WO2010048592A1 - Compositions comprising 4- (2- ( 5-br0m0-4- ( 1-cyclopropylnaphthalen-4-yl) -4h-1, 2, 4-triaz0l-3-ylthi0) acetamido -3-chlorobenzoic acid and pharmaceutically acceptable salts thereof - Google Patents

Compositions comprising 4- (2- ( 5-br0m0-4- ( 1-cyclopropylnaphthalen-4-yl) -4h-1, 2, 4-triaz0l-3-ylthi0) acetamido -3-chlorobenzoic acid and pharmaceutically acceptable salts thereof Download PDF

Info

Publication number
WO2010048592A1
WO2010048592A1 PCT/US2009/061969 US2009061969W WO2010048592A1 WO 2010048592 A1 WO2010048592 A1 WO 2010048592A1 US 2009061969 W US2009061969 W US 2009061969W WO 2010048592 A1 WO2010048592 A1 WO 2010048592A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
subject
pharmaceutical composition
mixture
composition
Prior art date
Application number
PCT/US2009/061969
Other languages
French (fr)
Inventor
Colin Edward Rowlings
Barry D. Quart
Original Assignee
Ardea Biosciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ardea Biosciences, Inc. filed Critical Ardea Biosciences, Inc.
Publication of WO2010048592A1 publication Critical patent/WO2010048592A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4816Wall or shell material
    • A61K9/4825Proteins, e.g. gelatin
    • 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
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5026Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents

Definitions

  • compositions comprising 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof and methods of using the compositions, for example in the treatment of diseases.
  • composition comprising greater than about 35% by weight of a compound or mixture of compounds of structure (I):
  • the composition comprises greater than about 60% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises greater than about 70% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises greater than about 80% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least lOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 250mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 500mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises
  • WSGR Docket No 18545-763.601 comprises at least 750mg of a compound or mixture of compounds of structure (I).
  • the composition comprises at least 800mg of a compound or mixture of compounds of structure (I).
  • the composition comprises about lOOmg, about 200mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 800mg, about 900mg or about lOOOmg of a compound or mixture of compounds of structure (I).
  • the composition comprises about 600mg, about 800mg or about lOOOmg of a compound or mixture of compounds of structure (I).
  • the composition comprises about 600mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 800mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition further comprises one or more pharmaceutically acceptable diluents, binders, lubricants, coating agents, barrier coatings, plasticizers, dispersing agents or film coating additives. In some embodiments, the composition further comprises one or more pharmaceutically acceptable diluents.
  • the diluent is microcrystalline cellulose, silicified microcrystalline cellulose, cellulose, lactose, compressible sugar, mannitol, calcium silicate and calcium phosphate, sodium phosphate, sodium carbonate, or a combination thereof.
  • the diluent is in powder form.
  • the diluent is in granular form.
  • the diluent is microcrystalline cellulose.
  • the composition further comprises one or more pharmaceutically acceptable binders.
  • the binder is hypromellose, povidone, hydroxypropyl cellulose, hydroxyethyl cellulose or starch. In some embodiments, the binder is hypromellose.
  • the composition further comprises one or more pharmaceutically acceptable lubricant.
  • the lubricant is magnesium stearate, stearic acid or sodium stearyl fumarate. In some embodiments, the lubricant is magnesium stearate.
  • the composition further comprises one or more pharmaceutically acceptable barrier coatings.
  • the pharmaceutically acceptable barrier coating is a pharmaceutically acceptable enteric coating. In some embodiments, the barrier coating is hypromellose or polyvinyl alcohol. In some embodiments, the barrier coating is hypromellose. In some embodiments, the composition further comprises one or more pharmaceutically acceptable coating agents.
  • the coating agent is a methacrylic acid based copolymer, Eudragit L30D55, Acryl- Eze, hydroxypropylmethyl cellulose acetate succinate, polyvinylacetate phthalate, cellulose acetate phthalate or a combination thereof.
  • the coating agent is a methacrylic acid based copolymer, Eudragit L30D55 or Acryl-Eze.
  • the composition further comprises one or more pharmaceutically acceptable plasticizers.
  • the plasticizer is triethyl citrate, triacetin, dibutyl phthalate, diethyl phthalate or glycerin. In some embodiments, the plasticizer is triethyl citrate. In some embodiments, the composition further comprises one or more pharmaceutically acceptable film coating additives. In some embodiments, the film coating additive is talc, glycerol monostearate or colloidal silicon dioxide. In some embodiments, the film coating additive is talc. In some embodiments, the composition further comprises one or more pharmaceutically active compounds. In some embodiments, at least one of the one or more pharmaceutically active compounds has anti-viral activity. In some embodiments, the composition further comprises two pharmaceutically active compounds.
  • the composition is a unitary dosage form.
  • M is hydrogen or potassium.
  • M is potassium.
  • the composition is suitable for oral administration to a mammal.
  • the composition is suitable for oral administration to a human. [0004] Disclosed herein, in certain embodiments, is a composition comprising: a compound or
  • the composition is a unitary dosage form. [0005] In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule. In some embodiments, M is hydrogen or potassium. In some embodiments, M is potassium.
  • the diluent is microcrystalline cellulose, silicified microcrystalline cellulose, cellulose, lactose, compressible sugar, mannitol, calcium silicate and calcium phosphate in powder and granular forms, sodium phosphate or sodium carbonate.
  • the diluent is microcrystalline cellulose.
  • the binder is hypromellose, povidone, hydroxypropyl cellulose, hydroxyethyl cellulose or starch. In some embodiments, the binder is hypromellose.
  • the coating agent is a methacrylic acid based copolymers, Eudragit L30D55, Acryl-Eze, hydroxypropylmethyl cellulose acetate succinate, polyvinylacetate phthalate or cellulose acetate phthalate.
  • the coating agent is a methacrylic acid based copolymer.
  • the dispersing agent is talc, glycerol monostearate or colloidal silicon dioxide.
  • the dispersing agent is talc.
  • the plasticizer is triethyl citrate, triacetin, dibutyl phthalate, diethyl phthalate or glycerin.
  • the plasticizer is triethyl citrate.
  • the composition comprises from about lmg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about lOmg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 50mg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 200mg of the compound or mixture of compounds of structure (I).
  • the composition comprises about 300mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 400mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 500mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 600mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 700mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 800mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 900mg of the compound or mixture of compounds of structure (I).
  • the composition comprises about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 10% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 25% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 50% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 65% to about 90% by weight of the compound or mixture of compounds of structure (I).
  • composition comprising: a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • the composition comprises: about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form: (IB); about 35mg of microcrystalline cellulose; about 29mg of hypromellose; about 30mg of methacrylic acid copolymer dispersion; about 6mg of talc; and about 3mg of triethyl citrate.
  • the composition is a unitary dosage form.
  • the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule.
  • composition comprising: from about 60% to about 90% by weight of a compound of structure (IB) or a mixture of a compound of structure
  • (IB) and its free acid form (IB); from about 5% to about 15% by weight of microcrystalline cellulose; from about 5% to about 15% by weight of hypromellose; from about 5% to about 15% by weight of methacrylic acid copolymer dispersion; from about 0.5% to about 5% by weight of talc; and from about 0.1% to about 3% by weight of triethyl citrate.
  • the composition comprises from about 60% to about 80% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 60% to about 75% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 65% to about 70% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 67% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form.
  • the composition comprises from about 7% to about 13% by weight of microcrystalline cellulose. In some embodiments, the composition comprises about 11% by weight of microcrystalline cellulose. In some embodiments, the composition comprises from about 7% to about 11% by weight of hypromellose. In some embodiments, the composition comprises about 9% by weight of hypromellose. In some embodiments, the composition comprises from about 5% to about 15% by weight of methacrylic acid copolymer dispersion. In some embodiments, the composition comprises from about 8% to about 12% by weight of methacrylic acid copolymer dispersion. In some embodiments, the composition comprises about 10% by weight of methacrylic acid copolymer dispersion.
  • the composition comprises from about 1% to about 4% by weight of talc. In some embodiments, the composition comprises about 2% by weight of talc. In some embodiments, the composition comprises from about 0.2% to about 2.5% by weight of triethyl citrate. In some embodiments, the composition comprises from about 0.5% to about 2% by weight of triethyl citrate. In some embodiments, the composition comprises about 1% by weight of triethyl citrate.
  • the composition comprises: about 67% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form; about 11% by weight of microcrystalline cellulose; about 9% by weight of hypromellose; about 10% by weight of methacrylic acid copolymer dispersion; about 2% by weight of talc; and about 1% by weight of triethyl citrate.
  • composition comprising: a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • the composition is in the form of granules.
  • the granules will not pass through a 40 mesh screen.
  • the granules are coated with hypromellose.
  • the coated granules are further coated with a composition comprising: methacrylic acid copolymer dispersion; talc; and triethyl citrate.
  • the composition is a unitary dosage form. In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule.
  • composition comprising: about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule.
  • not less than about 85% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form is released within 30 mins; and not less than about 90% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form is released within 45 mins; as measured using United States Pharmacopoeia ⁇ 711> method
  • Disclosed herein, in certain embodiments is a, using Disclosed herein, in certain embodiments, is apparatus 2 operating at 50 rpm in 900 mL of dissolution medium at pH 6.8 at 37°C.
  • a composition comprising: a compound or
  • the diluent is microcrystalline cellulose.
  • the microcrystalline cellulose comprises from about 40% to about 60% by weight of the composition.
  • the microcrystalline cellulose comprises about 50% by weight of the composition.
  • the disintegrant is croscarmellose sodium.
  • the croscarmellose sodium comprises from about 0.1% to about 2% by weight of the composition.
  • the croscarmellose sodium comprises from about 0.5% by weight of the composition.
  • the composition comprises: about 50% by weight microcrystalline cellulose; and about 0.5% by weight croscarmellose sodium.
  • M is hydrogen and/or potassium. In some embodiments, M is potassium. In some embodiments, the composition comprises at least 50% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 60% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 70% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 80% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises: about 50% by weight of a compound or mixture of compounds of structure (I); about 49.4% by weight of microcrystalline cellulose; and about 0.55% by weight of croscarmellose sodium.
  • the composition is a unitary dosage form. In some embodiments, the composition is encapsulated within a capsule. In some embodiments, the capsule comprises hard gelatin. In some embodiments, the hard gelatin capsule is over-encapsulated. In some embodiments, the hard gelatin capsule is over-encapsulated within a hypromellose capsule. [0018] Disclosed herein, in certain embodiments, is a composition comprising: about lOOmg of a
  • the composition is encapsulated.
  • composition comprising: about 106.8mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid
  • composition comprising: about 213mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • composition comprising: about 430mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • composition comprising: about 860mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • composition comprising: about lOOOmg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
  • the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule. In some embodiments, the hard gelatin capsule is over-encapsulated. In some embodiments, the hard gelatin capsule is over- encapsulated within a hypromellose capsule. In some embodiments, not less than about 80% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 30 minutes, as measured using United States Pharmacopoeia A apparatus 1 operating at 75 rpm in 900 mL water at 37°C.
  • the in vitro dissolution rate as measured using United States Pharmacopoeia A apparatus 1 operating at 75 rpm in 900 mL water at 37°C is not less than about 80% of the compound of structure (IB) is released within 30 minutes.
  • not less than about 78% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 30 minutes; and not less than about 95% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 45 minutes; as measured using United States Pharmacopoeia
  • apparatus 1 operating at 75 rpm in 900 mL water at 37°C.
  • a composition comprising: a compound of
  • M is hydrogen, sodium, potassium, calcium, or arginine; a diluent; a binder; a lubricant; and a coating.
  • the composition is a unitary dosage form.
  • the composition is a monolithic solid. [0026] Disclosed herein, in certain embodiments, is a composition comprising: about 200mg of a
  • composition comprising: from about 60% to about 90% by weight of a compound of structure (IB) or a mixture of a compound of structure
  • (IB) and its free acid form from about 5% to about 15% by weight of microcrystalline cellulose; from about 2.5% to about 10% by weight of acryl-eze white from about 2.5% to about 10% by weight of hypromellose; from about 0.25% to about 2% by weight of magnesium stearate.
  • composition comprising: about 213.6mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid
  • the granules are blended with magnesium stearate.
  • the composition is compressed into a monolithic solid.
  • the composition is coated with hypromellose.
  • the composition is further coated with acryl-eze white.
  • the in vitro dissolution rate as measured using United States Pharmacopoeia A apparatus 2 operating at 50 rpm in 700 mL of dissolution medium for two hours at pH 1.2 at 37°C is about 0% to about 5% of the compound of structure (IB) released within 2 hours; and after 2 hours, in 900 mL of buffer at pH 6.8 at 37°C is about 15% to about 45% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 30 mins; is about 50% to about 85% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 45 mins; is not less than about 80% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 60 mins; is not less than about 90% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 90
  • a method for decreasing uric acid levels in one or more tissues or organs of a subject in need of decreased uric acid levels comprising administering to the subject a uric acid level decreasing amount of a composition disclosed herein.
  • Disclosed herein is a method for reducing uric acid production, increasing uric acid excretion or both in a subject, comprising administering to the subject an effective amount of a composition disclosed herein.
  • Disclosed herein, in certain embodiments, is a method for treating or preventing hyperuricemia in a subject comprising administering to the subject an effective amount of a composition disclosed herein.
  • a method of treating a subject suffering from a condition characterized by abnormal tissue or organ levels of uric acid comprising administering to the subject an effective amount of a composition disclosed herein.
  • the condition is selected from gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosis.
  • a method for preventing a condition characterized by abnormal tissue levels of uric acid in a subject at increased risk of developing the condition comprising administering to the subject an effective amount of a composition disclosed herein.
  • a method for treating gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosisin a subject comprising administering to the subject an effective amount of a composition disclosed herein.
  • Disclosed herein, in certain embodiments, is a method for treating gout in a subject comprising administering to the subject an effective amount of a composition disclosed herein.
  • a method for preventing the formation or reducing the size of tophi/tophus in a subject comprising administering to the subject an effective amount of a composition disclosed herein.
  • the AUC ⁇ ( ⁇ g » hr/mL) of a composition disclosed herein is between about 0.6 and about 18. In some embodiments, the AUC ⁇ ( ⁇ g»hr/mL) of a composition disclosed herein is between about 2.5 and about 11. In some embodiments, the AUC ⁇ ( ⁇ g » hr/mL) of a composition disclosed herein is between about 4.8 and about 8. In some embodiments, the Cmax ( ⁇ g/mL) of a composition disclosed herein is between about 0.15 and about 6. In some embodiments, the Cmax ( ⁇ g/mL) of a composition disclosed herein is between about 0.5 and about 5. In some embodiments, the Cmax ( ⁇ g/mL) of a composition disclosed herein is between about 1 and about 4.
  • a composition disclosed herein provides the metabolite M6 (2-(5- bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetic acid).
  • a composition disclosed herein provides an AUC ⁇ ( ⁇ g » hr/mL) of metabolite M6 between about 2.5 and about 30.
  • a composition disclosed herein provides an AUC ⁇ ( ⁇ g » hr/mL) of metabolite M6 between about 8 and about 25.
  • a composition disclosed herein provides an AUC ⁇ ( ( ⁇ g » hr/mL) of metabolite M6 between about 12 and about 20.
  • the Cmax ( ⁇ g/mL) of metabolite M6 is between about 0.25 and about 4.
  • the Cmax ( ⁇ g/mL) of metabolite M6 is between about 1 and about 3.
  • the Cmax ( ⁇ g/mL) of metabolite M6 is between about 2.4 and about 3.
  • the molar ratio of the AUC ⁇ ( ⁇ g » hr/mL) of the metabolite M6 to the AUC ⁇ ( ⁇ g » hr/mL) of Compound I is about 2 to about 11.
  • Compound I is about 4 to about 8. In some embodiments, the molar ratio of the Cmax ( ⁇ g/mL) of the metabolite M6 to the Cmax ( ⁇ g/mL) of Compound I is about is about 1 to about 7. In some embodiments, the molar ratio of the Cmax ( ⁇ g/mL) of the metabolite M6 to the Cmax ( ⁇ g/mL) of Compound I is about is about 2 to about 6.
  • Figure 2 shows a flow chart depicting the steps for the preparation of the composition described in example 3A.
  • Figure 3 shows a flow chart depicting the steps for the preparation of the composition described in example 4A.
  • Figure 4 shows a flow chart depicting the steps for the preparation of the composition described in example 5A.
  • Figure 5 shows Serum Uric Acid Levels (measured on Day 9), after administration of compositions comprising compound 1. DETAILED DESCRIPTION OF THE INVENTION
  • compositions comprising 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof.
  • the pharmaceutical compositions comprise an effective amount of 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof.
  • the pharmaceutical compositions comprise an effective amount of 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical compositions are for the treatment of disorders.
  • the pharmaceutical compositions are for the treatment of disorders in a mammal.
  • the pharmaceutical compositions are for the treatment of disorders in a human.
  • the method includes administering an effective amount of a composition as described herein to a subject with aberrant levels of uric acid such as to restore acceptable or non-aberrant levels of uric acid.
  • the present invention also provides methods useful for decreasing uric acid levels in one or more tissues or organs of a subject in need of decreased uric acid levels, comprising administering to the subject a uric acid level decreasing amount of a composition as described herein.
  • the present invention also provides methods useful for reducing uric acid production, increasing uric acid excretion or both in a subject, comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for treating or preventing hyperuricemia in a subject comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for treating a subject suffering from a condition characterized by abnormal tissue or organ levels of uric acid comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for treating a subject suffering from gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosis, comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for preventing a condition characterized by abnormal tissue levels of uric acid in a subject at increased risk of developing the condition, comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for treating gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley- Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosisin a subject comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for treating gout in a subject comprising administering to the subject an effective amount of a composition as described herein.
  • the present invention also provides methods useful for preventing the formation or reducing the size of tophi/tophus in a subject, comprising administering to the subject an effective amount of a composition as described herein.
  • Terminology [0050] The term "subject”, “patient” or “individual” as used herein in reference to individuals suffering from a disorder, and the like, encompasses mammals and non-mammals.
  • mammals include, but are not limited to, any member of the Mammalian class: humans, non- human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.
  • non- mammals include, but are not limited to, birds, fish and the like.
  • the mammal is a human.
  • an “effective amount” refers to an amount of at least one agent or compound being administered that is sufficient to treat or prevent the particular disease or condition. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in a disease.
  • An appropriate “effective” amount in any individual case may be determined using techniques, such as a dose escalation study.
  • pharmaceutically acceptable refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compounds described herein, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • composition and “pharmaceutical composition,” as used herein, refer to 4-(2- (5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof, optionally mixed with at least one pharmaceutically acceptable chemical component, such as, though not limited to carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients and the like.
  • pharmaceutically acceptable chemical component such as, though not limited to carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients and the like.
  • pharmaceutically acceptable salt includes salts of 4-(2-(5- bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoate with any pharmaceutically acceptable cation, that retain the biological effectiveness of the free acid.
  • the free acid group of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H- l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid may react with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • bases include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N + (C 1-4 alkyl)4, and the like.
  • Representative organic amines useful for the formation of base addition salts include arginine, lysine, ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. Salts can be prepared in situ during final isolation and purification, or by separate reaction and isolation of the salt thus formed.
  • the term "prodrug” as used herein, refers to a drug precursor that, following administration to a subject and subsequent absorption, is converted to an active, or a more active species via some process, such as conversion by a metabolic pathway.
  • the term encompasses any derivative of a compound, which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or a pharmaceutically active metabolite or residue thereof.
  • Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not.
  • Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g. by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g. the brain or lymphatic system).
  • the amount will normally be determined by the prescribing physician with the dosage generally varying according to the age, sex, diet, weight, general health and response of the individual patient, the severity of the patient's symptoms, the precise indication or condition being treated, the severity of the indication or condition being treated, time of administration, route of administration, the disposition of the composition, rate of excretion, drug combination, and the discretion of the prescribing physician.
  • the pharmaceutical composition may be in unit dosage form. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose. Determination of the proper dosage for a particular situation is within the skill of the art.
  • the total daily dosage may be divided and administered in portions during the day if desired.
  • the amount and frequency of administration, and if applicable other therapeutic agents and/or therapies, will be regulated according to the judgment of the attending clinician (physician) considering such factors as described above.
  • Administration may occur in an amount of between about 0.001 mg/kg of body weight to about 100 mg/kg of body weight per day (administered in single or divided doses), or at least about 0.1 mg/kg of body weight per day.
  • a particular therapeutic dosage includes, e.g., from about 0.01 mg to about 7000 mg of compound, or, e.g., from about 0.05 mg to about 2500 mg.
  • the quantity of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof in a unit dose may occur in an amount of between about lmg to 3000 mg, from about 2 mg to 2000 mg, or 10 mg to 2000 mg, according to the particular application. In some embodiments, the amount is from about lOOmg to about 1500mg, from about 150mg to about 1200mg, or from about 200mg to about lOOOmg.
  • the amount is at least lOOmg, at least 200mg, at least 250mg, at least 300mg, at least 400mg, at least 500mg, at least 600mg, at least 700mg, at least 750mg, at least 800mg, at least 900mg or at least lOOOmg. In further or additional embodiments, the amount is about lOOmg, about 200mg, about 250mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 750mg, about 800mg, about 900mg or about lOOOmg. In some embodiments the compositions are administered once daily.
  • compositions are administered twice daily. In further or additional embodiments, the compositions are administered at least twice daily. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day. In combinational applications in which the compound is not the sole therapy, it may be possible to administer lesser amounts of compound and still have therapeutic or prophylactic effect.
  • compositions described herein are typically useful for oral administration as solid dosage forms, such as tablets, capsules, pills, powders, granules and the like. They may be administered for immediate release, delayed release or sustained release of 4- (2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or a pharmaceutically acceptable salts thereof. Kits
  • kits for the treatment of diseases and disorders comprise a compositions described herein in a container and, optionally, instructions teaching the use of the kit.
  • kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider.
  • Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • compositions described herein are also useful for diagnostics and as research reagents.
  • the compounds described herein may be useful as tools in differential and/or combinatorial analyses to elucidate expression patterns of genes expressed within cells and tissues.
  • expression patterns within cells or tissues treated with one or more compounds are compared to control cells or tissues not treated with compounds and the patterns produced are analyzed for differential levels of gene expression as they pertain, for example, to disease association, signaling pathway, cellular localization, expression level, size, structure or function of the genes examined. These analyses can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds which affect expression patterns.
  • the compositions described herein may also be useful for veterinary treatment of other animals.
  • Example 2B Identification of compound 1 in capsules prepared in example 2A
  • An isocratic reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase of 45% 10 mM KH 2 PO 4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the identity of compound 1.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water.
  • the identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 0.97 - 1.03 relative to reference standard, confirming the presence of compound 1.
  • Example 2C Determination of quantity of compound 1 in capsules prepared in example 2A
  • An isocratic reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column,; mobile phase of 45% 10 mM KH 2 PO 4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water. The quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly.
  • Example 2D Determination of impurities in capsules prepared in example 2A
  • a gradient elution reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase A lO mM KH 2 PO 4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water.
  • Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly.
  • Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
  • Example 2E Determination of water content in capsules prepared in example 2 A
  • Example 2F Dissolution Profile of compound 1 from capsules prepared in example 2A
  • Dissolution of compound 1 from the capsules was determined in 900 mL of water at 37°C using USP Apparatus 1 operating at 75 rpm.
  • a filtered sample of the dissolution medium was taken at the specified time(s) and analyzed by an HPLC procedure using the same chromatographic conditions as for content determination, as described above. The release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
  • Vcaps caps were separated from the capsule bodies. A size # 2 dark green opaque hard gelatin
  • Coni-Snap capsule (prepared as described in example 2A above) was placed into each of the
  • Example 3B Identification of compound 1 in capsules prepared in example 3 A
  • An isocratic reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase of 45% 10 mM KH 2 PO 4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the of identity of compound 1.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water.
  • the identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 0.97 - 1.03 relative to reference standard, confirming the presence of compound 1.
  • Example 3C Determination of quantity of compound 1 in capsules prepared in example 3 A
  • An isocratic reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column,; mobile phase of 45% 10 mM KH 2 PO 4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water.
  • the quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly and is shown below.
  • Example 3D Determination of impurities in capsules prepared in example 3 A
  • a gradient elution reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase A lO mM KH 2 PO 4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content.
  • the capsules were extracted with 50/50 (v/v) acetonitrile/water.
  • Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly.
  • Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
  • Example 3E Determination of water content in capsules prepared in example 3 A
  • Example 3F Dissolution Profile of compound 1 from capsules prepared in example 3 A
  • Dissolution of compound 1 from the capsules was determined in 900 mL of water at 37°C using USP Apparatus 1 operating at 75 rpm.
  • a filtered sample of the dissolution medium was taken at the specified times (15, 30, 45 and 60 minutes) and analyzed by HPLC using the same chromatographic conditions as described above for content determination.
  • Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
  • Batch number 1 2 Batch number 1 2
  • Example 4B Identification of compound 1 in capsules prepared in example 4A
  • Example 4C Determination of quantity of compound 1 in capsules prepared in example
  • Example 4D Determination of impurities in capsules prepared in example 4A
  • a gradient elution reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase A lO mM KH 2 PO 4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content.
  • the capsules were extracted with 20/80 (v/v) methanol/phosphate buffer (pH 7.4).
  • Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly.
  • Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
  • Example 4E Determination of water content in capsules prepared in example 4 A [0088] Water content was determined using the USP ⁇ 921>, Karl Fischer method and measured at 6.9%.
  • Example 4F Dissolution Profile of compound 1 from capsules prepared in example 4A
  • Dissolution of compound 1 from the capsules was determined following the principles of the USP ⁇ 711> method A for delayed-release dosage forms, using USP Apparatus 2 at 37°C, set at 50 rpm. Acid stage was performed for 2 hours in 700 mL of dissolution medium at pH 1.2 followed by buffer stage performed in 900 mL of dissolution medium at pH 6.8. A filtered aliquot of the test dissolution medium was taken at the specified times (15, 30, 45 and 60 minutes) and analyzed by HPLC using the same chromatographic conditions as described above for content determination. Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
  • a 7% w/w coating solution of hypromellose 2910, in purified water was prepared using a lab mixer and applied to the compressed tablets using a Vector LCDS-3 coating system to achieve an approximate 3% weight gain ( ⁇ 8 mg/tablet).
  • An 18% w/w enteric coating suspension of Acryl-Eze White (methacrylic acid co-polymer) in purified water was prepared using a lab mixer, and the suspension applied to the previously coated tablets using the Vector LCDS-3 coating system to achieve an approximate 10% weight gain ( ⁇ 27 mg/tablet).
  • the isolated tablets were white, caplet-shaped tablets approximately 0.2" x 0.43" in dimension, weighing approximately 298 mg each and containing 200mg of compound 1 (present as the potassium salt).
  • One batch of 1700 capsules was prepared according to this procedure. The capsules were analyzed for identity, strength, purity, content uniformity and dissolution profiles, as described below. Unit Composition
  • Example 5C Determination of quantity of compound 1 in capsules prepared in example 5 A
  • An isocratic reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column,; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present.
  • the capsules were extracted with extracted with 20:80 (v/v) methanol/phosphate buffer (pH 7.4) and diluted 1 :10 with methanol/water (20:80 v/v ratio).
  • the target concentration of compound 1 for assay is 0.1 mg/mL.
  • the quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly, and confirmed the quantity to be 102.8%, within the 90.0 - 110.0 % limit set.
  • Example 5D Determination of impurities in capsules prepared in example 5 A
  • a gradient elution reversed-phase HPLC method (4.6 x 150 mm YMC ODS AQ, 3 ⁇ m size particles column; mobile phase A lO mM KH 2 PO 4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content.
  • the capsules were extracted with 20/80 (v/v) methanol/phosphate buffer (pH 7.4).
  • Target concentration of compound 1 for impurities assay was 1 mg/mL.
  • Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly. Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
  • Example 5E Determination of water content in capsules prepared in example 5 A
  • Example 5F Dissolution Profile of compound 1 from capsules prepared in example 5 A
  • Dissolution of compound 1 from the capsules was determined following the principles of the USP ⁇ 711> method A for delayed-release dosage forms, using USP Apparatus 2 at 37°C, set at 50 rpm. Acid stage was performed for 2 hours in 700 mL of dissolution medium at pH 1.2 followed by buffer stage performed in 900 mL of dissolution medium at pH 6.8. A filtered aliquot of the test dissolution medium was taken at the specified times (30, 45, 60, 90 and 100 minutes) and analyzed by HPLC using isocratic elution and UV detection at 226 nm (same chromatographic conditions as described above for content determination). Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
  • compositions were administered over a 7-day treatment period plus one morning dose for PK purposes on day 8. (It should be noted that in some instances multiple doses of the same composition were administered, in order to achieve the required total dose of compound 1; i.e. in order to achieve a dose of 400mg compound 1, a patient may have taken four lOOmg compositions or two 200mg compositions, depending on the dosage form.)
  • Four sequential dose cohorts were administered as follows:
  • Serum uric acid levels were recorded on day 9 of the study, and are shown in the table below and graphically in figure 5.
  • Example 7 [0099] Additional human clinical studies were undertaken to further investigate the PK properties of the compositions described herein. The results are summarized in the table below.
  • M6 is 2-(5-bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetic acid - the predominant metabolite of Compound 1

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to compositions comprising 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H- 1,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid or pharmaceutically acceptable salts thereof, and to the preparation and use of such compositions, in particular for the treatment of diseases.

Description

COMPOSITIONS COMPRISING
4- (2- ( 5-BROMO-4- ( l-CYCLOPROPYLNAPHTHALEN-4-YL) -4H-1 , 2 , 4-TRIAZ0L-3-YLTHI0)
ACETAMIDO ) - S -CHLOROBENZOI C ACID AND PHARMACEUTICALLY ACCEPTABLE SALTS THEREOF ,
RELATED APPLICATION
[0001] This application claims priority to U.S. Provisional Application No.61/108,412filed October 24, 2008, which is incorporated herein by reference in its entirety. BACKGROUND OF THE INVENTION
[0002] 4-(2-(5-Bromo-4-(l-cyclopropylnaphthalen-4-yl)-4/f-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof, are useful in the treatment and prevention of diseases. Described herein are compositions comprising 4-(2-(5- bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof and methods of using the compositions, for example in the treatment of diseases.
Figure imgf000002_0001
SUMMARY OF THE INVENTION
[0003] Disclosed herein, in certain embodiments, is a composition comprising greater than about 35% by weight of a compound or mixture of compounds of structure (I):
Figure imgf000002_0002
(I); wherein M is hydrogen, sodium, potassium, calcium or arginine. In some embodiments, the composition comprises greater than about 60% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises greater than about 70% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises greater than about 80% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least lOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 250mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 500mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition
- 1 -
WSGR Docket No 18545-763.601 comprises at least 750mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 800mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOmg, about 200mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 800mg, about 900mg or about lOOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 600mg, about 800mg or about lOOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 600mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 800mg of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOOmg of a compound or mixture of compounds of structure (I). In some embodiments, the composition further comprises one or more pharmaceutically acceptable diluents, binders, lubricants, coating agents, barrier coatings, plasticizers, dispersing agents or film coating additives. In some embodiments, the composition further comprises one or more pharmaceutically acceptable diluents. In some embodiments, the diluent is microcrystalline cellulose, silicified microcrystalline cellulose, cellulose, lactose, compressible sugar, mannitol, calcium silicate and calcium phosphate, sodium phosphate, sodium carbonate, or a combination thereof. In some embodiments, the diluent is in powder form. In some embodiments, the diluent is in granular form. In some embodiments, the diluent is microcrystalline cellulose. In some embodiments, the composition further comprises one or more pharmaceutically acceptable binders. In some embodiments, the binder is hypromellose, povidone, hydroxypropyl cellulose, hydroxyethyl cellulose or starch. In some embodiments, the binder is hypromellose. In some embodiments, the composition further comprises one or more pharmaceutically acceptable lubricant. In some embodiments, the lubricant is magnesium stearate, stearic acid or sodium stearyl fumarate. In some embodiments, the lubricant is magnesium stearate. In some embodiments, the composition further comprises one or more pharmaceutically acceptable barrier coatings. In some embodiments, the pharmaceutically acceptable barrier coating is a pharmaceutically acceptable enteric coating. In some embodiments, the barrier coating is hypromellose or polyvinyl alcohol. In some embodiments, the barrier coating is hypromellose. In some embodiments, the composition further comprises one or more pharmaceutically acceptable coating agents. In some embodiments, the coating agent is a methacrylic acid based copolymer, Eudragit L30D55, Acryl- Eze, hydroxypropylmethyl cellulose acetate succinate, polyvinylacetate phthalate, cellulose acetate phthalate or a combination thereof. In some embodiments, the coating agent is a methacrylic acid based copolymer, Eudragit L30D55 or Acryl-Eze. In some embodiments, the composition further comprises one or more pharmaceutically acceptable plasticizers. In some
- 7 - embodiments, the plasticizer is triethyl citrate, triacetin, dibutyl phthalate, diethyl phthalate or glycerin. In some embodiments, the plasticizer is triethyl citrate. In some embodiments, the composition further comprises one or more pharmaceutically acceptable film coating additives. In some embodiments, the film coating additive is talc, glycerol monostearate or colloidal silicon dioxide. In some embodiments, the film coating additive is talc. In some embodiments, the composition further comprises one or more pharmaceutically active compounds. In some embodiments, at least one of the one or more pharmaceutically active compounds has anti-viral activity. In some embodiments, the composition further comprises two pharmaceutically active compounds. In some embodiments, at least one of the two pharmaceutically active compounds has anti-viral activity. In some embodiments, the composition is a unitary dosage form. In some embodiments, M is hydrogen or potassium. In some embodiments, M is potassium. In some embodiments, the composition is suitable for oral administration to a mammal. In some embodiments, the composition is suitable for oral administration to a human. [0004] Disclosed herein, in certain embodiments, is a composition comprising: a compound or
mixture of compounds of structure (I):
Figure imgf000004_0001
(I); wherein M is hydrogen, sodium, potassium, calcium, or arginine; and one or more diluents; one or more binders; one or more coating agents; one or more dispersing agents; and one or more plasticizers. In some embodiments, the composition is a unitary dosage form. [0005] In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule. In some embodiments, M is hydrogen or potassium. In some embodiments, M is potassium. In some embodiments, the diluent is microcrystalline cellulose, silicified microcrystalline cellulose, cellulose, lactose, compressible sugar, mannitol, calcium silicate and calcium phosphate in powder and granular forms, sodium phosphate or sodium carbonate. In some embodiments, the diluent is microcrystalline cellulose. In some embodiments, the binder is hypromellose, povidone, hydroxypropyl cellulose, hydroxyethyl cellulose or starch. In some embodiments, the binder is hypromellose. In some embodiments, the coating agent is a methacrylic acid based copolymers, Eudragit L30D55, Acryl-Eze, hydroxypropylmethyl cellulose acetate succinate, polyvinylacetate phthalate or cellulose acetate phthalate. In some embodiments, the coating agent is a methacrylic acid based copolymer. In some embodiments, the dispersing agent is talc, glycerol monostearate or colloidal silicon dioxide. In some embodiments, the dispersing agent is talc. In some embodiments, the plasticizer is triethyl citrate, triacetin, dibutyl phthalate, diethyl phthalate or glycerin. In some embodiments, the plasticizer is triethyl citrate. In some embodiments, the composition comprises from about lmg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about lOmg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 50mg to about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 200mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 300mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 400mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 500mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 600mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 700mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 800mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about 900mg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises about lOOOmg of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 10% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 25% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 50% to about 90% by weight of the compound or mixture of compounds of structure (I). In some embodiments, the composition comprises from about 65% to about 90% by weight of the compound or mixture of compounds of structure (I).
[0006] Disclosed herein, in certain embodiments, is a composition comprising: a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000005_0001
(IB); microcrystalline cellulose; hypromellose; methacrylic acid copolymer dispersion; talc; and triethyl citrate. [0007] In some embodiments, the composition comprises: about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000006_0001
(IB); about 35mg of microcrystalline cellulose; about 29mg of hypromellose; about 30mg of methacrylic acid copolymer dispersion; about 6mg of talc; and about 3mg of triethyl citrate. In some embodiments, the composition is a unitary dosage form. In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule.
[0008] Disclosed herein, in certain embodiments, is a composition comprising: from about 60% to about 90% by weight of a compound of structure (IB) or a mixture of a compound of structure
(IB) and its free acid form:
Figure imgf000006_0002
(IB); from about 5% to about 15% by weight of microcrystalline cellulose; from about 5% to about 15% by weight of hypromellose; from about 5% to about 15% by weight of methacrylic acid copolymer dispersion; from about 0.5% to about 5% by weight of talc; and from about 0.1% to about 3% by weight of triethyl citrate.
[0009] In some embodiments, the composition comprises from about 60% to about 80% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 60% to about 75% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 65% to about 70% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 67% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form. In some embodiments, the composition comprises from about 7% to about 13% by weight of microcrystalline cellulose. In some embodiments, the composition comprises about 11% by weight of microcrystalline cellulose. In some embodiments, the composition comprises from about 7% to about 11% by weight of hypromellose. In some embodiments, the composition comprises about 9% by weight of hypromellose. In some embodiments, the composition comprises from about 5% to about 15% by weight of methacrylic acid copolymer dispersion. In some embodiments, the composition comprises from about 8% to about 12% by weight of methacrylic acid copolymer dispersion. In some embodiments, the composition comprises about 10% by weight of methacrylic acid copolymer dispersion. In some embodiments, the composition comprises from about 1% to about 4% by weight of talc. In some embodiments, the composition comprises about 2% by weight of talc. In some embodiments, the composition comprises from about 0.2% to about 2.5% by weight of triethyl citrate. In some embodiments, the composition comprises from about 0.5% to about 2% by weight of triethyl citrate. In some embodiments, the composition comprises about 1% by weight of triethyl citrate. In some embodiments, the composition comprises: about 67% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form; about 11% by weight of microcrystalline cellulose; about 9% by weight of hypromellose; about 10% by weight of methacrylic acid copolymer dispersion; about 2% by weight of talc; and about 1% by weight of triethyl citrate.
[0010] Disclosed herein, in certain embodiments, is a composition comprising: a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000007_0001
(IB); microcrystalline cellulose; and hypromellose; wherein the composition is in the form of granules. In some embodiments, the granules will not pass through a 40 mesh screen. In some embodiments, the granules are coated with hypromellose. In some embodiments, the coated granules are further coated with a composition comprising: methacrylic acid copolymer dispersion; talc; and triethyl citrate.
[0011] In some embodiments, the composition is a unitary dosage form. In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule.
[0012] Disclosed herein, in certain embodiments, is a composition comprising: about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000007_0002
(IB); about 35mg of microcrystalline cellulose; about 13.5 mg of hypromellose; wherein the composition is in the form of granules which do not pass through a 40 mesh screen; and wherein the granules are coated with about 15.3 mg hypromellose; and wherein the coated granules are further coated with a composition comprising: about 30.4mg of methacrylic acid copolymer dispersion; about 6.1mg of talc; and about 3.0mg of triethyl citrate. [0013] In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule. In some embodiments, not less than about 85% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form is released within 30 mins; and not less than about 90% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form is released within 45 mins; as measured using United States Pharmacopoeia <711> method Disclosed herein, in certain embodiments, is a, using Disclosed herein, in certain embodiments, is apparatus 2 operating at 50 rpm in 900 mL of dissolution medium at pH 6.8 at 37°C. [0014] Disclosed herein, in certain embodiments, is a composition comprising: a compound or
mixture of compounds of structure (I):
Figure imgf000008_0001
(I); wherein M is hydrogen, sodium, potassium, calcium, or arginine; a diluent; and a disintegrant.
[0015] In some embodiments, the diluent is microcrystalline cellulose. In some embodiments, the microcrystalline cellulose comprises from about 40% to about 60% by weight of the composition. In some embodiments, the microcrystalline cellulose comprises about 50% by weight of the composition. In some embodiments, the disintegrant is croscarmellose sodium. In some embodiments, the croscarmellose sodium comprises from about 0.1% to about 2% by weight of the composition. In some embodiments, the croscarmellose sodium comprises from about 0.5% by weight of the composition. In some embodiments, the composition comprises: about 50% by weight microcrystalline cellulose; and about 0.5% by weight croscarmellose sodium.
[0016] In some embodiments, M is hydrogen and/or potassium. In some embodiments, M is potassium. In some embodiments, the composition comprises at least 50% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 60% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 70% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises at least 80% by weight of a compound or mixture of compounds of structure (I). In some embodiments, the composition comprises: about 50% by weight of a compound or mixture of compounds of structure (I); about 49.4% by weight of microcrystalline cellulose; and about 0.55% by weight of croscarmellose sodium.
[0017] In some embodiments, the composition is a unitary dosage form. In some embodiments, the composition is encapsulated within a capsule. In some embodiments, the capsule comprises hard gelatin. In some embodiments, the hard gelatin capsule is over-encapsulated. In some embodiments, the hard gelatin capsule is over-encapsulated within a hypromellose capsule. [0018] Disclosed herein, in certain embodiments, is a composition comprising: about lOOmg of a
compound of structure
Figure imgf000009_0001
about 98.9mg of microcrystalline cellulose; and about l.lmg of croscarmellose sodium. In some embodiments, the composition is encapsulated.
[0019] Disclosed herein, in certain embodiments, is a composition comprising: about 106.8mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid
form:
Figure imgf000009_0002
(IB); about 92.1mg of microcrystalline cellulose; and about l.lmg of croscarmellose sodium.
[0020] Disclosed herein, in certain embodiments, is a composition comprising: about 213mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000009_0003
(IB); microcrystalline cellulose; and croscarmellose sodium. [0021] Disclosed herein, in certain embodiments, is a composition comprising: about 430mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000009_0004
(IB); microcrystalline cellulose; and croscarmellose sodium. [0022] Disclosed herein, in certain embodiments, is a composition comprising: about 860mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000009_0005
(IB); microcrystalline cellulose; and croscarmellose sodium. [0023] Disclosed herein, in certain embodiments, is a composition comprising: about lOOOmg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000010_0001
(IB); microcrystalline cellulose; and croscarmellose sodium. [0024] In some embodiments, the composition is encapsulated. In some embodiments, the composition is encapsulated within a hard gelatin capsule. In some embodiments, the hard gelatin capsule is over-encapsulated. In some embodiments, the hard gelatin capsule is over- encapsulated within a hypromellose capsule. In some embodiments, not less than about 80% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 30 minutes, as measured using United States Pharmacopoeia A apparatus 1 operating at 75 rpm in 900 mL water at 37°C. In some embodiments, the in vitro dissolution rate as measured using United States Pharmacopoeia A apparatus 1 operating at 75 rpm in 900 mL water at 37°C is not less than about 80% of the compound of structure (IB) is released within 30 minutes. In some embodiments, not less than about 78% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 30 minutes; and not less than about 95% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form dissolves within 45 minutes; as measured using United States Pharmacopoeia Disclosed herein, in certain embodiments, is apparatus 1 operating at 75 rpm in 900 mL water at 37°C. [0025] Disclosed herein, in certain embodiments, is a composition comprising: a compound of
structure (I) or a mixture of compounds of structure (I):
Figure imgf000010_0002
(I); wherein
M is hydrogen, sodium, potassium, calcium, or arginine; a diluent; a binder; a lubricant; and a coating. In some embodiments, the composition is a unitary dosage form. In some embodiments, the composition is a monolithic solid. [0026] Disclosed herein, in certain embodiments, is a composition comprising: about 200mg of a
compound of structure
Figure imgf000010_0003
[0027] Disclosed herein, in certain embodiments, is a composition comprising: from about 60% to about 90% by weight of a compound of structure (IB) or a mixture of a compound of structure
(IB) and its free acid form:
Figure imgf000011_0001
from about 5% to about 15% by weight of microcrystalline cellulose; from about 2.5% to about 10% by weight of acryl-eze white from about 2.5% to about 10% by weight of hypromellose; from about 0.25% to about 2% by weight of magnesium stearate.
[0028] Disclosed herein, in certain embodiments, is a composition comprising: about 213.6mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid
form:
Figure imgf000011_0002
about 34.0mg of microcrystalline cellulose; about 23.1mg of hypromellose; about 1.3mg of magnesium stearate; and about 27.1mg of acryl-eze white.
[0029] In some embodiments, the granules are blended with magnesium stearate. In some embodiments, the composition is compressed into a monolithic solid. In some embodiments, the composition is coated with hypromellose. In some embodiments, the composition is further coated with acryl-eze white. In some embodiments, the in vitro dissolution rate as measured using United States Pharmacopoeia A apparatus 2 operating at 50 rpm in 700 mL of dissolution medium for two hours at pH 1.2 at 37°C is about 0% to about 5% of the compound of structure (IB) released within 2 hours; and after 2 hours, in 900 mL of buffer at pH 6.8 at 37°C is about 15% to about 45% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 30 mins; is about 50% to about 85% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 45 mins; is not less than about 80% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 60 mins; is not less than about 90% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 90 mins; and is not less than about 95% of the compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form released within 100 mins.
[0030] Disclosed herein, in certain embodiments, is a method for decreasing uric acid levels in one or more tissues or organs of a subject in need of decreased uric acid levels, comprising administering to the subject a uric acid level decreasing amount of a composition disclosed herein.
[0031] Disclosed herein, in certain embodiments, is a method for reducing uric acid production, increasing uric acid excretion or both in a subject, comprising administering to the subject an effective amount of a composition disclosed herein.
[0032] Disclosed herein, in certain embodiments, is a method for treating or preventing hyperuricemia in a subject comprising administering to the subject an effective amount of a composition disclosed herein. [0033] Disclosed herein, in certain embodiments, is a method of treating a subject suffering from a condition characterized by abnormal tissue or organ levels of uric acid comprising administering to the subject an effective amount of a composition disclosed herein. In some embodiments, the condition is selected from gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosis. [0034] Disclosed herein, in certain embodiments, is a method for preventing a condition characterized by abnormal tissue levels of uric acid in a subject at increased risk of developing the condition, comprising administering to the subject an effective amount of a composition disclosed herein. [0035] Disclosed herein, in certain embodiments, is a method for treating gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosisin a subject comprising administering to the subject an effective amount of a composition disclosed herein.
[0036] Disclosed herein, in certain embodiments, is a method for treating gout in a subject comprising administering to the subject an effective amount of a composition disclosed herein. [0037] Disclosed herein, in certain embodiments, is a method for preventing the formation or reducing the size of tophi/tophus in a subject, comprising administering to the subject an effective amount of a composition disclosed herein.
[0038] In some embodiments, the AUCτ (μg»hr/mL) of a composition disclosed herein is between about 0.6 and about 18. In some embodiments, the AUCτ (μg»hr/mL) of a composition disclosed herein is between about 2.5 and about 11. In some embodiments, the AUCτ (μg»hr/mL) of a composition disclosed herein is between about 4.8 and about 8. In some embodiments, the Cmax (μg/mL) of a composition disclosed herein is between about 0.15 and about 6. In some embodiments, the Cmax (μg/mL) of a composition disclosed herein is between about 0.5 and about 5. In some embodiments, the Cmax (μg/mL) of a composition disclosed herein is between about 1 and about 4.
[0039] In some embodiments, a composition disclosed herein provides the metabolite M6 (2-(5- bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetic acid). In some embodiments, a composition disclosed herein provides an AUCτ (μg»hr/mL) of metabolite M6 between about 2.5 and about 30. In some embodiments, a composition disclosed herein provides an AUCτ (μg»hr/mL) of metabolite M6 between about 8 and about 25. In some embodiments, a composition disclosed herein provides an AUCτ ( (μg»hr/mL) of metabolite M6 between about 12 and about 20. In some embodiments, the Cmax (μg/mL) of metabolite M6 is between about 0.25 and about 4. In some embodiments, the Cmax (μg/mL) of metabolite M6 is between about 1 and about 3. In some embodiments, the Cmax (μg/mL) of metabolite M6 is between about 2.4 and about 3. In some embodiments, the molar ratio of the AUCτ (μg»hr/mL) of the metabolite M6 to the AUCτ (μg»hr/mL) of Compound I is about 2 to about 11. In some embodiments, the molar ratio of the AUCτ (μg»hr/mL) of the metabolite M6 to the AUCτ (μg»hr/mL) of
Compound I is about 4 to about 8. In some embodiments, the molar ratio of the Cmax (μg/mL) of the metabolite M6 to the Cmax (μg/mL) of Compound I is about is about 1 to about 7. In some embodiments, the molar ratio of the Cmax (μg/mL) of the metabolite M6 to the Cmax (μg/mL) of Compound I is about is about 2 to about 6. INCORPORATION BY REFERENCE
[0040] All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS [0041] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which: [0042] Figure 1 shows a flow chart depicting the steps for the preparation of the composition described in example 2 A.
[0043] Figure 2 shows a flow chart depicting the steps for the preparation of the composition described in example 3A.
[0044] Figure 3 shows a flow chart depicting the steps for the preparation of the composition described in example 4A. [0045] Figure 4 shows a flow chart depicting the steps for the preparation of the composition described in example 5A.
[0046] Figure 5 shows Serum Uric Acid Levels (measured on Day 9), after administration of compositions comprising compound 1. DETAILED DESCRIPTION OF THE INVENTION
[0047] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. Pharmaceutical compositions
[0048] Described herein are pharmaceutical compositions comprising 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof. In some embodiments, the pharmaceutical compositions comprise an effective amount of 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof. In some embodiments, the pharmaceutical compositions comprise an effective amount of 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof and at least one pharmaceutically acceptable carrier. In some embodiments the pharmaceutical compositions are for the treatment of disorders. In some embodiments the pharmaceutical compositions are for the treatment of disorders in a mammal. In some embodiments the pharmaceutical compositions are for the treatment of disorders in a human. Methods: Aberrant uric Acid Levels [0049] The present invention also provides methods useful for diseases or disorders related to aberrant uric acid levels. The method includes administering an effective amount of a composition as described herein to a subject with aberrant levels of uric acid such as to restore acceptable or non-aberrant levels of uric acid. The present invention also provides methods useful for decreasing uric acid levels in one or more tissues or organs of a subject in need of decreased uric acid levels, comprising administering to the subject a uric acid level decreasing amount of a composition as described herein. The present invention also provides methods useful for reducing uric acid production, increasing uric acid excretion or both in a subject, comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for treating or preventing hyperuricemia in a subject comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for treating a subject suffering from a condition characterized by abnormal tissue or organ levels of uric acid comprising administering to the subject an effective amount of a composition as described herein The present invention also provides methods useful for treating a subject suffering from gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosis, comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for preventing a condition characterized by abnormal tissue levels of uric acid in a subject at increased risk of developing the condition, comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for treating gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley- Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosisin a subject comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for treating gout in a subject comprising administering to the subject an effective amount of a composition as described herein. The present invention also provides methods useful for preventing the formation or reducing the size of tophi/tophus in a subject, comprising administering to the subject an effective amount of a composition as described herein. Terminology [0050] The term "subject", "patient" or "individual" as used herein in reference to individuals suffering from a disorder, and the like, encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non- human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non- mammals include, but are not limited to, birds, fish and the like. In one embodiment of the methods and compositions provided herein, the mammal is a human.
[0051] The terms "effective amount", "therapeutically effective amount" or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound being administered that is sufficient to treat or prevent the particular disease or condition. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in a disease. An appropriate "effective" amount in any individual case may be determined using techniques, such as a dose escalation study.
[0052] The term "pharmaceutically acceptable" as used herein, refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compounds described herein, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[0053] The terms "composition" and "pharmaceutical composition," as used herein, refer to 4-(2- (5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof, optionally mixed with at least one pharmaceutically acceptable chemical component, such as, though not limited to carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients and the like.
[0054] The term "pharmaceutically acceptable salt" as used herein, includes salts of 4-(2-(5- bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoate with any pharmaceutically acceptable cation, that retain the biological effectiveness of the free acid. For example, the free acid group of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H- l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid may react with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Illustrative examples of bases include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N+(C 1-4 alkyl)4, and the like. Representative organic amines useful for the formation of base addition salts include arginine, lysine, ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. Salts can be prepared in situ during final isolation and purification, or by separate reaction and isolation of the salt thus formed. [0055] The term "prodrug" as used herein, refers to a drug precursor that, following administration to a subject and subsequent absorption, is converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Thus, the term encompasses any derivative of a compound, which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or a pharmaceutically active metabolite or residue thereof. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g. by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g. the brain or lymphatic system). Doses
[0056] The total amount of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3- ylthio)acetamido)-3-chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof, administered will firstly be dependent on the mammal being treated. In the instances where administration is to a human subject, the amount will normally be determined by the prescribing physician with the dosage generally varying according to the age, sex, diet, weight, general health and response of the individual patient, the severity of the patient's symptoms, the precise indication or condition being treated, the severity of the indication or condition being treated, time of administration, route of administration, the disposition of the composition, rate of excretion, drug combination, and the discretion of the prescribing physician. The pharmaceutical composition may be in unit dosage form. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose. Determination of the proper dosage for a particular situation is within the skill of the art. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. The amount and frequency of administration, and if applicable other therapeutic agents and/or therapies, will be regulated according to the judgment of the attending clinician (physician) considering such factors as described above. Thus the amount of pharmaceutical composition to be administered may vary widely. Administration may occur in an amount of between about 0.001 mg/kg of body weight to about 100 mg/kg of body weight per day (administered in single or divided doses), or at least about 0.1 mg/kg of body weight per day. A particular therapeutic dosage includes, e.g., from about 0.01 mg to about 7000 mg of compound, or, e.g., from about 0.05 mg to about 2500 mg. The quantity of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or pharmaceutically acceptable salts thereof in a unit dose may occur in an amount of between about lmg to 3000 mg, from about 2 mg to 2000 mg, or 10 mg to 2000 mg, according to the particular application. In some embodiments, the amount is from about lOOmg to about 1500mg, from about 150mg to about 1200mg, or from about 200mg to about lOOOmg. In further or additional embodiments, the amount is at least lOOmg, at least 200mg, at least 250mg, at least 300mg, at least 400mg, at least 500mg, at least 600mg, at least 700mg, at least 750mg, at least 800mg, at least 900mg or at least lOOOmg. In further or additional embodiments, the amount is about lOOmg, about 200mg, about 250mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 750mg, about 800mg, about 900mg or about lOOOmg. In some embodiments the compositions are administered once daily. In further or additional embodiments, the compositions are administered twice daily. In further or additional embodiments, the compositions are administered at least twice daily. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day. In combinational applications in which the compound is not the sole therapy, it may be possible to administer lesser amounts of compound and still have therapeutic or prophylactic effect. Dosage Forms
[0057] The pharmaceutical compositions described herein are typically useful for oral administration as solid dosage forms, such as tablets, capsules, pills, powders, granules and the like. They may be administered for immediate release, delayed release or sustained release of 4- (2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid, prodrugs, and/or a pharmaceutically acceptable salts thereof. Kits
[0058] The compositions and methods described herein provide kits for the treatment of diseases and disorders. These kits comprise a compositions described herein in a container and, optionally, instructions teaching the use of the kit. Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
[0059] The compositions described herein are also useful for diagnostics and as research reagents. For example, the compounds described herein, either alone or in combination with other compounds, may be useful as tools in differential and/or combinatorial analyses to elucidate expression patterns of genes expressed within cells and tissues. As one non-limiting example, expression patterns within cells or tissues treated with one or more compounds are compared to control cells or tissues not treated with compounds and the patterns produced are analyzed for differential levels of gene expression as they pertain, for example, to disease association, signaling pathway, cellular localization, expression level, size, structure or function of the genes examined. These analyses can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds which affect expression patterns. [0060] Besides being useful for human treatment, the compositions described herein may also be useful for veterinary treatment of other animals.
EXAMPLES
[0061] The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. / Chemical Syntheses
Example 1 : Synthesis of 4-(2-(5-bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3- ylthio)acetamido)-3-chlorobenzoic acid (compound 1)
Figure imgf000019_0001
[0062] 4-(2-(5-Bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid (compound 1) was prepared as described previously (see US published patent application US 2006-0270725) and outlined below.
Figure imgf000020_0001
-C yclopropylnaphthalen- 1 -amine [0063] Palladium acetate (23mg, 0.1 mmol) was added to a solution of 4-bromonaphthalen-l- amine (500mg, 2.01 mmol), cyclopropyl boronic acid (225mg, 2.62 mmol), potassium phosphate (1.49g, 7.04 mmol) and tricyclohexylphosphine (56mg, 0.2 mmol) in toluene (10 mL) and water (0.4 mL) under nitrogen atmosphere. The mixture was heated at 1000C for 3h and then cooled to room temperature. Water was added and the mixture extracted with ethyl acetate, dried over sodium sulfate and concentrated to give crude 4-cyclopropylnaphthalen-l -amine (550mg) which was used in the next step without further purification.
Figure imgf000020_0002
l-Cyclopropyl-4-isothiocyanatonaphthalene [0064] Sodium bicarbonate (7mL, sat. solution) and thiophosgene (0.2mL, 2.6mmol) were added to a solution of 4-cyclopropylnaphthalen-l -amine (440mg, 2.6mmol) in dichloromethane (14mL) and the mixture stirred at room temperature for Ih. The organic layer was separated, dried over sodium sulfate and concentrated to give crude l-cyclopropyl-4-isothiocyanatonaphthalene (877mg, 99%) which was used in the next step without further purification.
5-Amino-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazole-3-thiol
Figure imgf000020_0003
[0065] Aminoguanidine hydrochloride (355mg, 3.2mmol) and diisopropyl ethylamine (0.56mL, 3.2mmol) were added to a solution of l-cyclopropyl-4-isothiocyanatonaphthalene (447mg, 2.1mmol) in dimethylformamide (DMF, 3mL) and the mixture stirred at 500C for 18 hours. The mixture was then concentrated and aqueous sodium hydroxide solution (2M, 10 mL) added. The mixture was stirred at 500C for 18 hours, cooled to room temperature and neutralized with aqueous IN HCl. The resulting precipitate was isolated to give 5-amino-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazole-3 -thiol (240mg, 44%). 4-(2-(5-Amino-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid
Figure imgf000020_0004
[0066] 5-Amino-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazole-3-thiol (789mg, 2.79mmol) and 3-chloro-4-(2-chloroacetamido)benzoic acid (693mg, 2.79mmol) were dissolved in DMF (6mL) and the solution stirred at 500C for 18 hours. Water was then added and the mixture extracted with ethyl acetate. The organic layer was separated, dried over sodium sulfate and concentrated to give 4-(2-(5-amino-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3- ylthio)acetamido)-3-chlorobenzoic acid (1.04g, 75%).
4-(2-(5-Bromo-4-(l-cvclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid
Figure imgf000021_0001
[0067] Dichloroacetic acid (0.35 rnL, 4.2 mmol) was added to a mixture of give 4-(2-(5-amino- 4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid (1.04g, 2.1mmol), benzyltriethyl ammonium bromide (1.65g, β.lmmol) and sodium nitrite (2.9g, 42.1mmol) in dibromomethane (44mL). The mixture was stirred at room temperature for 18 hours in the dark, then concentrated and the resulting residue purified by column chromatography (95% dichloromethane /5% methanol) to give 4-(2-(5-bromo-4-(l- cyclopropylnaphthalen-4-yl)-4H- 1 ,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid (393 mg, 34%). II Preparation and Analysis of Compositions Example 2A: Preparation of Immediate Release Capsules, 100 mg
[0068] 4-(2-(5-Bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid (compound 1 , prepared as described herein) and microcrystalline cellulose were screened through a #40 mesh screen. The screened mixture and croscarmellose sodium were blended together using a Turbular Type TlOB Shaker Mixer for approximately 15 minutes. 200mg of the powder blend was encapsulated into a size # 2 dark green opaque hard gelatin Coni-Snap capsule.
[0069] Two batches of capsules were prepared according to this procedure; the first batch size was 3380 capsules and the second 4400 capsules. The capsules were analyzed for identity, strength, purity, content uniformity and dissolution profiles, as described below. Unit Composition
Figure imgf000021_0002
Batch Composition
Figure imgf000022_0001
* As free acid, equivalent to 106.8 mg of potassium salt. The quantity of compound 1 used was adjusted based on the actual potency and a corresponding adjustment with microcrystalline cellulose made.
Example 2B: Identification of compound 1 in capsules prepared in example 2A [0070] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the identity of compound 1. The capsules were extracted with 50/50 (v/v) acetonitrile/water. The identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 0.97 - 1.03 relative to reference standard, confirming the presence of compound 1. Example 2C: Determination of quantity of compound 1 in capsules prepared in example 2A [0071] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column,; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present. The capsules were extracted with 50/50 (v/v) acetonitrile/water. The quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly. Example 2D: Determination of impurities in capsules prepared in example 2A
[0072] A gradient elution reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase A lO mM KH2PO4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content. The capsules were extracted with 50/50 (v/v) acetonitrile/water. Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly. Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
Figure imgf000023_0001
NMT - not more than; RRT = relative retention time
Example 2E: Determination of water content in capsules prepared in example 2 A
[0073] Water content was determined using the USP <921>, Karl Fischer method. The results are shown below:
Figure imgf000023_0002
Example 2F: Dissolution Profile of compound 1 from capsules prepared in example 2A [0074] Dissolution of compound 1 from the capsules was determined in 900 mL of water at 37°C using USP Apparatus 1 operating at 75 rpm. A filtered sample of the dissolution medium was taken at the specified time(s) and analyzed by an HPLC procedure using the same chromatographic conditions as for content determination, as described above. The release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
Figure imgf000023_0003
NLT = not less than; RRT = relative retention time
Example 3A: Preparation of Over-Encapsulated Capsules, 100 mg
[0075] Size 1 white Vcaps capsule shells were loaded into Profill encapsulator trays and the
Vcaps caps were separated from the capsule bodies. A size # 2 dark green opaque hard gelatin
Coni-Snap capsule (prepared as described in example 2A above) was placed into each of the
Vcaps capsule bodies. The capsule caps were placed back onto capsule bodies using the Profill equipment and gently pressed shut to secure the caps on the bodies. This process was repeated using additional trays as necessary. [0076] Two batches of capsules were prepared according to this procedure; the first batch size was 680 capsules and the second 2200 capsules. The capsules were analyzed for identity, strength, purity, content uniformity and dissolution profiles, as described below. Unit Composition
Figure imgf000024_0001
a As free acid, equivalent to 106.8 mg of potassium salt. The quantity of compound 1 used was adjusted based on the actual potency and a corresponding adjustment with microcrystalline cellulose made.
Example 3B: Identification of compound 1 in capsules prepared in example 3 A [0077] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the of identity of compound 1. The capsules were extracted with 50/50 (v/v) acetonitrile/water. The identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 0.97 - 1.03 relative to reference standard, confirming the presence of compound 1.
Example 3C: Determination of quantity of compound 1 in capsules prepared in example 3 A [0078] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column,; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present. The capsules were extracted with 50/50 (v/v) acetonitrile/water. The quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly and is shown below.
Figure imgf000024_0002
Example 3D: Determination of impurities in capsules prepared in example 3 A [0079] A gradient elution reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase A lO mM KH2PO4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content. The capsules were extracted with 50/50 (v/v) acetonitrile/water. Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly. Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
Figure imgf000025_0001
NMT - not more than; RRT = relative retention time
Example 3E: Determination of water content in capsules prepared in example 3 A
[0080] Water content was determined using the USP <921>, Karl Fischer method. The results are shown below:
Figure imgf000025_0002
Example 3F: Dissolution Profile of compound 1 from capsules prepared in example 3 A [0081] Dissolution of compound 1 from the capsules was determined in 900 mL of water at 37°C using USP Apparatus 1 operating at 75 rpm. A filtered sample of the dissolution medium was taken at the specified times (15, 30, 45 and 60 minutes) and analyzed by HPLC using the same chromatographic conditions as described above for content determination. Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below. Batch number 1 2
Dissolution profile Ave. RSD Min. Max. Ave. RSD Min. Max.
15min = 7% 122% 0% 15min = 8% 163% 0%
20% 34%
30min = 92% 11% 77% 30min = 93% 4% 87%
102% 97%
45min = 101% 2% 98% 45min = 107% 3% 101%
103% 109%
60min = 101% 2% 98% 60min = 107% 3% 102%
103% 109%
NLT = not less than; RRT = relative retention time
Example 4A: Preparation of Enteric Coated Granules, 200 mg
[0082] 4-(2-(5-Bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid (compound 1 , prepared as described herein) was milled and then blended in a planetary mixer with microcrystalline cellulose and hypromellose. The blended ingredients in the planetary mixer were granulated with purified water and the wet granulation was sieved through a No. 8 mesh screen and dried on trays in an oven at 4O0C to a moisture content of less than 5% as determined by loss on drying (LOD). The dried granulation was sieved through a No. 40 mesh screen; the granules retained on the 40 mesh screen were collected, and the remaining fines transferred to recovered waste. Using a fluidized bed coating unit, a hypromellose solution (7% w/w in purified water) was applied to the > 40 mesh granules, followed by application of a talc, triethyl citrate and methacrylic acid dispersion (previously sieved through an 80 mesh screen).
Approximately 317 mg of the coated granules was manually filled into each of 1700 hard gelatin capsules, Swedish Orange Opaque, Size 1.
[0083] One batch of 1700 capsules was prepared according to this procedure. The capsules were analyzed for identity, strength, purity, content uniformity and dissolution profiles, as described below.
Unit Composition
Figure imgf000026_0001
Batch Composition
Figure imgf000027_0001
a As free acid, equivalent to 213.6 mg of potassium salt. The quantity of compound 1 used was adjusted based on the actual potency and a corresponding adjustment with microcrystalline cellulose made. b Quantity sufficient to facilitate granule formation, removed during the drying process c Eudragit L30D-55 is a dispersion containing 30% solids
Example 4B: Identification of compound 1 in capsules prepared in example 4A
[0084] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase of 45% 1OmM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the identity of compound 1. The capsules were extracted with 20:80 (v/v) methanol/phosphate buffer (pH 7.4) and diluted 1 :10 with methanol/water (20:80 v/v ratio). The target concentration of compound 1 for assay is 0.1 mg/mL. The identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 1.00 ± 0.03 relative to reference standard, confirming the presence of compound 1.
[0085] Example 4C: Determination of quantity of compound 1 in capsules prepared in example
4A
[0086] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column,; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present. The capsules were extracted with extracted with 20:80 (v/v) methanol/phosphate buffer (pH 7.4) and diluted 1 :10 with methanol/water (20:80 v/v ratio). The target concentration of compound 1 for assay is 0.1 mg/mL. The quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly, and confirmed the quantity to be 98.2%, within the 90.0 - 110.0 % limit set.
Example 4D: Determination of impurities in capsules prepared in example 4A [0087] A gradient elution reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase A lO mM KH2PO4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content. The capsules were extracted with 20/80 (v/v) methanol/phosphate buffer (pH 7.4). Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly. Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
Figure imgf000028_0001
Example 4E: Determination of water content in capsules prepared in example 4 A [0088] Water content was determined using the USP <921>, Karl Fischer method and measured at 6.9%.
Example 4F: Dissolution Profile of compound 1 from capsules prepared in example 4A [0089] Dissolution of compound 1 from the capsules was determined following the principles of the USP <711> method A for delayed-release dosage forms, using USP Apparatus 2 at 37°C, set at 50 rpm. Acid stage was performed for 2 hours in 700 mL of dissolution medium at pH 1.2 followed by buffer stage performed in 900 mL of dissolution medium at pH 6.8. A filtered aliquot of the test dissolution medium was taken at the specified times (15, 30, 45 and 60 minutes) and analyzed by HPLC using the same chromatographic conditions as described above for content determination. Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
Figure imgf000029_0001
NMT = not more than; RRT = relative retention time
Example 5A: Preparation of Enteric Coated Tablets, 200 mg
[0090] 4-(2-(5-Bromo-4-(l-cyclopropylnaphthalen-4-yl)-4H-l,2,4-triazol-3-ylthio)acetamido)-3- chlorobenzoic acid (compound 1, prepared as described herein) was milled through a Fitzmill fitted with a 0.033" round-hole screen. The milled compound 1, microcrystalline cellulose and hypromellose were blended with a Robot Coupe RSI-3 VG high-shear roto-granulator for approximately 5 minutes and the blended ingredients granulated with purified water. The wet granulation was sieved through a No. 10 mesh screen and dried on trays in an oven at 4O0C to a moisture content of NMT 3% as determined by loss on drying (LOD). The dried granulation was sieved through a No. 10 mesh screen and approximately half of the granulation was charged into a Bohle MC-5 (5.0 liter) bin. Magnesium stearate, NF was sieved through a No. 30 mesh screen into the MC-5 bin and the remaining granulation was charged to the bin. The mixture was blended with a Bohle LM 40 Bin blender at 25 RPM for 3 minutes and the blended granulation compressed to a target tablet weight of 262 mg using a rotary tablet press. A 7% w/w coating solution of hypromellose 2910, in purified water was prepared using a lab mixer and applied to the compressed tablets using a Vector LCDS-3 coating system to achieve an approximate 3% weight gain (~8 mg/tablet). An 18% w/w enteric coating suspension of Acryl-Eze White (methacrylic acid co-polymer) in purified water was prepared using a lab mixer, and the suspension applied to the previously coated tablets using the Vector LCDS-3 coating system to achieve an approximate 10% weight gain (~27 mg/tablet). The isolated tablets were white, caplet-shaped tablets approximately 0.2" x 0.43" in dimension, weighing approximately 298 mg each and containing 200mg of compound 1 (present as the potassium salt). [0091] One batch of 1700 capsules was prepared according to this procedure. The capsules were analyzed for identity, strength, purity, content uniformity and dissolution profiles, as described below. Unit Composition
Figure imgf000030_0001
Batch Composition
Figure imgf000030_0002
a As free acid, equivalent to 213.6 mg of potassium salt. The quantity of compound 1 used was adjusted based on the actual potency and a corresponding adjustment with microcrystalline cellulose made. b Quantity sufficient to facilitate granule formation, removed during the drying process Example 5B: Identification of compound 1 in capsules prepared in example 5 A
[0092] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase of 45% 1OmM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to confirm the identity of compound 1. The capsules were extracted with 20:80 (v/v) methanol/phosphate buffer (pH 7.4) and diluted 1 :10 with methanol/water (20:80 v/v ratio). The target concentration of compound 1 for assay is 0.1 mg/mL. The identity of compound 1 was determined by comparing the retention time of the peak in the sample preparation with that of a standard preparation and showed relative retention values (Rr) 1.00 ± 0.03 relative to reference standard, confirming the presence of compound 1. Example 5C: Determination of quantity of compound 1 in capsules prepared in example 5 A [0093] An isocratic reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column,; mobile phase of 45% 10 mM KH2PO4 (pH 3.0) and 55% acetonitrile) with UV detection at 220 nm, was used to determine the amount of compound 1 present. The capsules were extracted with extracted with 20:80 (v/v) methanol/phosphate buffer (pH 7.4) and diluted 1 :10 with methanol/water (20:80 v/v ratio). The target concentration of compound 1 for assay is 0.1 mg/mL. The quantity of compound 1 present was determined by comparing sample peaks with standard preparations obtained concomitantly, and confirmed the quantity to be 102.8%, within the 90.0 - 110.0 % limit set.
Example 5D: Determination of impurities in capsules prepared in example 5 A [0094] A gradient elution reversed-phase HPLC method, (4.6 x 150 mm YMC ODS AQ, 3 μm size particles column; mobile phase A lO mM KH2PO4 (pH 3.0) and mobile phase B acetonitrile) with UV detection at 220 nm, was used to determine impurity content. The capsules were extracted with 20/80 (v/v) methanol/phosphate buffer (pH 7.4). Target concentration of compound 1 for impurities assay was 1 mg/mL. Impurities were determined by comparing the impurity peak areas in the sample preparation chromatogram to the area of the compound 1 peak in the standard preparation obtained concomitantly. Known impurities were calculated by applying the response factor of the respective impurities. The method reporting limit was 0.05%. The results of the analyses are shown in the table below.
Figure imgf000031_0001
Figure imgf000032_0001
Example 5E: Determination of water content in capsules prepared in example 5 A
[0095] Water content was determined using the USP <921>, Karl Fischer method and measured at 4.75%.
Example 5F: Dissolution Profile of compound 1 from capsules prepared in example 5 A
[0096] Dissolution of compound 1 from the capsules was determined following the principles of the USP <711> method A for delayed-release dosage forms, using USP Apparatus 2 at 37°C, set at 50 rpm. Acid stage was performed for 2 hours in 700 mL of dissolution medium at pH 1.2 followed by buffer stage performed in 900 mL of dissolution medium at pH 6.8. A filtered aliquot of the test dissolution medium was taken at the specified times (30, 45, 60, 90 and 100 minutes) and analyzed by HPLC using isocratic elution and UV detection at 226 nm (same chromatographic conditions as described above for content determination). Compound release was determined by comparing the peak responses of the sample chromatograms to the peak responses of the standard chromatograms obtained concomitantly and the results are shown below.
Figure imgf000032_0002
Mean = 91.2%
% RSD = 4.1%
60 min
Min = 84.3%
Max = 95.2%
Mean = 96.8%
% RSD = 2.0%
90 min
Min = 94.4%
Max = 99.4%
Mean = 101.3%
% RSD = 2.8%
100 min
Min = 97.0%
Max = 105.1%
NMT = not more than III Human Clinical Studies Example 6
[0097] Human clinical studies using the compositions described in examples 2A, 3A, 4A, 5A were conducted as described: A multi-center, double-blind, placebo-controlled study in 48 treatment-naϊve HIV-I -infected subjects, randomized 3: 1 (treatmentplacebo). Patients were male, 18-65 years with chronic HIV infection, but no history of AIDS-defining illness. Patients were antiretro viral treatment naϊve or < 14 days prior therapy and had no pre-existing RTI or PI drug resistance and no co-infection with acute HAV, chronic HBV, active HCV. Patient CD4+ cell count was > 50 cells/mm3 for 2 cohorts, then > 200 cells/mm3 or > 350 cells/mm3 depending on site.
[0098] The compositions were administered over a 7-day treatment period plus one morning dose for PK purposes on day 8. (It should be noted that in some instances multiple doses of the same composition were administered, in order to achieve the required total dose of compound 1; i.e. in order to achieve a dose of 400mg compound 1, a patient may have taken four lOOmg compositions or two 200mg compositions, depending on the dosage form.) Four sequential dose cohorts were administered as follows:
Capsules EC Tablets
400mg BID Fasted 800mg QD Fed
600mg QD Fasted lOOOmg QD Fasted Assessments were made as follows:
• PK and tolerability Days 1-10 & 2 wks post-dose • Safety labs, immunology Days 1, 4, 9 & 2 wks post-dose
• ECGs Days 1, 3, 4, 7, 9 and 2 wks post-dose
• Genotype and phenotype Days 1 , 9 & 2 wks post-dose
Safety / Tolerability Observations: No serious adverse events (grade 3/4) or premature discontinuations occurred during the trail. There was no indication of CNS toxicity, no drug related rashes, no clinically significant laboratory abnormalities, no apparent effects on lipid profile, no clinically relevant ECG findings and no characteristic changes in genotypes or phenotypic susceptibility observed. Adverse events were generally mild (grade 1) with no required intervention. Patient Baseline Characteristics were determined as shown below:
Compound 1 PLACEBO
400mg BID* 600mg QD* 800mg QD lOOOmg QD*
N=9 N=9 N=9 N=9 N=12
Age
Mean years 35.3 39.9 31.2 33.0 36.3
Race
Caucasian 7 9 7 7 8
Black 2 - 1 2 1
Asian - - - - -
Other - - 1 - -
CD4 Cell
Count
Mean
288.1 319.9 303.6 407.2 325.9 cells/mL
Viral Load
Copies/ml 31,815 46,845 40,161 39,852 32,551
4880- 6060- 15900- 5730-
Range 7520-469000
113000 879000 244000 233000
The Steady-State Pharmacokinetics of the various dosage forms were determined and are shown below:
Figure imgf000034_0001
Serum uric acid levels were recorded on day 9 of the study, and are shown in the table below and graphically in figure 5.
PLACEBO 600mg QD 800mg QD 100<jmg 400mg BID
Mean serum UA
-6 -21 -21 -28 -30 reduction (%) T-test vs placebo 0.0011 0.0010 <0.0001 <0.0001
Example 7 [0099] Additional human clinical studies were undertaken to further investigate the PK properties of the compositions described herein. The results are summarized in the table below.
Figure imgf000035_0001
1 State: Fa = Fasted; EF = Evening Fasted; FeS = Fed Standard Breakfast; FeH = Fed FDA High
Fat Breakfast
2M6 is 2-(5-bromo-4-(4-cyclopropylnaphthalen-l-yl)-4H-l,2,4-triazol-3-ylthio)acetic acid - the predominant metabolite of Compound 1
3Molar Ratio M6/Compound 1

Claims

WHAT IS CLAIMED IS:
1. A pharmaceutical composition comprising: a compound or mixture of compounds of structure (I):
Figure imgf000036_0001
(I), wherein M is hydrogen, sodium, potassium, calcium, or arginine; and one or more diluents; one or more binders; one or more coating agents; one or more dispersing agents; and one or more plasticizers.
2. The pharmaceutical composition of claim 1, wherein the diluent is microcrystalline cellulose, silicifϊed microcrystalline cellulose, cellulose, lactose, compressible sugar, mannitol, calcium silicate and calcium phosphate in powder and granular forms, sodium phosphate or sodium carbonate.
3. The pharmaceutical composition of claim 1 , wherein the binder is hypromellose, povidone, hydroxypropyl cellulose, hydroxyethyl cellulose or starch.
4. The pharmaceutical composition of claim 1 , wherein the coating agent is a methacrylic acid based copolymers, Eudragit L30D55, Acryl-Eze, hydroxypropylmethyl cellulose acetate succinate, polyvinylacetate phthalate or cellulose acetate phthalate.
5. The pharmaceutical composition of claim 1, wherein the dispersing agent is talc, glycerol monostearate or colloidal silicon dioxide.
6. The pharmaceutical composition of claim 1, wherein the plasticizer is triethyl citrate, triacetin, dibutyl phthalate, diethyl phthalate or glycerin.
7. The pharmaceutical composition of claim 1, comprising from about lOmg to about lOOOmg of the compound or mixture of compounds of structure (I).
8. The pharmaceutical composition of claim 1, comprising about lOOmg, or about 200mg, or about 300mg, or about 400mg, or about 500mg, or about 600mg, or about 700mg, or about 800mg, or about 900mg, or about lOOOmg of the compound or mixture of compounds of structure (I).
9. A pharmaceutical composition comprising: a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000036_0002
(IB); microcrystalline cellulose; hypromellose; methacrylic acid copolymer dispersion; talc; and triethyl citrate.
10. The composition of claim 9 comprising: about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form; about 35mg of microcrystalline cellulose; about 29mg of hypromellose; about 30mg of methacrylic acid copolymer dispersion; about 6mg of talc; and about 3mg of triethyl citrate.
11. A pharmaceutical composition comprising: from about 60% to about 90% by weight of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000037_0001
(IB); from about 5% to about 15% by weight of microcrystalline cellulose; from about 5% to about 15% by weight of hypromellose; from about 5% to about 15% by weight of methacrylic acid copolymer dispersion; from about 0.5% to about 5% by weight of talc; and from about 0.1% to about 3% by weight of triethyl citrate.
12. A pharmaceutical composition comprising: a compound of structure (IB) or a mixture of
a compound of structure (IB) and its free acid form:
Figure imgf000037_0002
(IB); microcrystalline cellulose; and hypromellose; wherein the composition is in the form of granules.
13. A pharmaceutical composition comprising : about 214mg of a compound of structure (IB) or a mixture of a compound of structure (IB) and its free acid form:
Figure imgf000037_0003
(IB); about 35mg of microcrystalline cellulose; about 13.5 mg of hypromellose; wherein the composition is in the form of granules which do not pass through a 40 mesh screen; and wherein the granules are coated with about 15.3 mg hypromellose; and wherein the coated granules are further coated with a composition comprising: about 30.4mg of methacrylic acid copolymer dispersion; about 6.1mg of talc; and about 3.0mg of triethyl citrate.
14. The composition of any preceding claim, wherein the in vitro dissolution rate as measured using United States Pharmacopoeia Apparatus 1 operating at 75 rpm in 900 mL water at 37°C is not less than about 80% of the compound of structure (IB) is released within 30 minutes.
15. A method for decreasing uric acid levels in one or more tissues or organs of a subject in need of decreased uric acid levels, comprising administering to the subject a uric acid level decreasing amount of the pharmacuetical composition according to any of the preceding claims.
16. A method for reducing uric acid production, increasing uric acid excretion or both in a subject, comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
17. A method for treating or preventing hyperuricemia in a subject comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
18. A method of treating a subject suffering from a condition characterized by abnormal tissue or organ levels of uric acid comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
19. A method for preventing a condition characterized by abnormal tissue levels of uric acid in a subject at increased risk of developing the condition, comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
20. A method for treating gout, a recurrent gout attack, gouty arthritis, hyperuricaemia, hypertension, a cardiovascular disease, coronary heart disease, Lesch-Nyhan syndrome, Kelley-Seegmiller syndrome, kidney disease, kidney stones, kidney failure, joint inflammation, arthritis, urolithiasis, plumbism, hyperparathyroidism, psoriasis or sarcoidosisin a subject comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
21. A method for treating gout in a subject comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
22. A method for preventing the formation or reducing the size of tophi/tophus in a subject, comprising administering to the subject an effective amount of the pharmaceutical composition according to any of the preceding claims.
PCT/US2009/061969 2008-10-24 2009-10-23 Compositions comprising 4- (2- ( 5-br0m0-4- ( 1-cyclopropylnaphthalen-4-yl) -4h-1, 2, 4-triaz0l-3-ylthi0) acetamido -3-chlorobenzoic acid and pharmaceutically acceptable salts thereof WO2010048592A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10841208P 2008-10-24 2008-10-24
US61/108,412 2008-10-24

Publications (1)

Publication Number Publication Date
WO2010048592A1 true WO2010048592A1 (en) 2010-04-29

Family

ID=41494050

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/061969 WO2010048592A1 (en) 2008-10-24 2009-10-23 Compositions comprising 4- (2- ( 5-br0m0-4- ( 1-cyclopropylnaphthalen-4-yl) -4h-1, 2, 4-triaz0l-3-ylthi0) acetamido -3-chlorobenzoic acid and pharmaceutically acceptable salts thereof

Country Status (2)

Country Link
TW (1) TW201020240A (en)
WO (1) WO2010048592A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7947721B2 (en) 2004-08-25 2011-05-24 Ardes Biosciences Inc. S-triazolyl α-mercaptoacetanilides as inhibitors of HIV reverse transcriptase
WO2011159840A2 (en) 2010-06-16 2011-12-22 Ardea Biosciences, Inc. Phenylthioacetate compounds, compositions and methods of use
US8084483B2 (en) 2007-11-27 2011-12-27 Ardea Biosciences, Inc. Compounds and compositions and methods of use
US8106205B2 (en) 2004-08-25 2012-01-31 Ardea Biosciences, Inc. N[S(4-aryl-triazol-3-yl)α-mercaptoacetyl]p-amino benzoic acids as HIV reverse transcriptase inhibitors
US10266493B2 (en) 2010-06-16 2019-04-23 Ardea Biosciences, Inc. Thioacetate compounds, compositions and methods of use

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006026356A2 (en) * 2004-08-25 2006-03-09 Ardea Biosciences, Inc. S-TRIAZOLYL α-MERCAPTOACETANILDES AS INHIBITORS OF HIV REVERSE TRANSCRIPTASE
WO2007050087A1 (en) * 2004-08-25 2007-05-03 Ardea Biosciences, Inc. N[S(4-aryl-triazol-3-yl)α -mercaptoacetyl]-p-amino benozoic acids AS HIV REVERSE TRANSCRIPTASE INHIBITORS
WO2009070740A2 (en) * 2007-11-27 2009-06-04 Ardea Biosciences Inc. Novel compounds and compositions and methods of use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006026356A2 (en) * 2004-08-25 2006-03-09 Ardea Biosciences, Inc. S-TRIAZOLYL α-MERCAPTOACETANILDES AS INHIBITORS OF HIV REVERSE TRANSCRIPTASE
WO2007050087A1 (en) * 2004-08-25 2007-05-03 Ardea Biosciences, Inc. N[S(4-aryl-triazol-3-yl)α -mercaptoacetyl]-p-amino benozoic acids AS HIV REVERSE TRANSCRIPTASE INHIBITORS
WO2009070740A2 (en) * 2007-11-27 2009-06-04 Ardea Biosciences Inc. Novel compounds and compositions and methods of use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SWEENEY ZACHARY K ET AL: "Improving non-nucleoside reverse transcriptase inhibitors for first-line treatment of HIV infection: the development pipeline and recent clinical data.", CURRENT OPINION IN DRUG DISCOVERY & DEVELOPMENT JUL 2008, vol. 11, no. 4, July 2008 (2008-07-01), pages 458 - 470, XP009127983, ISSN: 1367-6733 *
YEH LITAIN ET AL: "RDEA594, a potential uric acid lowering agent through inhibition of uric acid reuptake, shows better pharmacokinetics than its prodru RDEA806", ARTHRITIS & RHEUMATISM, vol. 58, no. 9, Suppl. S, September 2008 (2008-09-01), & 72ND ANNUAL SCIENTIFIC MEETING OF THE AMERICAN-COLLEGE-OF-RHEUMATOLOG Y/43RD ANNUAL SCIENTIFIC MEETIN; SAN FRANCISCO, CA, USA; OCTOBER 24 29, 2008, pages S178, XP009127986, ISSN: 0004-3591 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8481581B2 (en) 2004-08-25 2013-07-09 Ardea Biosciences, Inc. S-triazolyl α-mercaptoacetanilides as inhibitors of HIV reverse transcriptase
US8106205B2 (en) 2004-08-25 2012-01-31 Ardea Biosciences, Inc. N[S(4-aryl-triazol-3-yl)α-mercaptoacetyl]p-amino benzoic acids as HIV reverse transcriptase inhibitors
US7947721B2 (en) 2004-08-25 2011-05-24 Ardes Biosciences Inc. S-triazolyl α-mercaptoacetanilides as inhibitors of HIV reverse transcriptase
US8552043B2 (en) 2004-08-25 2013-10-08 Ardea Biosciences, Inc. N[S(4-aryl-triazol-3-yl)α-mercaptoacetyl]-p-amino benzoic acids as HIV reverse transcriptase inhibitors
US8252828B2 (en) 2004-08-25 2012-08-28 Ardea Biosciences, Inc. S-triazolyl α-mercapto acetanilides as inhibitors of HIV reverse transcriptase
US8946273B2 (en) 2004-08-25 2015-02-03 Ardea Biosciences, Inc. S-Triazolyl alpha-mercapto acetanilides as inhibitors of HIV reverse transcriptase
US8084483B2 (en) 2007-11-27 2011-12-27 Ardea Biosciences, Inc. Compounds and compositions and methods of use
US8357713B2 (en) 2007-11-27 2013-01-22 Ardea Biosciences Inc. Compounds and compositions and methods of use
US8283369B2 (en) 2007-11-27 2012-10-09 Ardea Biosciences. Inc. Compounds and compositions and methods of use
US10183012B2 (en) 2007-11-27 2019-01-22 Ardea Biosciences, Inc. Compounds and compositions and methods of use
US8546437B2 (en) 2007-11-27 2013-10-01 Ardea Biosciences, Inc. Compounds and compositions and methods of use
EP2582664A2 (en) * 2010-06-16 2013-04-24 Ardea Biosciences, Inc. Phenylthioacetate compounds, compositions and methods of use
US10919858B2 (en) 2010-06-16 2021-02-16 Ardea Biosciences, Inc Thioacetate compounds, compositions and methods of use
WO2011159840A2 (en) 2010-06-16 2011-12-22 Ardea Biosciences, Inc. Phenylthioacetate compounds, compositions and methods of use
EP2582664A4 (en) * 2010-06-16 2014-07-09 Ardea Biosciences Inc Phenylthioacetate compounds, compositions and methods of use
TWI499412B (en) * 2010-06-16 2015-09-11 Ardea Biosciences Inc Phenylthioacetate compositions and methods of use
US9212135B2 (en) 2010-06-16 2015-12-15 Ardea Biosciences, Inc. Phenylthioacetate compounds, compositions and methods of use
US10266493B2 (en) 2010-06-16 2019-04-23 Ardea Biosciences, Inc. Thioacetate compounds, compositions and methods of use

Also Published As

Publication number Publication date
TW201020240A (en) 2010-06-01

Similar Documents

Publication Publication Date Title
US20180221285A1 (en) Oral formulations of deferasirox
US20210113472A1 (en) Pharmaceutical composition comprising a potent inhibitor of urat1
JP2509044B2 (en) Preparation for oral administration containing gastric juice containing bile salt
JP2916978B2 (en) Controlled release initiation type formulation
KR101697800B1 (en) Controlled release pharmaceutical formulations of nitazoxanide
JP2509045B2 (en) Gastric juice resistant formulation for oral administration containing bile acids
US20130259906A1 (en) Pharmaceutical composition comprising one or more fumaric acid esters
KR20110116027A (en) Pharmaceutical formulation comprising one or more fumaric acid esters in an erosion matrix
WO2010048592A1 (en) Compositions comprising 4- (2- ( 5-br0m0-4- ( 1-cyclopropylnaphthalen-4-yl) -4h-1, 2, 4-triaz0l-3-ylthi0) acetamido -3-chlorobenzoic acid and pharmaceutically acceptable salts thereof
US20190248830A1 (en) Proliposomal testosterone undecanoate formulations
US20210386674A1 (en) Modified release tablet formulations containing phosphodiesterase inhibitor
AU2007259408B2 (en) Oral DTPA for radionuclide chelation
US20220175766A1 (en) Compositions and methods of treatment
WO2010023690A2 (en) Prolonged release formulation of amisulpride
US20110244049A1 (en) Compositions comprising 4-(2-(5-bromo-4-(1-cyclopropylnaphthalen-4-yl)-4h-1,2,4-triazol-3-ylthio)acetamido)-3-chlorobenzoic acid and pharmaceutically acceptable salts thereof, and methods for preparing and using same
CN110662533A (en) Methods and compositions for delivering mycophenolic acid active agents to non-human mammals
KR102104507B1 (en) Pharmaceutical formulations comprising sodium palmitoyl-l-prolyl-l-prolyl-glycyl-l-tyrosinate and methods for preparing the same
JP2003519172A (en) Sustained-release anthelmintic composition containing praziquantel
US20180071220A1 (en) Oral formulations of deferasirox
KR20190007370A (en) Combination formulation having improved stability and dissolution rate comprising Bazedoxifene or its pharmaceutically acceptable salt, and Cholecalciferol or its pharmaceutically acceptable salt
WO2019004713A2 (en) Sustained-release pharmaceutical composition comprising ivabradine and method for producing same
JP7195354B2 (en) Pharmaceutical formulation containing one or more fumarates in an eroding matrix
KR101324425B1 (en) Bisphosphonate composition having enhanced oral bioavailability
TW202313072A (en) Pediatric formulations of ferric citrate
WO2023002004A1 (en) Multiparticulate pharmaceutical composition

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: 09749250

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09749250

Country of ref document: EP

Kind code of ref document: A1