of Claim 1, wherein the second delayed-release coating comprises a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WTP:EP ratio of 4: 1 to 1 :2.
6. The method of Claim 1, wherein the second delayed-release coating comprises multiple coating layers.
7. The method of Claim 1, wherein the second delayed-release coating comprises a swelling layer and an outer insoluble but semi-permeable polymer coating.
8. The method of Claim 7, wherein the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
9. The method of Claim 1, wherein the second delay ed-release coating comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
10. The method of Claim 1, further comprising the step of pressing the third mixture into a tablet form.
11. The method of Claim 1, wherein the third mixture is formed by combining the first component and the second component with a third component, wherein the third component comprises a third active ingredient comprising one or more analgesic agents.
12. A pharmaceutical composition manufactured using the method of Claim 1.
13. A method for manufacturing a pharmaceutical composition for reducing the frequency of urination, comprising:
forming a first mixture comprising a first active ingredient comprising one or more analgesic agents;
coating the first mixture with a first delayed-release coating to form a core structure, wherein the first delayed-release coating releases the first mixture with a lag time of 1-4 hours after direct contact with a body fluid;
coating the core structure with a second mixture to form a coated core structure, wherein the second mixture comprises a second active ingredient comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen; and
coating the coated core structure with a second delayed-release coating to form a double-coated core structure, wherein the second delayed-release coating releases the second mixture with a lag time of 1-4 hours after oral administration.
14. The method of Claim 13, wherein the second delayed-release coating is an enteric coating.
15. The method of Claim 13, wherein the one or more analgesic agents in the first active ingredient and the second active ingredient are selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
16. The method of Claim 13, wherein the first active ingredient or the second active ingredient or both further comprise an agent selected from the group consisting of
antimuscarinic agents, antidiuretic agents and spasmolytics.
17. The method of Claim 13, further comprising the step of coating the double-coated core structure with a third coating, wherein the third coating comprises a third active ingredient comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen.
18. The method of Claim 17, wherein the third active ingredient further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
19. A pharmaceutical composition manufactured using the method of Claim 13.
20. A pharmaceutical composition, comprising:
a first component comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen, wherein the first component is formulated for delayed-release with a lag time of 1-4 hours; and
a second component comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen, wherein the second component is formulated for delayed-release with a lag time of 4-6 hours.
21. The pharmaceutical composition of Claim 20, wherein the pharmaceutical composition is coated with an enteric coating.
22. The pharmaceutical composition of Claim 20, wherein the first component and/or the second component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
23. The pharmaceutical composition of Claim 20, wherein the second component is coated with a blend of a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
24. The pharmaceutical composition of Claim 20, wherein the second component is coated with multiple coating layers.
25. The pharmaceutical composition of Claim 20, wherein the second component is covered by a swelling layer and an outer insoluble but semi-permeable polymer coating.
26. The pharmaceutical composition of Claim 20, wherein the swelling layer comprises a material selected from the group consisting of hydroxypropyl methylcellulose,
methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose,
carboxymethylcellulose, acrylate copolymers, polyethylene glycols, polyvinylpyrrolidone, methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and ethylcellulose.
27. The pharmaceutical composition of Claim 20, wherein the second component formulation comprises a water insoluble capsule body closed at one end with an insoluble, but permeable and swellable hydrogel plug, wherein said plug comprises a material selected from the group consisting of polymethacrylates, erodible compressed polymers, congealed melted polymer and enzymatically controlled erodible polymers.
28. A pharmaceutical composition, comprising:
a first component comprising acetaminophen in an amount of 5-2000 mg, wherein the first component is formulated for immediate-release; and
a second component comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen, wherein the second component is formulated for delayed-release with a lag time of 1-4 hours; and
a third component comprising one or more analgesic agents selected from the group consisting of aspirin, ibuprofen, naproxen, naproxen sodium, indomethacin, nabumetone, and acetaminophen, wherein the third component is formulated for delayed-release with a lag time of 4-6 hours.
29. The pharmaceutical composition of Claim 28, wherein the second component is coated with an enteric coating.
30. The pharmaceutical composition of Claim 28, wherein the first component and/or the second component and/or the third component further comprises an agent selected from the group consisting of antimuscarinic agents, antidiuretic agents and spasmolytics.
31. The pharmaceutical composition of Claim 30, wherein the second component is coated with an enteric coating.
32. The pharmaceutical composition of Claim 28, wherein the second component or the third component is coated with a water insoluble polymer (WIP) and an enteric polymer (EP) at a WIP:EP ratio of 4: 1 to 1 :2.
33. The pharmaceutical composition of Claim 28, wherein the third component is coated with multiple coating layers.
PCT/US2016/041354 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof WO2017039833A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP16842483.6A EP3344241A4 (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof
AU2016317093A AU2016317093A1 (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof
JP2018530487A JP2018526442A (en) 2015-09-01 2016-07-07 Delayed release formulations, methods of making and using the same
SG11201805812QA SG11201805812QA (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof
RU2018111401A RU2018111401A (en) 2015-09-01 2016-07-07 DELAYED DELIVERY COMPOSITIONS, METHODS FOR PRODUCING AND USING THEM
KR1020187009153A KR20180054656A (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of manufacture and uses
MX2018002449A MX2018002449A (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof.
CN201680050660.8A CN108348487A (en) 2015-09-01 2016-07-07 Delayed release preparation, preparation method and use
HK19101249.6A HK1259196A1 (en) 2015-09-01 2019-01-24 Delayed-release formulations, methods of making and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14/842,539 2015-09-01
US14/842,539 US10278925B2 (en) 2012-01-04 2015-09-01 Delayed-release formulations, methods of making and use thereof

Publications (1)

Publication Number Publication Date
WO2017039833A1 true WO2017039833A1 (en) 2017-03-09

Family

ID=58188685

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/041354 WO2017039833A1 (en) 2015-09-01 2016-07-07 Delayed-release formulations, methods of making and use thereof

Country Status (10)

Country Link
EP (1) EP3344241A4 (en)
JP (1) JP2018526442A (en)
KR (1) KR20180054656A (en)
CN (1) CN108348487A (en)
AU (1) AU2016317093A1 (en)
HK (1) HK1259196A1 (en)
MX (1) MX2018002449A (en)
RU (1) RU2018111401A (en)
SG (1) SG11201805812QA (en)
WO (1) WO2017039833A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140154314A1 (en) * 2010-07-08 2014-06-05 Wellesley Pharmaceuticals, Llc Delayed-release formulation for reducing the frequency of urination and method of use thereof
US20150010599A1 (en) * 2010-07-08 2015-01-08 WELLESLEY PHARMACELJTlCALS, LLC Pharmaceutical formulation for reducing bladder spasms and method of use thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1758904A (en) * 2003-03-21 2006-04-12 麦克内尔-Ppc股份有限公司 Non-steroidal anti-inflammatory drug dosing regimen
US9415048B2 (en) * 2010-07-08 2016-08-16 Wellesley Pharmaceuticals, Llc Pharmaceutical formulation for reducing frequency of urination and method of use thereof
US20120237574A1 (en) * 2010-07-08 2012-09-20 Wellesley Pharmaceuticals, Llc Delayed-release formulation for reducing the frequency of urination and method of use thereof
CN107648212A (en) * 2012-01-04 2018-02-02 韦尔斯利医药有限公司 For alleviating the sustained release preparation and its application method of frequent micturition
SG11201500407XA (en) * 2012-01-04 2015-03-30 Wellesley Pharmaceuticals Llc Extended-release formulation for reducing the frequency of urination and method of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140154314A1 (en) * 2010-07-08 2014-06-05 Wellesley Pharmaceuticals, Llc Delayed-release formulation for reducing the frequency of urination and method of use thereof
US20150010599A1 (en) * 2010-07-08 2015-01-08 WELLESLEY PHARMACELJTlCALS, LLC Pharmaceutical formulation for reducing bladder spasms and method of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3344241A4 *

Also Published As

Publication number Publication date
EP3344241A1 (en) 2018-07-11
AU2016317093A1 (en) 2018-04-26
SG11201805812QA (en) 2018-08-30
EP3344241A4 (en) 2019-04-03
CN108348487A (en) 2018-07-31
MX2018002449A (en) 2018-08-24
JP2018526442A (en) 2018-09-13
RU2018111401A (en) 2019-10-02
HK1259196A1 (en) 2019-11-29
KR20180054656A (en) 2018-05-24

Similar Documents

Publication Publication Date Title
US8703184B2 (en) Delayed-release formulation for reducing the frequency of urination and method of use thereof
AU2017203418B2 (en) Extended-release formulation for reducing the frequency of urination and method of use thereof
US8445015B2 (en) Extended-release formulation for reducing the frequency of urination and method of use thereof
US20120141554A1 (en) Delayed release formulation for reducing the frequency of urination and method of use thereof
US8236856B2 (en) Delayed release formulation for reducing the frequency of urination and method of use thereof
US8445011B2 (en) Delayed-release formulation for reducing the frequency of urination and method of use thereof
US20190224131A1 (en) Delayed-release formulations, methods of making and use thereof
EP2612662B1 (en) Delayed-release formulation for reducing the frequency of urination and method of use thereof
EP3344240A1 (en) Extended, delayed and immediate release formulation method of manufacturing and use thereof
EP2612661B1 (en) Delayed release formulation for reducing the frequency of urination and method of use thereof
WO2017039833A1 (en) Delayed-release formulations, methods of making and use thereof
EP2612663A1 (en) Extended-release formulation for reducing the frequency of urination and method of use thereof
TWI590822B (en) Delayed release formulation for reducing the frequency of urination and method of use thereof
NZ626622B2 (en) Delayed-release formulation for reducing the frequency of urination and method of use thereof
WO2013103356A1 (en) Delayed release formulation for reducing the frequency of urination and method of use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16842483

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: MX/A/2018/002449

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2018530487

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20187009153

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2018111401

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2016317093

Country of ref document: AU

Date of ref document: 20160707

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 11201805812Q

Country of ref document: SG