Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR TREATMENT OF HYPERURICEMIA
Document Type and Number:
WIPO Patent Application WO/2012/122532
Kind Code:
A2
Abstract:
The present invention is directed to methods and compositions for reduction of uric acid levels in subjects, and methods and compositions that are useful for treating, suppressing and/or preventing afflictions related to elevated uric acid levels in subjects. The methods comprise administering to the subject an effective amount of compound I, or a pharmaceutically acceptable salt or hydrate thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt or hydrate thereof, and a pharmaceutically acceptable carrier. The compositions and/or methods may further comprise administering to the subject an effective amount of xanthine oxidase inhibitor such as allopurinol, or a pharmaceutically acceptable salt or hydrate thereof.

Inventors:
HOLLISTER ALAN (US)
SHERIDAN WILLIAM P (US)
BANTIA SHANTA (US)
FLYNT AMY (US)
ELDER JENNA (US)
WAUGH ANITA (US)
LYMAN SCOTT (US)
Application Number:
PCT/US2012/028616
Publication Date:
September 13, 2012
Filing Date:
March 09, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BIOCRYST PHARM INC (US)
HOLLISTER ALAN (US)
SHERIDAN WILLIAM P (US)
BANTIA SHANTA (US)
FLYNT AMY (US)
ELDER JENNA (US)
WAUGH ANITA (US)
LYMAN SCOTT (US)
International Classes:
B64C25/04
Foreign References:
US20090239885A12009-09-24
US20090197825A12009-08-06
US5494912A1996-02-27
US20100160351A12010-06-24
Other References:
FITZ-PATRICK ET AL.: 'Effects of a Purine Nucleoside Phosphorylase Inhibitor, BCX4208, on the Serum Uric Acid Concentrations in Patients with Gout.' APS- ACR POSTER SESSION A. 08 November 2010,
GOMES ET AL.: 'A Phase 2 Study of the Purine Nucleoside Phosphorylase (PNP) inhibitor R05092888 (BCX4208) in Patients with Moderate to Severe Chronic Plaque Psoriasis: Safety' TOLERABILITY AND LYMPHOCYTE EFFECTS - PRESENTED AT THE AMERICAN SOCIETY OF HEMATOLOGY 50TH ANNUAL MEETING & EXPOSITION (ASH) 06 December 2008, SAN FRANCISCO, CALIFORNIA,
LEWANDOWICZ ET AL.: 'Energetic Mapping of Transition State Analogue Interactions with Human and Plasmodium falciparum Purine Nucleoside Phosphorylases.' THE JOURNAL OF BIOLOGICAL CHEMISTRY vol. 280, no. 34, 26 August 2005, pages 30320 - 30328
Attorney, Agent or Firm:
LOVE, Jane, M. et al. (7 World Trade Center250 Greenwich Stree, New York NY, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for reducing serum uric acid levels in a subject comprising administering to said subject a therapeutically effective amount of compound I:

or pharmaceutically acceptable salt thereof.

2. The method of claim 1, further comprising administration of a xanthine oxidase

inhibitor, a urate oxidase, or a uricosuric agent.

3. The method of claim 2, wherein the xanthine oxidase inhibitor, urate oxidase or

uricosuric agent is selected from the group consisting of allopurinol, oxypurinol, febuxostat, pegloticase, probenecid, sulfinpyrazone, benzbromarone, lesinurad, pyrazinamide, pyrazinoate, aspirin, ethambutol, losartan, atorvastatin and fenofibrate.

4. The method of claims 2 or 3, wherein the xanthine oxidase inhibitor is allopurinol.

5. The method of claims 2 or 3 wherein the xanthine oxidase inhibitor is febuxostat.

6. The method of any of claims 1-5, wherein the subject is treated with from about 1 mg to about 500 mg of compound I.

7. The method of any of claims 1-6, wherein the subject is treated with from about 5 mg to about 240 mg of compound I.

8. The method of any of claims 1-7, wherein the subject is treated with from about 20 mg to about 80 mg of compound I.

9. The method of any of claims 1-8, wherein the subject is treated with about 20 mg of compound I.

10. The method of any of claims 1-8, wherein the subject is treated with about 40 mg of compound I.

11. The method of any of claims 1-8, wherein the subject is treated with about 80 mg of compound I.

12. The method of any of claims 4 or 6-11, wherein the subject is treated with from about 100 mg to about 300 mg of allopurinol.

13. The method of any of claims 4 or 6-12, wherein the subject is treated with about 200 mg of allopurinol.

14. The method of any of claims 4 or 6-13, wherein the subject is treated with about 300 mg of allopurinol.

15. The method of any of claims 1-14, wherein serum uric acid concentration in the

subject is reduced to less than 6.0 mg/dL.

16. The method of any of claims 1-15, wherein administration is oral.

17. The method of any of claims 1-16, wherein the subject has hyperuricemia.

18. The method of any of claims 1-17, wherein the subject is asymptomatic for

hyperuricemia.

19. The method of any of claims 1-18, wherein the subject has gout, hereditary

xanthinuria, hypoxanthine-guanine phosphoribosyltransferase deficiency, or Lesch- Nyhan syndrome.

20. The method of any of claims 1-19, wherein the subject has gout.

21. The method of any of claims 1-20, wherein the subject is a human.

22. A method for treating gout in a subject in need thereof comprising administering to said subject a therapeutically effective amount of compound I:

or pharmaceutically acceptable salt thereof.

23. The method of claim 22, further comprising administration of a xanthine oxidase inhibitor, a urate oxidase, or a uricosuric agent.

24. The method of claim 23, wherein the xanthine oxidase inhibitor, urate oxidase or uricosuric agent is selected from the group consisting of allopurinol, oxypurinol, febuxostat, pegloticase, probenecid, sulfinpyrazone, benzbromarone, lesinurad, pyrazinamide, pyrazinoate, aspirin, ethambutol, losartan, atorvastatin and fenofibrate.

25. The method of claims 23 or 24, wherein the xanthine oxidase inhibitor is allopurinol.

26. The method of claims 23 or 24, wherein the xanthine oxidase inhibitor is febuxostat.

27. The method of any of claims 22-26, wherein the subject is treated with from about 1 mg to about 500 mg of compound I.

28. The method of any of claims 22-27, wherein the subject is treated with from about 20 mg to about 240 mg of compound I.

29. The method of any of claims 22-28, wherein the subject is treated with from about 20 mg to about 80 mg of compound I.

30. The method of any of claims 22-29, wherein the subject is treated with about 20 mg of compound I.

31. The method of any of claims 22-30, wherein the subject is treated with about 40 mg of compound I.

32. The method of any of claims 22-31 , wherein the subject is treated with about 80 mg of compound I.

33. The method of any of claims 22-32, wherein the subject is treated with from about 100 mg to about 300 mg of allopurinol.

34. The method of any of claims 22-33, wherein the subject is treated with about 200 mg of allopurinol.

35. The method of any of claims 22-33, wherein the subject is treated with about 300 mg of allopurinol.

36. The method of any of claims 22-35, wherein serum uric acid concentration in the subject is reduced to less than 6.0 mg/dL.

37. The method of any of claims 22-36, wherein administration is oral.

38. The method of any of claims 22-37, wherein the subject is a human.

39. A method for treating hyperuricemia or an affliction related to hyperuricemia in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of compound I:

or pharmaceutically acceptable salt thereof, wherein the subject is undergoing a treatment regimen with a xanthine oxidase inhibitor.

40. The method of claim 39, wherein the treatment regimen with a xanthine oxidase inhibitor fails to reduce serum uric acid levels to less than 6.0 mg/dL.

41. The method of claims 39 or 40, wherein the subject is treated with from about 1 mg to about 240 mg of compound I.

42. The method of any of claims 39-41, wherein the subject is treated with from about 40 mg to about 160 mg of compound I.

43. The method of any of claims 39-42, wherein the subject is treated with about 40 mg of compound I.

44. The method of any of claims 39-42, wherein the subject is treated with about 20 mg of compound I.

45. The method of any of claims 39-42, wherein the subject is treated with about 10 mg of compound I.

46. The method of any of claims 37-39, wherein the subject is treated with about 5 mg of compound I.

47. The method of any of claims 39-46, wherein serum uric acid concentration in the subject is reduced to less than 6.0 mg/dL.

48. The method of any of claims 39-47, wherein the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, febuxostat or combinations thereof.

49. The method of any of claims 39-48, wherein the xanthine oxidase inhibitor is

allopurinol.

50. The method of any of claims 39-48, wherein the xanthine oxidase inhibitor is

febuxostat.

51. The method of any of claims 39-50, wherein the subject has gout, hereditary

xanthinuria, hypoxanthine-guanine phosphoribosyltransferase deficiency, or Lesch- Nyhan syndrome.

52. The method of any of claims 39-51 , wherein the subject has gout.

53. The method of any of claims 39-52, wherein the subject is human.

54. The method of any of claims 39-49 or 51-53, wherein the xanthine oxidase inhibitor is allopurinol, the treatment regimen with the xanthine oxidase inhibitor is 100 mg, 200 mg, or 300 mg of allopurinol daily and the subject is treated with about 5 mg, 10 mg, 20 mg, or 40 mg of compound I daily.

55. The method of any of claims 1-54, wherein said salt is selected from the group

consisting of hemisuccinate, hemifumarate, hemimalate salt, or hydrates of any of the foregoing.

56. The method of any of claims 1-55, wherein said salt is a hemisuccinate salt or a

hydrate thereof.

57. The use of compound I or a pharmaceutically acceptable salt thereof for manufacture of a medicament, wherein the medicament is prepared to be administered to reduce serum uric acid levels in a subject

58. A pharmaceutical composition comprising compound I:

or pharmaceutically acceptable salt thereof in conjunction with a pharmaceutically acceptable carrier.

59. The pharmaceutical composition of claim 58, further comprising a xanthine oxidase inhibitor or a uricosuric agent.

60. The pharmaceutical composition of claim 59, wherein the xanthine oxidase inhibitor is allopurinol.

61. The pharmaceutical composition of claim 59, wherein the xanthine oxidase inhibitor is febuxostat.

62. A pharmaceutical dosage form comprising compound I:

or pharmaceutically acceptable salt thereof in conjunction with a pharmaceutically acceptable carrier.

63. The pharmaceutical dosage form of claim 62, further comprising a xanthine oxidase inhibitor or a uricosuric agent. The pharmaceutical dosage form of claim 63, wherein the xanthine oxidase inhibitor is allopurinol.

The pharmaceutical dosage form of claim 63, wherein the xanthine oxidase inhibitor is febuxostat.

The pharmaceutical composition or dosage form of any of claims 58-65, wherein said salt is selected from the group consisting of hemisuccinate, hemifumarate, hemimalate salt, or hydrates of any of the foregoing.

The pharmaceutical composition or dosage form of any of claims 58-65, wherein said salt is a hemisuccinate salt or a hydrate thereof.

Description:
COMPOSITIONS AND METHODS FOR TREATMENT OF HYPERURICEMIA

[0001] This application claims priority to U.S. Provisional Application No. 61/450,771, filed on March 9, 2011 and U.S. Provisional Application No. 61/504,161, filed on July 1, 2011, both of which are incorporated herein by reference.

[0002] The disclosures of all patents, patent applications and publications cited herein are hereby incorporated by reference in their entirety.

BACKGROUND

[0003] Excessive blood concentrations of uric acid (hyperuricemia) lead to deposition of monosodium urate crystals in tissue, particularly the joints and kidneys. These crystals cause recurrent attacks of joint inflammation (arthritis). Gout is a common form of inflammatory arthritis caused by hyperuricemia, affecting approximately 8.3 million individuals in the United States. It is also is the most common form of inflammatory arthritis in males. Gout is a chronic, progressive disease that can lead to joint destruction, decreased kidney function and kidney stones.

[0004] Uric acid is a breakdown product of purines, which are present in many dietary constituents. Often, the ability of the body to process uric acid is a causative factor for gout; however, hyperuricemia is not always accompanied by manifestations of gout. This state of asymptomatic hyperuricemia is considered a precursor state to the development of gout.

[0005] Although there is presently no cure for gout, current treatments provide only palliative relief. Typical pharmaceutical treatments for gout include pain management such as analgesics, anti-inflammatory agents such as nonsteroidal anti-inflammatory drugs (NSAIDS), colchicine or corticosteroids, and medications for managing the underlying metabolic imbalance of elevated uric acid levels. Medications such as probenecid and sulfinpyrazone increase excretion of uric acid, while allopurinol lowers uric acid levels by preventing production of uric acid. While allopurinol is a popular therapeutic for reduction of uric acid, patient compliance is poor due to untoward side effects such as common flares, particularly during the initial treatment period, and kidney stones. Although many patients tolerate allopurinol therapy, most do not reach treatment goal for reduced levels of uric acid levels. Allopurinol is not recommended for the treatment of asymptomatic hyperuricemia. Probenecid, a uricosuric agent, requires dosing of several times daily and exhibits adverse drug-drug interactions. For example, probenecid accelerates oxypurinol excretion, making allopurinol less effective for patients being treated with both agents. Gout patients frequently have co-morbidities requiring multiple medications, thus increasing the risk of drug-drug interactions with probenecid. Febuxostat has been recently approved for chronic

management of hyperuricemia, requiring patient monitoring for uric acid levels and liver function. Recently, intravenous treatments with pegloticase (PEGylated uricase) to lower uric acid levels has been used for patients who have failed other conventional treatments. There remains a need for development of new uric acid lowering compounds and methods

SUMMARY OF THE INVENTION

[0006] The invention provides methods and compositions for reduction of uric acid levels in subjects, and methods and compositions that are useful for treating, suppressing and/or preventing afflictions related to elevated uric acid levels in subjects.

[0007] In one aspect, invention provides methods for reducing serum uric acid in a subject in need thereof comprising administering to the subject a therapeutically effective amount of compound I:

or a pharmaceutically acceptable salt thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor, a urate oxidase, or a uricosuric agent. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor or a uricosuric agent. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor.

[0008] In one aspect, invention provides methods for treating hyperuricemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of compound I, or a pharmaceutically acceptable salt thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor, a urate oxidase, or a uricosuric agent. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor or a uricosuric agent. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor.

[0009] In some embodiments, the methods further comprise administering to the subject an therapeutically effective amount of allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with a therapeutically effective amount of compound I, or a pharmaceutically acceptable salt thereof, or a composition comprising allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with compound I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

[0010] In some embodiments, the methods and compositions are useful for treating, suppressing and/or preventing gout in subjects. In some embodiments, the methods and compositions are useful for treating, suppressing and/or preventing hereditary xanthinuria in subjects. In some embodiments, the methods and compositions are useful for treating, suppressing and/or preventing hypoxanthine-guanine phosphoribosyltransferase deficiency (HPRT) in subjects. In some embodiments, the methods and compositions are useful for treating, suppressing and/or preventing Lesch-Nyhan syndrome in subjects.

[0011] The present invention is based, in part, on certain discoveries which are described more fully in the Examples section of the present application. For example, the present invention is based, in part, on the discovery that levels of serum uric acid were markedly reduced upon treatment with compound I alone, and in combination with allopurinol. Thus, in some embodiments, the present invention also provides methods for reducing serum uric acid levels in blood comprising treating a subject with compound I, or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention further provides methods for reducing serum uric acid levels in blood comprising treating a subject with allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in synergistic combination with compound I, or a pharmaceutically acceptable salt thereof.

[0012] In some embodiments, the present invention provides methods for treating hyperuricemia by combined administration of allopurinol and the compound of formula I in subjects wherein the use of either allopurinol or the compound of formula I alone does not reduce serum uric acid levels below about 6.0 mg/dL. Surprisingly, it has been found that when allopurinol is administered together with compound I to patients whose serum uric acid levels are poorly controlled on allopurinol alone, that serum uric acid levels are reduced. More surprisingly, it has been found that there is a synergism between allopurinol and the compound of formula I.

[0013] In some embodiments, the present invention provides methods for treating

hyperuricemia wherein the combination of allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof and compound I, or pharmaceutically acceptable salt thereof is synergistically effective.

[0014] In some embodiments, the compositions and/or methods reduce serum uric acid concentration. In some embodiments, the compositions and/or methods treat, suppress and/or prevent hyperuricemia. In some embodiments, the compositions and/or methods treat, suppress and/or prevent gout.

[0015] In some embodiments, the hyperuricemia is asymptomatic. In some embodiments, the hyperuricemia results in gout.

[0016] In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.

[0017] In some embodiments, the subject has gout, hereditary xanthinuria, hypoxanthine- guanine phosphoribosyltransferase (HPRT) deficiency, or Lesch-Nyhan Syndrome. In some embodiments, the subject has gout. In some embodiments, the subject has hereditary xanthinuria. In some embodiments, the subject has HPRT deficiency. In some embodiments, the subject has Lesch-Nyhan syndrome.

[0018] In some embodiments, the subject has uric acid levels above about 8.0 mg/dL. In some embodiments, the subject has uric acid levels above about 7.0 mg/dL. In some embodiments, the subject has uric acid levels above about 6.0 mg/dL. In some embodiments, the subject has uric acid levels above 8.0 mg/dL. In some embodiments, the subject has uric acid levels above 7.0 mg/dL. In some embodiments, the subject has uric acid levels above 6.0 mg/dL.

[0019] In some embodiments, the subject is being treated with or has been treated with a xanthine oxidase inhibitor, a urate oxidase or uricosuric agent. In some embodiments, the subject is being treated with or has been treated with a xanthine oxidase inhibitor or uricosuric agent. In some embodiments, the subject is being treated with or has been treated with a xanthine oxidase inhibitor. [0020] In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with allopurinol. In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with about 300 mg/day allopurinol. In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with about 200 mg/day allopurinol.

[0021] In some embodiments, compound I is administered as co-therapy with a xanthine oxidase inhibitor, a urate oxidase or uricosuric agent. In some embodiments, compound I is administered as co-therapy with a xanthine oxidase inhibitor or uricosuric agent. In some embodiments, compound I is administered as co-therapy with a xanthine oxidase inhibitor.

[0022] In some embodiments, compound I is administered as add-on therapy to treatment with a xanthine oxidase inhibitor, a urate oxidase or uricosuric agent. In some embodiments, compound I is administered as add-on therapy to treatment with a xanthine oxidase inhibitor or uricosuric agent. In some embodiments, compound I is administered as add-on therapy to treatment with a xanthine oxidase inhibitor.

[0023] In another aspect, the invention provides methods for reduction of cholesterol in a subject in need thereof comprising administering to the subject a therapeutically effective amount of compound I, or a pharmaceutically acceptable salt thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt thereof, and a

pharmaceutically acceptable carrier. In some embodiments, the methods reduce total cholesterol. In some embodiments, the methods reduce LDL cholesterol.

[0024] These and other embodiments of the invention are further described in the following sections of the application, including the Detailed Description, Examples, and Claims. Still other objects and advantages of the invention will become apparent by those of skill in the art from the disclosure herein, which are simply illustrative and not restrictive. Thus, other embodiments will be recognized by the ordinarily skilled artisan without departing from the spirit and scope of the invention.

SUMMARY OF FIGURES

[0025] FIG. 1 shows serum uric acid percentage change over time for monotherapy with compound I.

[0026] FIG. 2 shows synergistic reduction in mean sUA with compound I and allopurinol versus allopurinol monotherapy. [0027] FIG. 3 shows synergistic percent reductions in mean sUA with compound I and allopurinol versus allopurinol monotherapy.

[0028] FIG. 4 shows percentage of subjects in reaching goal sUA reduction (sUA < 6 mg/dL) with compound I in combination with allopurinol.

[0029] FIG. 5 shows percentage of subjects reaching goal sUA reduction (sUA < 6 mg/dL) with compound I added to allopurinol therapy.

[0030] FIG. 6 shows percentage reduction of A) total cholesterol, and B) LDL cholesterol in subjects treated with compound I.

DETAILED DESCRIPTION

[0031] In some embodiments, the invention provides methods and compositions for reduction of uric acid levels in subjects, and methods and compositions that are useful for treating, suppressing and/or preventing afflictions related to elevated uric acid levels, such as gout, in subjects.

[0032] In some embodiments, the methods comprise administering to the subject an therapeutically effective amount of compound I:

or a pharmaceutically acceptable salt thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the methods further comprise administration of a xanthine oxidase inhibitor or a uricosuric agent.

[0033] In some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in a therapeutically effective amount. In some embodiments, the compound of formula I or pharmaceutically acceptable salt thereof is a part of a

pharmaceutical composition, and may be delivered alone or in combination with other agents and a pharmaceutically acceptable carrier. In some embodiments, the other agent is an analgesic, anti-inflammatory agent, xanthine oxidase inhibitor, urate oxidase, uricosuric agent, uricostatic agent, other agent that increases excretion of uric acid, other agent that decreases the production of uric acid, or a combination thereof.

[0034] The compositions or methods may optionally comprise one or more additional agents selected from the group consisting of analgesics, anti-inflammatory agents, xanthine oxidase inhibitors, uricosuric agents, other agents that increase excretion of uric acid and other agents that prevent the production of uric acid. The methods and compositions are useful for treating, suppressing and/or preventing conditions in subjects that may arise from

hyperuricemia.

[0035] In some embodiments, the present invention relates to methods of treatment, suppression or and/or prevention of diseases or conditions relating to hyperuricemia comprising administration of a therapeutically effective amount of compound I, or pharmaceutically acceptable salt thereof.

[0036] In some embodiments, the present invention relates to methods of treatment, suppression or and/or prevention of diseases or conditions relating to hyperuricemia comprising administration of a therapeutically effective amount of a xanthine oxidase inhibitor, or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof in combination with a therapeutically effective amount of compound I, or pharmaceutically acceptable salt thereof.

[0037] In some embodiments, the present invention relates to methods of treatment, suppression and/or prevention of diseases or conditions relating to hyperuricemia comprising administration of a therapeutically effective amount of allopurinol, or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof in combination with a therapeutically effective amount of compound I, or pharmaceutically acceptable salt thereof.

[0038] In some embodiments, the present invention relates to compositions for treatment, suppression and/or prevention of diseases or conditions relating to hyperuricemia comprising a therapeutically effective amount of a xanthine oxidase inhibitor, or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof and compound I, or pharmaceutically acceptable salt thereof.

[0039] The present invention is based, in part, on the discovery that levels of serum uric acid were markedly reduced upon treatment with compound I alone or in combination with a xanthine oxidase inhibitor such as allopurinol. Thus, in some embodiments, the present invention provides methods for reducing serum uric acid levels in blood comprising of treating a subject with compound I alone or in combination with a xanthine oxidase inhibitor such as, for example, allopurinol. When used in combination, the particular sequence of administration of xanthine oxidase inhibitor and compound I is not important. Thus, in some embodiments, the xanthine oxidase inhibitor and compound I may be administered at the same time. In some embodiments, the xanthine oxidase inhibitor and compound I may be administered at different times. In some embodiments, the xanthine oxidase inhibitor and compound I may be administered sequentially. In some embodiments, the xanthine oxidase inhibitor is administered prior to administration of compound I. In some embodiments, compound I is administered prior to the administration of the xanthine oxidase inhibitor. In some embodiments, the subject is currently taking a xanthine oxidase inhibitor, such as allopurinol, and compound I is administered while the subject maintains treatment with the xanthine oxidase inhibitor.

[0040] In some embodiments, the present invention provides a method for treating hyperuricemia by combined use of xanthine oxidase inhibitor and compound I in subjects wherein the use of either xanthine oxidase inhibitor or compound I alone does not reduce serum uric acid levels below about 6.0 mg/dL. It has been found that when a xanthine oxidase inhibitor such as, for example, allopurinol is administered together with compound I to patients whose serum uric acid levels are poorly controlled on allopurinol alone, statistically significant increases in the proportion of subjects achieving serum uric acid levels below about 6.0 mg/dL are observed. In some embodiments, there is a synergistic effect between allopurinol and compound I.

[0041] In some embodiments, the present invention provides a method for treating, preventing or suppressing hyperuricemia or afflictions related to hyperuricemia by combined use of xanthine oxidase inhibitor and compound I in subjects wherein the use of either the xanthine oxidase inhibitor or the compound of formula I alone does not reduce serum uric acid levels below about 6.0 mg/dL. In some embodiments, the methods comprise

administering an amount of a xanthine oxidase inhibitor or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with an amount of compound I, or a pharmaceutically acceptable salt thereof to a subject to treat, prevent or suppress hyperuricemia. In one embodiment, a synergistic effect is observed between allopurinol and compound I. In some embodiments, the xanthine oxidase inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof and compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof are administered in a therapeutically effective amount. In some embodiments, the xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, along with compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, may be a part of a pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof. In some embodiments, the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, febuxostat and combinations thereof. In some embodiments, the xanthine oxidase inhibitor is allopurinol.

[0042] In some embodiments, the present invention provides a method for treating, preventing or suppressing hyperuricemia or afflictions related to hyperuricemia by administering compound I, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof, to a subject currently undergoing a treatment regimen with a xanthine oxidase inhibitor. In some embodiments, the treatment regimen with the xanthine oxidase inhibitor does not reduce serum uric acid levels below about 6.0 mg/dL or the subject is experiencing incomplete relief of symptoms from an affliction related to hyperuricemia. In some embodiments, the methods comprise administering an amount of compound I or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof to a patient undergoing a treatment regimen with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof to a subject to treat, prevent or suppress hyperuricemia. In some embodiments, a synergistic effect is observed between the xanthine oxidase inhibitor and compound I. In some embodiments, the xanthine oxidase inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof and compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof are administered in a therapeutically effective amount. In some embodiments, the xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, along with compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, may be a part of a pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof. In some embodiments, the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, febuxostat and combinations thereof. In some embodiments, the xanthine oxidase inhibitor is

allopurinol.

[0043] In some embodiments, the present invention provides a method for treating, preventing or suppressing gout or a symptom related to gout by administering compound I, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof, to a subject currently undergoing a treatment regimen with a xanthine oxidase inhibitor. In some embodiments, the treatment regimen with the xanthine oxidase inhibitor does not reduce serum uric acid levels below about 6.0 mg/dL or the subject is experiencing incomplete relief of symptoms from gout or a symptom related to gout, such as gout flares. In some

embodiments, the methods comprise administering an amount of compound I or a

pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof to a patient undergoing a treatment regimen with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof to a subject to treat, prevent or suppress gout or a symptom related to gout. In some embodiments, a synergistic effect is observed between the xanthine oxidase inhibitor and compound I. In some embodiments, the xanthine oxidase inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof and compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof are administered in a therapeutically effective amount. In some

embodiments, the xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, along with compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, may be a part of a

pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof. In some embodiments, the agent is a urate oxidase. In some embodiments, the urate oxidase is pegloticase. In some embodiments, the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, febuxostat and combinations thereof. In some embodiments, the xanthine oxidase inhibitor is allopurinol. [0044] In some embodiments, the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, and febuxostat. In some embodiments, the xanthine oxidase inhibitor is allopurinol. In some embodiments, the xanthine oxidase inhibitor is oxypurinol. In some embodiments, the xanthine oxidase inhibitor is febuxostat.

[0045] The xanthine oxidase inhibitors, urate oxidases, uricosuric agents and compound of formula I can be prepared in different forms, such as salts, hydrates, solvates, complexes, pro-drugs or salts of pro-drugs, and the invention includes compositions and methods encompassing all variant forms of the compounds.

[0046] Subjects treated with the compound of formula I also exhibited a reduction in total cholesterol and LDL cholesterol. Thus, in another aspect, the invention provides methods for reduction of cholesterol in a subject in need thereof comprising administering to the subject a therapeutically effective amount of compound I, or a pharmaceutically acceptable salt thereof, or a composition comprising compound I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the methods reduce total cholesterol. In some embodiments, the methods reduce LDL cholesterol.

Abbreviations and Definitions

[0047] The term "compound(s) of the invention" as used herein means compound I, and may include salts, tautomeric forms, pro-drugs, hydrates and/or solvates thereof. The compounds of the present invention are prepared in different forms, such as salts, hydrates, solvates, complexes, pro-drugs or salts of pro-drugs, and the invention includes compositions and methods encompassing all variant forms of the compounds. Compound I is described herein, and may include salts, tautomeric forms, pro-drugs, hydrates and/or solvates thereof.

[0048] The term "hyperuricemia" refers to elevated blood concentrations of uric acid.

Hyperuricemia may be primary, as in gout or hypoxanthine-guanine

phosphoribosyltransferase (HPRT) deficiency, or secondary to diseases such as, for example, acute and chronic leukemia, polycythemia vera, multiple myeloma, and psoriasis.

Hyperuricemia may occur with the use of diuretic agents, during renal dialysis, in the presence of renal damage, during starvation or reducing diets, and in the treatment of neoplastic disease where rapid resolution of tissue masses may occur. Urate crystal formation occurs when the extracellular uric acid concentration exceeds 6.8 mg/mL, and the goal of gout therapy is to reduce serum uric acid (sUA) concentrations to less than 6.0 mg/dL. [0049] The term "composition(s) of the invention" as used herein means compositions comprising compound I, or salts, tautomeric forms, hydrates, pro-drugs and solvates thereof. The compositions of the invention may further comprise xanthine oxidase inhibitors, urate oxidases and/or uricosuric agents, and may also include salts, tautomeric forms, hydrates, pro-drugs and solvates thereof. Exemplary xanthine oxidase inhibitors include allopurinol, oxypurinol, and febuxostat. Exemplary urate oxidases include pegloticase. Exemplary uricosuric agents include probenecid, sulfinpyrazone, benzbromarone, lesinurad,

pyrazinamide, pyrazinoate, aspirin, ethambutol, losartan, atorvastatin and fenofibrate.

[0050] The term "method(s) of the invention" as used herein means methods comprising treatment with the compounds and/or compositions of the invention.

[0051] The term "solvate" as used herein means a compound, or a pharmaceutically acceptable salt thereof, wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate."

[0052] A "pharmaceutical composition" refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts, tautomers, solvates, hydrates or prodrugs thereof, with other chemical components, such as physiologically acceptable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.

[0053] A "prodrug" or "pro-drug" refers to an agent which is converted into the parent drug in vivo. Pro-drugs are often useful because, in some situations, they are easier to administer than the parent drug. They are bioavailable, for instance, by oral administration whereas the parent drug is not. The pro-drug also has improved solubility in pharmaceutical compositions over the parent drug. For example, the compound carries protective groups which are split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing active compound or is oxidized or reduced in body fluids to release the compound. The term "pro-drug" may apply to such functionalities as, for example, the hydroxyl or amino functionalities of compounds. Pro-drugs may be comprised of structures wherein a hydroxyl group is masked as an alkoxy, ester, phosphate, sulfonate, or other derivative. Pro-drugs may also be comprised of structures wherein an amino group is derivatized as a carboxamide, carbamate, acetamide, acyloxy-alkyl carbamate, imine, enamine or Mannich base. Further examples of pro-drugs are discussed herein and, for example, by Alexander et al, J. Med. Chem. 1988, 31, 318 (hereby incorporated by reference in its entirety).

[0054] The term "pharmaceutically acceptable salt" is intended to include salts derived from inorganic or organic acids including, for example hydrochloric, hydrobromic, sulfuric, nitric, perchloric, phosphoric, formic, acetic, lactic, maleic, fumaric, succinic, tartaric, glycolic, salicylic, citric, methanesulfonic, benzenesulfonic, benzoic, malonic, trifluroacetic, trichloroacetic, naphthalene-2 sulfonic and other acids. Pharmaceutically acceptable salt forms may also include forms wherein the ratio of molecules comprising the salt is not 1 : 1. For example, the salt may comprise more than one inorganic or organic acid molecule per molecule of base, such as two hydrochloric acid molecules per molecule of compound of formula (I). As another example, the salt may comprise less than one inorganic or organic acid molecule per molecule of base, such as two molecules of compound of formula (I) per molecule of tartaric acid. Salts may also exist as solvates or hydrates. Pharmaceutically acceptable salts are also intended to encompass hemi-salts, wherein the ratio of

compound:acid is respectively 2: 1. Exemplary hemi-salts are those salts derived from acids comprising two carboxylic acid groups, such as malic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, glutaric acid, oxalic acid, adipic acid and citric acid. Other exemplary hemi-salts are those salts derived from diprotic mineral acids such as sulfuric acid. Exemplary preferred hemi-salts include, but are not limited to, hemimaleate, hemifumarate, and hemisuccinate. Other exemplary pharmaceutically acceptable salts are described herein.

[0055] The term "acid" contemplates all pharmaceutically acceptable inorganic or organic acids. Inorganic acids include mineral acids such as hydrohalic acids, such as hydrobromic and hydrochloric acids, sulfuric acids, phosphoric acids and nitric acids. Organic acids include all pharmaceutically acceptable aliphatic, alicyclic and aromatic carboxylic acids, dicarboxylic acids, tricarboxylic acids, and fatty acids. Preferred acids are straight chain or branched, saturated or unsaturated C1-C20 aliphatic carboxylic acids, which are optionally substituted by halogen or by hydroxyl groups, or C6-C12 aromatic carboxylic acids.

Examples of such acids are carbonic acid, formic acid, fumaric acid, acetic acid, propionic acid, isopropionic acid, valeric acid, alpha-hydroxy acids, such as glycolic acid and lactic acid, chloroacetic acid, benzoic acid, methane sulfonic acid, and salicylic acid. Examples of dicarboxylic acids include oxalic acid, malic acid, succinic acid, tataric acid and maleic acid. An example of a tricarboxylic acid is citric acid. Fatty acids include all pharmaceutically acceptable saturated or unsaturated aliphatic or aromatic carboxylic acids having 4 to 24 carbon atoms. Examples include butyric acid, isobutyric acid, sec-butyric acid, lauric acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid, and phenylsteric acid. Other acids include gluconic acid, glycoheptonic acid and lactobionic acid.

[0056] As used herein the term "about" is used herein to mean approximately, roughly, around, or in the region of. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term "about" is used herein to modify a numerical value above and below the stated value by a variance of 20 percent up or down (higher or lower).

[0057] An "effective amount", "sufficient amount" or "therapeutically effective amount" as used herein is an amount of a compound that is sufficient to effect beneficial or desired results, including clinical results. As such, the effective amount may be sufficient, for example, to reduce or ameliorate the severity and/or duration of the hyperuricemia, or one or more symptoms thereof, prevent the advancement of conditions related to hyperuricemia, prevent the recurrence, development, or onset of one or more symptoms associated with hyperuricemia, or enhance or otherwise improve the prophylactic or therapeutic effect(s) of another therapy. An effective amount also includes the amount of the compound that avoids or substantially attenuates undesirable side effects.

[0058] As used herein and as well understood in the art, "treatment" is an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results may include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminution of extent of disease, a stabilized (i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.

[0059] The term "in need thereof refers to the need for symptomatic or asymptomatic relief from a condition such as, for example, elevated levels of serum uric acid. The subject in need thereof may or may not be undergoing treatment for conditions related to elevated levels of serum uric acid.

[0060] The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which a compound is administered. Non-limiting examples of such pharmaceutical carriers include liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The pharmaceutical carriers may also be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. In addition, auxiliary, stabilizing, thickening, lubricating and coloring agents may be used. Other examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin (hereby incorporated by reference in its entirety).

[0061] The terms "animal," "subject" and "patient" as used herein include all members of the animal kingdom including, but not limited to, mammals, animals (e.g., cats, dogs, horses, swine, etc.) and humans.

[0062] The term "synergistic" or "synergistically effective" relates to the commonly used meaning in the art, and may pertain specifically to reduction of serum uric acid levels. In the present context, the terms "synergistically effective" or "synergistic effect" indicate that two or more compounds that are therapeutically effective, when used in combination, provide improved therapeutic effects greater than the additive effect that would be expected based on the effect of each compound used by itself.

Description

[0063] Gout is an inflammatory arthritis characterized by the presence of monosodium urate crystals in synovial fluid and is associated with hyperuricemia during the course of the disease (Bieber, J.D. et al, Arthritis Rheum. 2004, 50, 2400-14; hereby incorporated by reference in its entirety). Urate crystal formation occurs when the extracellular uric acid concentration exceeds 6.8 mg/mL, and the goal of gout therapy is to reduce serum uric acid (sUA) concentrations to less than 6.0 mg/dL. Hyperuricemia is a risk factor for destructive arthritis, renal disease, hypertension, cardiovascular disease, and each of the components of the metabolic syndrome (Feig, D. I. et al, N. Engl. J. Med. 2008, 359, 1811-21; Bieber, J.D. et al., Arthritis Rheum. 2004, 50, 2400-14; each of which are hereby incorporated by reference in their entirety).

[0064] Allopurinol acts on purine catabolism, reducing the production of uric acid via inhibition of uric acid biosynthesis. Allopurinol is a structural analogue of hypoxanthine, a natural purine base. Allopurinol inhibits xanthine oxidase, the enzyme responsible for conversion of hypoxanthine to xanthine, and of xanthine to uric acid. Allopurinol is also metabolized in humans to oxypurinol (alloxanthine), which is also an inhibitor of xanthine oxidase. While allopurinol lowers uric acid levels by preventing production of uric acid, doses must be adjusted to avoid xanthine lithiasis in some patients, and greater than 50% of gout patients treated with allopurinol fail to meet the therapeutic goal of serum uric acid levels less than 6.0 mg/dL. Additionally, allopurinol is not recommended for the treatment of asymptomatic hyperuricemia in the United States. Discontinuation of allopurinol therapy is frequently caused by adverse events such as gastrointestinal (diarrhea) and hypersensitivity issues (allopurinol hypersensitivity syndrome) (Sundy, J. S., Curr. Opin. Rheumatology 2010, 22, 188-193; hereby incorporated by reference in its entirety).

[0065] Xanthinuria is a rare disorder associated with xanthine dehydrogenase deficiency (also referred to as xanthine oxidoreductase), which catalyzes the conversion of hypoxanthine and xanthine to uric acid. Xanthinuria can occur secondary to therapy with xanthine oxidase inhibitors such as allopurinol (Simmonds, H. A., Hereditary xanthinuria. Orphanet

Encyclopedia, 1-4 (2003); hereby incorporated by reference in its entirety).

[0066] Deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT) activity is associated with purine metabolism, and associated with overproduction of uric acid. Uric acid elevation is thus present in all HPRT-deficient patients and is associated with gout and lithiasis, the most severe form of which is known as Lesch-Nyhan syndrome (Torres, R. J. et al, Orphanet Journal of Rare Diseases 2007, 2:48; hereby incorporated by reference in its entirety).

[0067] Purine nucleoside phosphorylase (PNP) controls the generation of uric acid precursors early in the metabolic pathway. PNP functions in the salvage/catabolism pathway, and reversibly catalyzes phosphoro lysis of dGuo and guanosine to guanine and deoxyribose 1- phosphate (or ribose-1 -phosphate in the case of guanosine), and deoxyinosine and inosine to hypoxanthine and deoxyribose 1 -phosphate (or ribose-1 -phosphate in the case of inosine). In PNP-deficient patients, decreases in uric acid in plasma and urine have been observed.

Compound I is an inhibitor of PNP, and treatment with compound I leads directly to a reduction in sUA. Compound I, when administered as monotherapy, has been shown to decrease serum uric acid levels in patients with gout. Compound I administered as co- therapy with allopurinol surprisingly shows synergistic reductions in serum uric acid levels.

[0068] Compound I is a 9-deazahypoxanthine derivative, and is described, for example, in U.S. Patent Nos. 7,553,839; 7,655,795; U.S. Patent Application No. 11/628,427; and

International Patent Publication No. WO 10/111381; each hereby incorporated by reference in its entirety. The compound I and structurally similar compounds are known as immucillins. Immucillins have been studied as PNP inhibitors (See, Evans et al., Org. Lett. 2003, 5, 3639; Taylor et al, J. Am. Chem. Soc. 2007, 129, 6984; Evans et al, J. Med. Chem. 2003, 46, 5271; Castilho et al, Bioorg. Med. Chem. 2006, 14, 516; Schramm et al, J. Biol. Chem. 2007, 282, 28297; and Bantia et al, Int. Immunopharmacol. 2010, 784; each hereby incorporated by reference in its entirety) and references therein. Some immucillins have also been studied as 5'-methylthioadenosine phosphorylase (MTAP) or 5'-methylthioadenosine nucleosidase (MTAN) inhibitors.

[0069] Exemplary forms of compound I are described in, for example, U.S. Patent Nos. 7,553,839; 7,655,795; U.S. Patent Application No. 11/628,427; and International Patent Publication No. WO 10/111381; each hereby incorporated by reference in its entirety. For example, WO 10/111381 relates to salt forms of compound I including, C 4 organic diacid salts, hemi salts, mono salts and mixtures and hydrates thereof. Representative C 4 organic diacids are succinic, fumaric, L-malic, maleic, L-tartaric, and L-aspartic acids.

Representative mixed salts are hemi(hemisuccinate, hemifumarate) monohydrate, hemi(hemisuccinate, hemimalate) monohydrate, hemi(hemifumarate, hemimalate) monohydrate, and hemi(l/3-succinate, 1/3-fumarate and 1/3-malate) monohydrate.

Anhydrous forms of each of the foregoing are also representative.

[0070] In some embodiments, compound I exists as a pharmaceutically acceptable salt. In some embodiments, the salt is selected from the group consisting of C 4 organic diacid salts, hemi salts, mono salts and mixtures and hydrates thereof. In some embodiments, the salt is selected from the group consisting of salts of succinic, fumaric, L-malic, maleic, L-tartaric, and L-aspartic acids. In some embodiments, the salt is selected from the group consisting of hemi salts of succinic, fumaric, L-malic, maleic, L-tartaric, and L-aspartic acids. In some embodiments, the salt is selected from the group consisting of hemi(hemisuccinate, hemifumarate) monohydrate, hemi(hemisuccinate, hemimalate) monohydrate,

hemi(hemifumarate, hemimalate) monohydrate, and hemi( 1/3 -succinate, 1/3-fumarate and 1/3-malate) monohydrate salts. In some embodiments, compound I exists as a hemi salt. In some embodiments, compound I exists as a succinate salt. In some embodiments, compound I exists as a hemisuccinate salt. In some embodiments, compound I exists as a hemisuccinate hydrate salt. In some embodiments, compound I exists as a hemisuccinate monohydrate salt. In some embodiments, compound I exists as a tautomer. In some embodiments, compound I exists as a solvate. In some embodiments, compound I exists as a hydrate. In some embodiments, compound I exists as a pro-drug. [0071] In some embodiments, the methods comprise administering to the subject a therapeutically effective amount of a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with a therapeutically effective amount of compound I, or a pharmaceutically acceptable salt thereof, or a composition comprising a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with compound I, or a pharmaceutically acceptable salt or hydrate thereof, and a pharmaceutically acceptable carrier. In some embodiments, the methods and compositions are useful for treating, suppressing and/or preventing gout in subjects.

[0072] In some embodiments, the methods and/or compositions of the invention comprise xanthine oxidase inhibitors. In some embodiments, the xanthine oxidase inhibitor is selected from the group consisting of allopurinol, oxypurinol, and febuxostat. In some embodiments, the xanthine oxidase inhibitor is allopurinol. In some embodiments, the xanthine oxidase inhibitor is oxypurinol. In some embodiments, the xanthine oxidase inhibitor is febuxostat.

[0073] Xanthine oxidase inhibitors such as, for example, allopurinol and/or the compound of formula I may exhibit tautomeric properties. Thus, the present invention also encompasses tautomeric forms, and mixtures thereof. It will further be appreciated that some compounds, including xanthine oxidase inhibitors, can exist as pharmaceutically acceptable salts, solvates, hydrates, tautomers or pro-drugs, each of which are also within the embodiments of the invention.

[0074] In some embodiments, the xanthine oxidase inhibitor exists as a pharmaceutically acceptable salt. In some embodiments, the xanthine oxidase inhibitor exists as a tautomer. In some embodiments, the xanthine oxidase inhibitor exists as a hydrate or solvate. In some embodiments, the xanthine oxidase inhibitor exists as a pro-drug.

[0075] The compounds and/or compositions of the disclosure therefore are useful in treating and/or preventing conditions related to elevated uric acid levels in a host or subject. The methods of the invention may be used in treating, suppressing and/or preventing disease states or conditions caused by and/or related to such elevated uric acid levels. Examples of such disease states or conditions include, but are not limited to, gout, acute gouty arthritis, hyperuricemia, uric acid nephropathy, nephrolithiasis, hereditary xanthinuria, HPRT deficiency and chronic gouty joint disease. [0076] In some embodiments, the compositions or methods may further comprise one or more additional agents in combination with compound I alone, or in combination with compound I and the xanthine oxidase inhibitor. Examples of such agents include, but are not limited to, analgesic agents, anti-inflammatory agents, xanthine oxidase inhibitors, urate oxidases, uricosuric agents, other agents that increase excretion of uric acid and agents that prevent the production of uric acid. Exemplary xanthine oxidase inhibitors include allopurinol, oxypurinol, and febuxostat. Exemplary uricosuric agents include probenecid, sulfinpyrazone, benzbromarone, lesinurad, pyrazinamide, pyrazinoate, aspirin, ethambutol, losartan, atorvastatin and fenofibrate. Exemplary urate oxidases include pegloticase.

[0077] Thus, in some embodiments the methods and/or compositions of the invention comprise uricosuric agents. In some embodiments, the uricosuric agent is selected from the group consisting of probenecid, sulfinpyrazone, benzbromarone, lesinurad, pyrazinamide, pyrazinoate, aspirin, ethambutol, losartan, atorvastatin and fenofibrate. In some

embodiments, the uricosuric agent is selected from the group consisting of probenecid, sulfinpyrazone, benzbromarone, lesinurad, and pyrazinamide. In some embodiments, the uricosuric agent is probenecid. In some embodiments, the uricosuric agent is aspirin. In some embodiments, the uricosuric agent is atorvastatin. In some embodiments, the uricosuric agent is fenofibrate. In some embodiments, the uricosuric agent is benzbromarone. In some embodiments, the uricosuric agent is sulfinpyrazone.

[0078] Exemplary anti-inflammatory agents include colchicine or one or more non-steroidal anti-inflammatory drugs ("NSAIDs"). NSAIDs used to treat subjects pursuant to the methods of the invention can be selected from the group consisting of: acetaminophen, amoxiprin, benorilate, choline magnesium salicylate, difunisal, faislamine, methyl salicylate, magnesium salicylate, salicyl salicylate, diclofenac, aceclofenac, acemetacin, bromfenac, etodolac, ketorolac, nabumetone, sulindac, tolmetin, ibuprofen, carprofen, fenbufen, fenoprofen, flurbiprofen, ketoprofen, loxoprofen, naproxen, tiaprofenic acid, mefenamic acid, meclofenamic acid, tolfenamic acid, phenylbutazone, azapropazone, metamizole,

oxyphenbutazone, piroxicam, lornoxicam, meloxicam, tenoxicam, celecoxib, etoricoxib, lumiracoxib, parecoxib, nimesulide, licofelone, indomethacin, pharmaceutically acceptable salts thereof and mixtures thereof.

[0079] Thus, in some embodiments, the methods of the invention further comprise administration of one or more additional analgesic or anti-inflammatory agents. In some embodiments, an additional analgesic or anti-inflammatory agent is selected from the group consisting of acetaminophen, amoxiprin, benorilate, choline magnesium salicylate, difunisal, faislamine, methyl salicylate, magnesium salicylate, salicyl salicylate, diclofenac, aceclofenac, acemetacin, bromfenac, etodolac, ketorolac, nabumetone, sulindac, tolmetin, ibuprofen, carprofen, fenbufen, fenoprofen, flurbiprofen, ketoprofen, loxoprofen, naproxen, tiaprofenic acid, mefenamic acid, meclofenamic acid, tolfenamic acid, phenylbutazone, azapropazone, metamizole, oxyphenbutazone, piroxicam, lornoxicam, meloxicam, tenoxicam, celecoxib, etoricoxib, lumiracoxib, parecoxib, nimesulide, licofelone, and indomethacin, or pharmaceutically acceptable salts thereof and mixtures thereof.

[0080] In some embodiments, the present invention provides methods for reducing serum uric acid levels in a subject in need thereof, comprising administration of compound I alone or further in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or pro-drug thereof.

[0081] In some embodiments, the present invention provides a method for treating a subject suffering from a gout comprising administering to said subject compound I alone or further in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or pro-drug thereof.

[0082] In some embodiments, the present invention provides methods for treating, preventing or suppressing gout. In some embodiments, the methods comprise administering compound I, or a pharmaceutically acceptable salt, hydrate, solvate, or pro-drug thereof to a subject to treat, prevent or suppress gout. In some embodiments, the methods further comprise identifying a subject in need of such treatment, prevention or suppression. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof is administered in a therapeutically effective amount. In some

embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof may be a part of a pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, xanthine oxidase inhibitor, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof.

[0083] In some embodiments, the present invention provides a method for suppressing manifestations of symptoms related to elevated uric acid concentrations in a subject comprising administering to the subject compound I alone or further in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof.

[0084] In some embodiments, the present invention provides methods for treating, preventing or suppressing hereditary xanthinuria. In some embodiments, the methods treating or suppress hereditary xanthinuria. In some embodiments, the methods comprise administering compound I, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof to a subject to treat, prevent or suppress hereditary xanthinuria. In some embodiments, the methods further comprise identifying a subject in need of such treatment, prevention or suppression. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof is administered in a therapeutically effective amount. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof may be a part of a

pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, xanthine oxidase inhibitor, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof.

[0085] In some embodiments, the present invention provides methods for treating, preventing or suppressing hypoxanthine-guanine phosphoribosyltransferase (HPRT) deficiency or a condition related to HPRT deficiency. In some embodiments, the methods treating or suppress HPRT deficiency or a condition related to HPRT deficiency. In some embodiments, the methods comprise administering compound I, or a pharmaceutically acceptable salt, hydrate, solvate, or pro-drug thereof to a subject to treat, prevent or suppress HPRT deficiency or a condition related to HPRT deficiency. In some embodiments, the methods further comprise identifying a subject in need of such treatment, prevention or suppression. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof is administered in a therapeutically effective amount. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof may be a part of a pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, xanthine oxidase inhibitor, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof. [0086] In some embodiments, the present invention provides methods for treating, preventing or suppressing Lesch-Nyhan syndrome. In some embodiments, the methods treating or suppress Lesch-Nyhan syndrome. In some embodiments, the methods comprise administering compound I, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof to a subject to treat, prevent or suppress Lesch-Nyhan syndrome. In some embodiments, the methods further comprise identifying a subject in need of such treatment, prevention or suppression. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof is administered in a therapeutically effective amount. In some embodiments, compound I or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof may be a part of a

pharmaceutical composition and may be delivered alone or with other agents in combination with a pharmaceutically acceptable carrier. In some embodiments, the agent is an analgesic, anti-inflammatory agent, xanthine oxidase inhibitor, uricosuric agent, other agent that increases excretion of uric acid, other agent that prevents the production of uric acid or a combination thereof.

[0087] In some embodiments, the present invention provides for the use of pharmaceutical compositions and/or medicaments comprised of compound I alone or further in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, or pro-drug thereof, in a method of treating a disease state, and/or condition caused by or related to hyperuricemia.

[0088] In some embodiments, the method of treatment comprises the steps of: i) identifying a subject in need of such treatment; (ii) providing compound I; and (iii) administering said compound I in a therapeutically effective amount to treat, suppress and/or prevent the hyperuricemia in a subject in need of such treatment.

[0089] In some embodiments, the method of treatment comprises the steps of: i) identifying a subject in need of such treatment; (ii) providing a composition comprising compound I; and (iii) administering said composition in a therapeutically effective amount to treat, suppress and/or prevent the hyperuricemia in a subject in need of such treatment.

[0090] In some embodiments, the method of treatment comprises the steps of: i) identifying a subject in need of such treatment; (ii) providing a xanthine oxidase inhibitor, or a

pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, and compound I; and (iii) administering said xanthine oxidase inhibitor and compound I in a therapeutically effective amount to treat, suppress and/or prevent the hyperuricemia in a subject in need of such treatment.

[0091] In some embodiments, the method of treatment comprises the steps of: i) identifying a subject in need of such treatment; (ii) providing a composition comprising a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, hydrate, solvate, pro-drug or tautomer thereof, and compound I and (iii) administering said composition in a therapeutically effective amount to treat, suppress and/or prevent the hyperuricemia in a subject in need of such treatment.

[0092] In some embodiments, the methods comprise administering to the subject an effective amount of compound I alone or in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof; or a composition comprising compound I alone or in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof, and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carriers are well-known to those skilled in the art, and include, for example, adjuvants, diluents, excipients, fillers, lubricants and vehicles. Often, the pharmaceutically acceptable carrier is chemically inert toward the active compounds and is non-toxic under the conditions of use. Examples of pharmaceutically acceptable carriers may include, for example, water or saline solution, polymers such as polyethylene glycol, carbohydrates and derivatives thereof, oils, fatty acids, or alcohols.

[0093] In some embodiments, the method of treatment, prevention and/or suppression of a condition related to hyperuricemia comprises the steps of: i) identifying a subject in need of such treatment; (ii) providing compound I alone or in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof; or a composition comprising compound I alone or in combination with a xanthine oxidase inhibitor, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or prodrug thereof, and a pharmaceutically acceptable carrier; and (iii) administering said compound(s) or composition in a therapeutically effective amount to treat, prevent and/or suppress hyperuricemia in a subject in need of such treatment.

[0094] In some embodiments, the invention provides a method for treating gout in a human in need thereof comprising administering compound I, or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof in an amount corresponding to from about 1 mg to about 240 mg of the free base of compound I.

[0095] In some embodiments, the invention provides a method for treating hyperuricemia in a subject in need thereof comprising administering to said subject a therapeutically effective amount of allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with a therapeutically effective amount of compound I, or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof.

[0096] In some embodiments, the invention provides a method for treating gout in a subject in need thereof comprising administering to said subject a therapeutically effective amount of allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in combination with a therapeutically effective amount of compound I, or

pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof.

[0097] In some embodiments, the invention provides a method for treating gout in a human in need thereof comprising administering allopurinol, or a pharmaceutically acceptable salt, tautomer, solvate, hydrate or pro-drug thereof in an amount corresponding to from about 100 mg to 300 mg of allopurinol free base; and compound I or pharmaceutically acceptable salt, tautomer, solvate, hydrate, or pro-drug thereof in an amount corresponding to from about 1 mg to about 80 mg of the free base of compound I.

[0098] Common co-morbidities associated with elevated serum uric acid concentrations and gout include, for example, obesity, hypertension, diabetes, and chronic kidney disease. Risks of drug-drug interactions thus exist in subjects treated for co-morbidities or other conditions. For example, currently available urate-lowering therapies such as colchicine, allopurinol, probenecid, in addition to anti-inflammatory therapies used for gout management are associated with drug-drug interactions. Treatment with compound I confers a low risk of drug-drug interactions with other medications. For example, compound I does not strongly induce or inhibit cytochrome P450 isoforms, has a low potential as a P-gp substrate or inducer, and is not a substrate or inhibitor of renal organic ion (anion and cation) transporters, indicating a low potential for hepatic and/or renal drug-drug interactions. Compound I also undergoes renal elimination and is not extensively metabolized in vivo, so the

pharmacokinetic profile should not be significantly altered by inhibitors of drug metabolizing enzymes. [0099] Thus, in some embodiments, compound I does not exhibit drug-drug interactions. In some embodiments, compound I does not exhibit drug-drug interactions with allopurinol or oxypurinol. In some embodiments, compound I does not exhibit drug-drug interactions with allopurinol.

[0100] In some embodiments, the present invention also encompasses methods comprising pro-drugs of a xanthine oxidase inhibitor and/or compound I and/or pharmaceutical compositions thereof. Pro-drugs include derivatives of compounds that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound of the invention. Examples of pro-drugs include, but are not limited to, derivatives and metabolites of a compound of the invention that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, and biohydrolyzable phosphate analogues. Prodrugs are also described in, for example, The Practice of Medicinal Chemistry (Camille Wermuth, ed., 1999, Academic Press; hereby incorporated by reference in its entirety). In certain embodiments, pro-drugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid. The carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule. Pro-drugs can typically be prepared using well-known methods, such as those described by Burger 's Medicinal Chemistry and Drug Discovery 6 th ed. (Donald J. Abraham ed., 2001, Wiley) and Design and Application of Pro-drugs (H. Bundgaard ed., 1985, Harwood Academic

Publishers Gmfh; each of which are hereby incorporated by reference in their entirety).

Biohydrolyzable moieties 1) do not interfere with the biological activity of the compound but can confer upon that compound advantageous properties in vivo, such as uptake, duration of action, or onset of action; or 2) are biologically inactive but are converted in vivo to the biologically active compound. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters. Examples of biohydrolyzable amides include, but are not limited to, lower alkyl amides, a-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. Examples of biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and

heteroaromatic amines, and polyether amines.

[0101] In some embodiments, the compounds of the invention are formulated into pharmaceutical compositions for administration to subjects in a biologically compatible form suitable for administration in vivo. According to another aspect, the present invention provides a pharmaceutical composition comprising a xanthine oxidase inhibitor and/or compound I in admixture with a pharmaceutically acceptable diluent and/or carrier. The pharmaceutically-acceptable carrier is "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. The pharmaceutically-acceptable carriers employed herein may be selected from various organic or inorganic materials that are used as materials for pharmaceutical formulations and which are incorporated as analgesic agents, buffers, binders, disintegrants, diluents, emulsifiers, excipients, extenders, glidants, solubilizers, stabilizers, suspending agents, tonicity agents, vehicles and viscosity-increasing agents. Pharmaceutical additives, such as antioxidants, aromatics, colorants, flavor-improving agents, preservatives, and sweeteners, may also be added. Examples of acceptable pharmaceutical carriers include carboxymethyl cellulose, crystalline cellulose, glycerin, gum arabic, lactose, magnesium stearate, methyl cellulose, powders, saline, sodium alginate, sucrose, starch, talc and water, among others. In some embodiments, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.

[0102] Surfactants such as, for example, detergents, are also suitable for use in the formulations. Specific examples of surfactants include polyvinylpyrrolidone, polyvinyl alcohols, copolymers of vinyl acetate and of vinylpyrrolidone, polyethylene glycols, benzyl alcohol, mannitol, glycerol, sorbitol or polyoxyethylenated esters of sorbitan; lecithin or sodium carboxymethylcellulose; or acrylic derivatives, such as methacrylates and others, anionic surfactants, such as alkaline stearates, in particular sodium, potassium or ammonium stearate; calcium stearate or triethanolamine stearate; alkyl sulfates, in particular sodium lauryl sufate and sodium cetyl sulfate; sodium dodecylbenzenesulphonate or sodium dioctyl sulphosuccinate; or fatty acids, in particular those derived from coconut oil, cationic surfactants, such as water-soluble quaternary ammonium salts of formula N R'R"R'"R""Y " , in which the R radicals are identical or different optionally hydroxylated hydrocarbon radicals and Y " is an anion of a strong acid, such as halide, sulfate and sulfonate anions;

cetyltrimethylammonium bromide is one of the cationic surfactants which can be used, amine salts of formula N R'R'R", in which the R radicals are identical or different optionally hydroxylated hydrocarbon radicals; octadecylamine hydrochloride is one of the cationic surfactants which can be used, non-ionic surfactants, such as optionally polyoxyethylenated esters of sorbitan, in particular Polysorbate 80, or polyoxyethylenated alkyl ethers;

polyethylene glycol stearate, polyoxyethylenated derivatives of castor oil, polyglycerol esters, polyoxyethylenated fatty alcohols, polyoxyethylenated fatty acids or copolymers of ethylene oxide and of propylene oxide, amphoteric surfactants, such as substituted lauryl compounds of betaine,

[0103] When administered to a subject, the xanthine oxidase inhibitor and/or compound I and pharmaceutically acceptable carrier can be sterile. Suitable pharmaceutical carriers may also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, polyethylene glycol 300, water, ethanol, polysorbate 20, and the like. The present compositions, if desired, may also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.

[0104] The pharmaceutical formulations of the present invention are prepared by methods well-known in the pharmaceutical arts. Optionally, one or more accessory ingredients (e.g., buffers, flavoring agents, surface active agents, and the like) also are added. The choice of carrier is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.

[0105] Additionally, the compounds and/or compositions of the present invention are administered to a human or animal subject by known procedures including oral

administration, sublingual or buccal administration. In some embodiments, the compound or composition is administered orally.

[0106] For oral administration, a formulation of the compounds of the invention may be presented in dosage forms such as capsules, tablets, powders, granules, or as a suspension or solution. Capsule formulations may be gelatin, soft-gel or solid. Tablets and capsule formulations may further contain one or more adjuvants, binders, diluents, disintegrants, excipients, fillers, or lubricants, each of which are known in the art. Examples of such include carbohydrates such as lactose or sucrose, dibasic calcium phosphate anhydrous, corn starch, mannitol, xylitol, cellulose or derivatives thereof, microcrystalline cellulose, gelatin, stearates, silicon dioxide, talc, sodium starch glycolate, acacia, flavoring agents,

preservatives, buffering agents, disintegrants, and colorants. Orally administered

compositions may contain one or more optional agents such as, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of wintergreen, or cherry; coloring agents; and preservative agents, to provide a pharmaceutically palatable preparation.

[0107] In some embodiments, the composition is in unit dose form such as a tablet, capsule or single-dose vial. Suitable unit doses, i.e., therapeutically effective amounts, may be determined during clinical trials designed appropriately for each of the conditions for which administration of a chosen compound is indicated and will, of course, vary depending on the desired clinical endpoint.

[0108] In accordance with the methods of the present invention, the compounds of the invention are administered to the subject in an amount effective to limit or decrease the level of serum uric acid in the subject. This amount is readily determined by the skilled artisan, based upon known procedures, including analysis of titration curves established in vivo and methods and assays disclosed herein.

[0109] In some embodiments, the present invention provides a method for treating, suppressing and/or preventing hyperuricemia by combined use of a xanthine oxidase inhibitor and compound I in subjects wherein the use of either a xanthine oxidase inhibitor or compound I alone does not reduce serum uric acid levels below about 6.0 mg/dL.

Surprisingly, it has been found that when a xanthine oxidase inhibitor such as allopurinol is administered together with compound I to patients whose serum uric acid levels are poorly controlled on allopurinol alone, statistically significant increases in the proportion of subjects achieving serum uric acid levels below about 6.0 mg/dL are observed. In some

embodiments, there is a synergistic effect between a xanthine oxidase inhibitor, such as allopurinol, and compound I. Thus, in some embodiments, a xanthine oxidase inhibitor and compound I are administered in amounts that exhibit synergistic lowering of serum uric acid levels. In some embodiments, a xanthine oxidase inhibitor and compound I are administered in amounts that exhibit synergistic treatment, suppression and/or prevention of

hyperuricemia. In some embodiments, a xanthine oxidase inhibitor and compound I are administered in amounts that exhibit synergistic treatment, suppression and/or prevention of gout.

[0110] In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with allopurinol or another xanthine oxidase inhibitor. In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with about 300 mg/day allopurinol. In some embodiments, the subject has uric acid levels above about 6.0 mg/dL following monotherapy with about 200 mg/day allopurinol or 100 mg/day allopurinol.

[0111] In some embodiments, the serum uric acid levels are reduced to from about 4.0 to about 6.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to from about 4.5 to about 5.8 mg/dL. In some embodiments, the serum uric acid levels are reduced to from about 5.0 to about 6.0 mg/dL.

[0112] In some embodiments, the serum uric acid levels are reduced to less than 6.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than 6.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than 5.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than 5.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than 4.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than 4.0 mg/dL.

[0113] In some embodiments, the serum uric acid levels are reduced to less than about 6.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than about 6.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than about 5.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than about 5.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than about 4.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to less than about 4.0 mg/dL.

[0114] In some embodiments, the serum uric acid levels are reduced to 6.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to 5.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to 5.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to 4.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to 4.0 mg/dL.

[0115] In some embodiments, the serum uric acid levels are reduced to about 6.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to about 5.5 mg/dL. In some embodiments, the serum uric acid levels are reduced to about 5.0 mg/dL. In some embodiments, the serum uric acid levels are reduced to about 4.5 mg/dL. In some

embodiments, the serum uric acid levels are reduced to about 4.0 mg/dL.

[0116] The precise dose to be employed in the compositions will also depend on the route of administration, and the seriousness of the affliction or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. In specific embodiments of the invention, suitable dose ranges for oral administration of a xanthine oxidase inhibitor such as, for example, allopurinol are generally about 50 mg/day to about 500 mg/day. In some embodiments, the oral dose of the xanthine oxidase inhibitor is about 100 mg/day to about 300 mg/day. In some embodiments, the oral dose of the xanthine oxidase inhibitor is about 100 mg/day to about 200 mg/day. In some embodiments, the oral dose of the xanthine oxidase inhibitor is about 100 mg/day. In some embodiments, the oral dose of the xanthine oxidase inhibitor is about 200 mg/day. In some embodiments, the oral dose of the xanthine oxidase inhibitor is about 300 mg/day.

[0117] In specific embodiments of the invention, suitable dose ranges for administration of compound I are generally about 1 mg/day to about 500 mg/day. In some embodiments, the dose of compound I is about 5 mg/day to about 500 mg/day. In some embodiments, the dose of compound I is about 1 mg/day to about 240 mg/day. In some embodiments, the dose of compound I is about 5 mg/day to about 240 mg/day. In some embodiments, the dose of compound I is about 5 mg/day to about 160 mg/day. In some embodiments, the dose of compound I is about 5 mg/day to about 160 mg/day. In some embodiments, the dose of compound I is about 5 mg/day to about 80 mg/day. In some embodiments, the dose of compound I is about 10 mg/day to about 160 mg/day. In some embodiments, the dose of compound I is about 10 mg/day to about 240 mg/day. In some embodiments, the dose of compound I is about 20 mg/day to about 40 mg/day. In some embodiments, the dose of compound I is about 20 mg/day to about 80 mg/day. In some embodiments, the dose of compound I is about 20 mg/day to about 120 mg/day. In some embodiments, the dose of compound I is about 20 mg/day to about 160 mg/day. In some embodiments, the dose of compound I is about 40 mg/day to about 80 mg/day. In some embodiments, the dose of compound I is about 40 mg/day to about 120 mg/day. In some embodiments, the dose of compound I is about 40 mg/day to about 160 mg/day. In some embodiments, the dose of compound I is about 40 mg/day to about 240 mg/day. In some embodiments, the dose of compound I is about 5 mg/day. In some embodiments, the dose of compound I is about 10 mg/day. In some embodiments, the dose of compound I is about 20 mg/day. In some embodiments, the dose of compound I is about 40 mg/day. In some embodiments, the dose of compound I is about 60 mg/day. In some embodiments, the dose of compound I is about 80 mg/day. In some embodiments, the dose of compound I is about 120 mg/day. In some embodiments, the dose of compound I is about 160 mg/day. In some embodiments, the dose of compound I is about 240 mg/day. In some embodiments, the dose of compound I is about 300 mg/day. In some embodiments, the dose of compound I is about 400 mg/day. In some embodiments, the dose of compound I is about 500 mg/day. In some embodiments, the doses are used when compound I is administered alone. In some embodiments, the doses are used when compound I is administered in combination with a xanthine oxidase inhibitor. In some embodiments, the doses are administered orally.

[0118] The xanthine oxidase inhibitor and compound I (or pharmaceutically acceptable salts, tautomers, solvates, hydrates or pro-drugs of either xanthine oxidase inhibitor or compound I or both) can be administered at different times or at the same time.

[0119] In some embodiments, the compositions of the invention comprise a compound of formula I and a xanthine oxidase inhibitor, a urate oxidase, or a uricosuric agent. In some embodiments, the compound of formula I and xanthine oxidase inhibitor, urate oxidase, or uricosuric agent are present together in a single dosage form such as, for example, an oral dosage form.

[0120] In some embodiments, the dose of the xanthine oxidase inhibitor is 100 mg, 200 mg or 300 mg daily and the dose of compound I is 5, 10, 20, 40, 60, 80, 100, 120, 140, 160, 180, 200, 220 or 240 mg daily. In specific embodiments, the xanthine oxidase inhibitor is allopurinol administered at 300 mg daily and compound I is administered at 5, 10, 20, 40 or 80 mg daily.

[0121] The present invention also provides articles of manufacture for treating, suppressing, and/or preventing disorders related to elevated uric acid levels, such as gout, in a subject. The articles of manufacture comprise a pharmaceutical composition of compound I, optionally further containing at least one additional uricostatic or uricosuric compound, as described herein. The articles of manufacture are packaged with indications for various disorders that the pharmaceutical compositions are capable of treating and/or preventing. For example, the articles of manufacture comprise a unit dose of a compound disclosed herein that is capable of treating or preventing a certain disorder, and an indication that the unit dose is capable of treating or preventing a certain disorder, for example elevated levels of uric acid.

[0122] In some embodiments, the invention provides compound I or a pharmaceutically acceptable salt thereof for use in reducing serum uric acid levels in a subject.

[0123] In some embodiments, the invention provides compound I or a pharmaceutically acceptable salt thereof for use in treatment of gout in a subject. [0124] In some embodiments, the invention provides compound I or a pharmaceutically acceptable salt thereof for use in treatment of hyperuricemia or an affliction related to hyperuricemia in a subject.

[0125] In some embodiments, the invention provides for the use of compound I or a pharmaceutically acceptable salt thereof for manufacture of a medicament, wherein the medicament is prepared to be administered to reduce serum uric acid levels in a subject.

[0126] In some embodiments, the invention provides for the use of compound I or a pharmaceutically acceptable salt thereof for manufacture of a medicament, wherein the medicament is prepared to be administered to treat gout in a subject.

[0127] In some embodiments, the invention provides for the use of compound I or a pharmaceutically acceptable salt thereof for manufacture of a medicament, wherein the medicament is prepared to be administered in treatment of hyperuricemia or an affliction related to hyperuricemia in a subject.

[0128] In some embodiments, the use further comprises a xanthine oxidase inhibitor, a urate oxidase, or a uricosuric agent.

[0129] In some embodiments, the medicament is administered in a dosage of from about 1 mg to about 500 mg of compound I. In some embodiments, the medicament is administered in a dosage of from about 5 mg to about 240 mg of compound I. In some embodiments, the medicament is administered in a dosage of from about 20 mg to about 80 mg of compound I. In some embodiments, the medicament is administered in a dosage of about 20 mg of compound I. In some embodiments, the medicament is administered in a dosage of about 40 mg of compound I. In some embodiments, the medicament is administered in a dosage of about 80 mg of compound I.

[0130] In some embodiments, the medicament is administered with about 100 mg to about 300 mg of allopurinol. In some embodiments, the medicament is administered with about 100 mg of allopurinol. In some embodiments, the medicament is administered with about 200 mg of allopurinol. In some embodiments, the medicament is administered with about 300 mg of allopurinol.

[0131] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be within the scope of the present invention. [0132] The invention is further described by the following non-limiting Examples.

EXAMPLES

[0133] Example 1: Effects on serum uric acid concentrations in gout patients via treatment with compound I (I).

[0134] The purpose of this study was to evaluate the sUA-lowering effects, safety and tolerability of compound I at 40, 80, 120, 160, and 240 mg monotherapy administered orally once daily for 3 weeks in gout patients whose untreated sUA was >8.0mg/dL (Fitzpatrick et al, 74 th ACR Scientific Meeting 2010, Atlanta; and Fitzpatrick, D. et al, Arthritis. Rheum. 2010; 62(10): S63 Abstract # 150; hereby incorporated by reference in its entirety).

[0135] Methods: Eligible subjects were male or female, aged 18-69 with ARA diagnosis of gout and baseline sUA > 8.0mg/dL, no active cardiac disease, gout flare, elevated liver enzymes, abnormal hemogram, CD4+ cell count < 500 cells/mm 3 , estimated creatinine clearance > 60 mL/min, and able to tolerate colchicine or naproxen flare prophylaxis. Part 1 : a parallel group (N=60) was randomized into four groups of N=15. Each group was administered either placebo, 40 mg/day, 80 mg/day or 120 mg/day of compound I. In part 2, a second group (N=60) was subjected to a sequential dose-escalation study with compound I at 160 mg/day (N=15) or placebo (N=5), compound I at 240 mg/day (N=15) or placebo (N=5), and compound I at 320 mg/day (N=15) or placebo (N=5). The 320 mg/day dosage was not performed due to efficacy criteria being met at a lower dose. The study design was a 30 day screening, with baseline within 7 days of the start of 21 day treatment with compound I. Days 22-50 follow up. Serum uric acid and safety labs and lymphocyte subsets were performed at baseline and days 2, 8, 15, 22, 29, 36, 43, and 50. Sparse PK day 1-2, and trough drug levels at days 8, 15, 22, 29, and 43. Adverse events were recorded at every visit.

[0136] Results: 96% of placebo and 95% of compound I treated subjects completed the study. One subject (160 mg/day) discontinued due to elevated CPK (also present at baseline). One subject (240 mg/day) discontinued at day 16 due to diarrhea, and one on day 2 due to an ectopic atrial rhythm. Gout flares greater than or equal to moderate severity requiring treatment occurred in 8 subjects (2 placebo, 6 compound I treated) during the treatment phase, and 3 subjects (all compound I treated) during follow-up to day 50.

[0137] Table 1. Demographics and Baseline Values

Placebo Compound I

40 mg/day 80 mg/day 120 160 240 Total (N=24) (N=15) (N=14) mg/day mg/day mg/day (N=75)

(N=16) (N=15) (N=15)

Age, mean (SD) 51 (9.9) 52 (11.9) 51 (11.7) 51 (13.6) 48 (10.5) 49 (10.9) 50 (11.6)

Male, n (%) 22 (92%) 15 (100%) 13 (93%) 15 (94%) 15 (100%) 15 (100%) 73 (97%)

White, n (%) 17 (71%) 10 (67%) 9 (64%) 10 (63%) 11 (73%) 8 (53%) 48 (64%)

Black/African

2 (8%) 3 (20%) 2 (14%) 1 (6%) 1 (7%) 2 (13%) 9 (12%) American, n (%)

Asian/Pacific

5 (21%) 2 (13%) 3 (21%) 5 (31%) 3 (20%) 5 (33%) 18 (24%) Islander/Other

BMI, kg/m2,

34.9 (5.23) 34.6 (9.87) 33.7 (4.87) 35.4 (8.52) (1 3 2 3 9 6 4) 32.8 (6.81) 34.1 (8.85) mean (SD)

Baseline sUA, mean

9.7 (1.04) 9.2 (0.71) 9.5 (0.84) 9.8 (1.36) 10.1 (1.76) 9.7 (1.51) — (SD)

Baseline Lymphocyte Subsets, count/mm3, median (range)

887 (622, 837 (607, 801 (541, 870 (506, 1106 (524, 848 (505,

CD4+ —

1678) 2201) 1539) 1312) 2135) 1483)

477 (169, 500 (230, 421 (194, 391 (171, 504 (149, 406 (175,

CD8+ —

788) 688) 704) 674) 1523) 697)

273 (85, 273 (81, 216 (97, 207 (73, 281 (92, 245 (107,

CD20+ —

649) 540) 714) 313) 690) 929)

189 (54, 254 (166, 192 (61, 163 (86, 180 (103, 283 (98,

CD56+ —

763) 623) 475) 390) 400) 472)

[0138] Table 2. Compound I Monotherapy: Absolute Values, Change from Baseline in Serum Uric Acid at Day 22

Placebo Compound I

40 mg/day 80 mg/day 120 mg/day 160 mg/day 240 mg/day (N=24) (N=15) (N=14) (N=16) (N=15) (N=15) sUA Mean (SD),

9.5 (1.35) 6.6 (1.13) 6.0 (0.89) 6.5 (1.50) 6.2 (1.72) 5.1 (1.46) mg/dL

CFB to Day 22

LS Means

-0.2 (0.27) -3.0 (0.35) -3.6 (0.36) -3.2 (0.33) -3.6 (0.35) -4.6 (0.34) (SEM) a ' b , mg/dL

Pairwise Comp arisons to Placebo

LS Mean

— -2.8 -3.4 -3.1 -3.4 -4.4

Difference 0 , mg/dL

95% CI — -3.7, -1.9 -4.3, -2.5 -3.9, -2.2 -4.3, -2.6 -5.3, -3.5 a Least Square (LS) Means from ANCOVA model with factors for treatment and baseline sUA.

bp < 0.001 for overall treatment effect by ANCOVA.

cp < 0.001 for pairwise differences between placebo and each dose of compound I.

[0139] Serum uric acid percentage change over time is shown in Figure 1.

[0140] Table 3: Proportion of Subjects Achieving Serum Uric Acid Levels < 6, < 5 and < 4 mg/dL at Day 22.

Placebo Compound I

40 mg/day 80 mg/day 120 mg/day 160 mg/day 240 mg/day All Subjects N=24 N=15 N=14 N=16 N=15 N=15

Day 22

<6.0 mg/dL a 0% 33%* 36%* 31%* 47%** 77%**

<5.0 mg/dL a 0% 0% 21% 25% 33%* 54%**

<4.0 mg/dL b 0% 0% 0% 6% 7% 23%

Subjects with BL

N=14 N=13 N=10 N=8 N=7 N=9 sUA <10 mg/dL

Day 22

<6.0 mg/dL a 0% 38% 30% 63%* 29% 89%*

<5.0 mg/dL a 0% 0% 10% 50%* 29% 67%*

<4.0 mg/dL b 0% 0% 0% 13% 14% 33% a P<0.001, t P<0.01 Overall Treatment Effect P-value by the Cochran- Armitage-Trend Test.

P<0.01, P<0.001 for comparison against placebo by Fisher's Exact test.

[0141] Table 4: First-Dose Pharmacokinetic Parameters for Compound I.

Cmax, ng/mL t 1ir AUCo-24, ng-h/mL C 2 4, ng/mL

Compound I Dose, mg/day

Mean (SD) Mean (SD) Mean (SD) Mean (SD)

40 13.0 (13.1) 3.9 (1.8) 327 (183) 10.6 (4.6)

80 22.8 (16.6) 4.5 (1.7) 453 (363) 17.1 (21.0)

120 35.2 (19.7) 4.4 (2.0) 547 (241) 12.7 (8.0)

160 46.5 (25.0) 4.1 (1.5) 678 (365) 18.4 (9.5)

240 71.3 (27.8) 4.0 (1.3) 1024 (385) 24.3 (12.0)

[0142] Table 5: Overview of Adverse Events.

Number of subjects (%)

Category

Placebo, N=24 Compound I (total), N=75

All AEs 15 (63%) 50 (67%)

All AEs at Least Possibly Related to

6 (25%) 25 (33%)

Study Drug

All Severe AEs a 3 (13%) 2 (3%)

All Serious AEs 0 1 (i%)

Withdrawals from Study Medication

1 (4%) 3 (4%)

Due to AEs

Deaths 0 0

All grades, all relationships to drug.

Note: Subjects may fall into more than 1 category.

aThe serious AE was hemorrhoidal bleeding 11 days after treatment with 40 mg/d compound I.

[0143] Table 6: Adverse Events that Occurred in > 1 Subject Per Dose Group, N(%) Placebo Compound I

Any adverse event 15 (63%) 10 (67%) 9 (64%) 10 (63%) 10 (67%) 1 1 (73%) 50 (67%)

Decreased lymphocytes 1 (4%) 0 2 (14%) 0 2 (13%) 1 (7%) 5 (7%)

Blood CPK increased 2 (8%) 0 1 (7%) 0 1 (7%) 1 (7%) 3 (4%)

Blood glucose increased 0 0 0 2 (13%) 0 1 (7%) 3 (4%)

Back pain 2 (8%) 1 (7%) 1 (7%) 2 (13%) 0 0 4 (5%)

URTI 1 (4%) 1 (7%) 2 (14%) 2 (13%) 0 1 (7%) 6 (8%)

Nasopharyngitis 3 (13%) 1 (7%) 1 (7%) 1 (6%) 0 0 3 (4%)

Gastroenteritis 3 (13%) 0 0 0 0 0 0

Edema peripheral 0 1 (7%) 1 (7%) 2 (13%) 1 (7%) 2 (13%) 7 (9%)

Fatigue 3 (13%) 0 2 (14%) 1 (6%) 1 (7%) 0 4 (5%)

Headache 4 (17%) 5 (33%) 1 (7%) 2 (13%) 0 2 (13%) 10 (13%)

Diarrhea 2 (8%) 1 (7%) 0 1 (6%) 1 (7%) 3 (20%) 6 (8%)

Cough 0 0 0 0 2 (13%) 0 2 (3%)

Rash 0 1 (7%) 0 0 0 2 (13%) 3 (4%)

Hypertension 1 (4%) 2 (13%) 0 0 0 0 2 (3%)

All grades, all relationships to drug.

[0144] Table 7. Lymphocyte Subset Percent Change from Baseline at Day 22 - Median (Range in %).

Placebo Compound I

** P<0.001, by ANCOVA with factors for treatment and baseline subset counts.

[0145] Table 8. Categorical Summary of CD4+ Cell Counts < 350 cells^L.

Placebo Compound I

40 mg/day 80 mg/day 120 mg/day 160 mg/day 240 mg/day

(N=24) (N=15) (N=14) (N=16) (N=15) (N=15)

Day 22 0/24 (0%) 0/15 (0%) 2/14 (14%) 0/16 (0%) 1/15 (7%) 1/13 (8%)

At Any

0/24 (0%) 0/15 (0%) 3/14 (21%) * 3/16 (19%) 1/15 (7%) 1/15 (7%) Time

P<0.05, for comparison to placebo by Fisher's Exact test.

No subject discontinued the drug treatment for a confirmed CD4+ cell count of less than 350 cells^L

[0146] Conclusions: Compound I dosed at 40, 80 120, 160, and 240 mg/day rapidly and significantly reduced sUA in gout subjects. There was a dose-related increase in the proportion of subjects who reach sUA <6.0 mg/dL and lower. First-dose exposures of compound I were dose proportional between 40 and 240 mg/day. Compound I was safe and well-tolerated. AEs were similar in frequency and severity between compound I and placebo. Measured lymphocyte subsets were reduced by therapy with compound I. No increase or change in severity of infections were observed in relation to the reduction in lymphocyte counts. These results demonstrate the potential for compound I in the treatment of gout.

[0147] Example 2: Synergistic reductions in serum uric acid in gout patients via treatment with the compound I combined with allopurinol.

[0148] The goal of this study was to determine the dose-response relationship of compound I on serum uric acid (sUA) when administered as monotherapy and in combination with allopurinol. The key efficacy endpoints were the change in sUA levels from baseline on Day 22 and the proportion of subjects at goal sUA < 6.0 mg/dL.

[0149] Methods: 87 adult subjects (M:F = 85:2) with gout and a sUA greater than or equal to 8.0 mg/dL were randomized to a 4X4 factorial study design using placebo, 20 mg, 40 mg, or 80 mg per day of compound I in combination with placebo, 100 mg, 200 mg, or 300 mg daily allopurinol. Drugs were administered in a double-blind manner for three weeks with weekly assessments of sUA, safety parameters, adverse events, and first dose abbreviated

pharmacokinetics (PK). All subjects received colchicine (0.6 mg/d) or naproxen (220-250 mg BID) for gout flare prophylaxis. The per protocol population analyses are presented.

[0150] Results: Compound I produced a significant reduction in sUA levels compared to placebo when administered as monotherapy and as combination therapy with allopurinol. Both compound I and allopurinol demonstrated dose-related reductions in sUA and increases in the proportion of subjects reaching goal. When compound I was combined with allopurinol, there was an additive or synergistic reduction in sUA. Tables 8-10 show reduction of sUA using mlTT population, including patients that received at least one dose of compound I. Figures 2-4 show reduction of sUA using per protocol population where all patients received 3 weeks of treatment.

[0151] Table 8: Change in sUA from Baseline at Day 22. * Compound I

Allopurinol O mg 20 mg 40 mg 80 mg

O mg -0.3 (0.59) -0.7 (0.64) -1.7 (0.57) § -3.2 (0.64) *

100 mg -1.6 (0.63) -2.4 (0.63) § -3.8 (0.58) * -4.5 (0.63) *

200 mg -2.6 (0.63) § -3.8 (0.65) * -4.1 (0.64) * -5.6 (0.64) *

300 mg -2.1 (0.64) § -4.3 (0.63) * -4.3 (0.63) * -4.6 (0.57) *

N per cell, 5-6

* LOCF; values are Least Squares Mean (SD) from ANCOVA with factors for Compound 1 dose, allopurinol dose, compound 1 and allopurinol interaction, and baseline sUA. Overall compound 1 dose -response effect, p <0.001, ANCOVA. Overall allopurinol dose-response effect, p <0.001, ANCOVA. Overall compound 1 and allopurinol dose -response effect, p = 0.775.

p <0.001 vs. placebo.

p <0.001 vs. placebo.

[0152] Table 9: Percent Reduction in sUA at Day 22.

Per protocol population, N = 4-6 per cell. Expressed as Least Square Means (parentheses indicate standard error).

[0153] Table 10: Proportion of Subjects With sUA < 6.0 mg/dL at Day 22. *

Overall compound 1 dose-response effect, p <0.001, ANCOVA. Overall allopurinol dose -response effect, p O.001, ANCOVA.

p <0.05 vs. placebo. [0154] Allopurinol in combination with compound I shows evidence of synergy. An initial analysis was performed to compare the 90% confidence interval (CI) for mean change from baseline in sUA at Day 22 for each drug when given alone versus the 90% CI for the combination treatment. As seen in Tables 11 and 12, there are 5 different treatments

(compound I plus allopurinol) where the 90%> CI for compound I dose when given alone does not overlap with the 90% CI for the combination dose and the 90% CI for the allopurinol dose when given alone does not overlap with the 90% CI for the combination dose. Since the CIs do not overlap, for the following dose combinations: 20 mg compound I plus 300 mg allopurinol, 40 mg compound I plus 100 mg allopurinol, 40 mg compound I plus 300 mg allopurinol, 80 mg compound I plus 100 mg allopurinol, and 80 mg compound I plus 200 mg allopurinol, there is a statistically significant difference between the change from baseline in sUA at Day 22 for the combination therapy and the change from baseline in sUA at Day 22 for each drug when given alone.

[0155] Table 11 : Analysis of Synergy at Day 22

[1] p-value is from a 2 sample t-test comparing the observed mean versus the expected mean.

[0156] Table 12: Change from Baseline in sUA at Day 22

Cmpd I Dose Alone Allopurinol Dose Alone Combination Dose Mean (SD) 90% CI Mean (SD) 90% CI Mean (SD) 90% CI

20 mg Cmpd I +

100 mg -0.94 (1.033) -1.93, 0.05 -1.70 (0.346) -2.03, -1.37 -2.46 (1.174) -3.58, -1.34 Allopurinol

40 mg Cmpd I +

100 mg -1.80 (1.715) -3.21, -0.39 -1.70 (0.346) -2.03, -1.37 -4.62 (1.436) -5.99, -3.25 Allopurinol

80 mg Cmpd I +

100 mg -2.94 (0.321) -3.25, -2.63 -1.70 (0.346) -2.03, -1.37 -4.66 (1.424) -6.02, -3.30 Allopurinol Cmpd I Dose Alone Allopurinol Dose Alone Combination Dose Mean (SD) 90% CI Mean (SD) 90% CI Mean (SD) 90% CI

20 mg Cmpd I +

200 mg -0.94 (1.033) -1.93, 0.05 -2.46 (0.844) -3.27, -1.65 -4.20 (2.217) -6.31, -2.09 Allopurinol

40 mg Cmpd I +

200 mg -1.80 (1.715) -3.21, -0.39 -2.46 (0.844) -3.27, -1.65 -3.86 (0.754) -4.58, -3.14 Allopurinol

80 mg Cmpd I +

200 mg -2.94 (0.321) -3.25, -2.63 -2.46 (0.844) -3.27, -1.65 -5.86 (1.877) -7.65, -4.07 Allopurinol

20 mg Cmpd I +

300 mg -0.94 (1.033) -1.93, 0.05 -1.74 (1.262) -2.94, -0.54 -4.24 (0.991) -5.19, -3.29 Allopurinol

40 mg Cmpd I +

300 mg -1.80 (1.715) -3.21, -0.39 -1.74 (1.262) -2.94, -0.54 -4.28 (0.630) -4.88, -3.68 Allopurinol

80 mg Cmpd I +

300 mg -2.94 (0.321) -3.25, -2.63 -1.74 (1.262) -2.94, -0.54 -4.45 (2.722) -6.69, -2.21 Allopurinol

[0157] Two different analyses were performed to test for synergy. First, a combination index was calculated based on a 40% responder rate, defined as the lowest compound I drug level with 40%) responder rate when given in combination divided by the lowest compound I drug level with 40% responder rate when given alone + the lowest allopurinol drug level with 40% responder rate when given in combination divided by the lowest allopurinol drug level with 40%) responder rate when given alone. For this analysis, a responder is defined as a subject how has a sUA less than 6.0 mg/dL at Day 22. A combination index value of <0.9 was set a priori as determining a synergistic relationship between the drugs. As observed in source Table 13, the combination index was 0.58 indicating synergy based on the a priori criteria.

[0158] Table 13. Combination Index for Response at Day 22

Compound I Dose Level Allopurinol Dose Level

Combination

Alone Combination Alone Combination Index Interpretation

40% Response 300

80 mg 20 mg 100 mg 0.58 Synergistic Rate mg

60% Response

NA 20 mg NA 100 mg NA

Rate

80% Response

NA 20 mg NA 200 mg NA

Rate Combination Index is calculated as compound I level at XX% response when given in

combination/compound I level at XX% response when given alone + allopurinol level at XX% response when given in combination/allopurinol level at XX% response when given alone.

[0159] For the second analysis, the expected mean for each combination was calculated by taking the mean change from baseline for sUA at day 22 for compound I dose when the dose was given with placebo allopurinol and adding it to the mean change from baseline for sUA at day 22 for the allopurinol dose when the dose was given with placebo compound I. The expected mean (the sum of the means from the 2 individual drugs when given alone) was compared to the observed mean (the observed mean change from baseline for sUA at Day 22 when the drugs were given in combination) using a 2 sample t-test. A p-value less than 0.10 indicates a significant difference between the expected mean and the observed mean. As observed in Table 14, the test comparing the expected mean versus the observed mean for the combination of 20 mg compound I plus 300 mg allopurinol results in a p-value of 0.093. Thus, there appears to be a synergistic effect when 20 mg compound I and 300 mg allopurinol is given in combination. In summary, the combination treatment gave a statistically greater reduction in sUA at Day 22 than was expected from the sum of the means of the reduction in sUA at Day 22 for each drug when given alone.

[0160] Table 14. Analysis of Synergy at Day 22

Change from Baseline Percent Change From Baseline Response [1]

Expected Observed Expected Observed

Mean Mean Mean Mean Expected Observed Value Value p-value Value Value p-value Rate Rate Ratio (mg/dL) (mg/dL) \2] (%) (%) \2] (%) (%) \3]

20 mg cmpd I +

100 mg -2.64 -2.46 0.806 -25.13 -23.62 0.796 0.00 0.00 NA

Allopurinol

40 mg cmpd I +

100 mg -3.50 -4.62 0.265 -34.70 -43.58 0.329 16.67 60.00 0.3

Allopurinol

80 mg cmpd I +

100 mg -4.64 -4.66 0.977 -48.90 -43.19 0.251 40.00 40.00 1.0

Allopurinol

20 mg cmpd I +

200 mg -3.40 -4.20 0.503 -34.86 -36.45 0.868 0.00 20.00 0.0

Allopurinol

40 mg cmpd I +

200 mg -4.26 -3.86 0.651 -44.44 -43.98 0.963 16.67 80.00 0.2

Allopurinol Change from Baseline Percent Change From Baseline Response [1]

Expected Observed Expected Observed

Mean Mean Mean Mean Expected Observed

Value Value p-value Value Value p-value Rate Rate Ratio (mg/dL) (mg/dL) [2] (%) (%) [2] (%) (%) [3]

80 mg cmpd I +

200 mg -5.40 -5.86 0.630 -58.64 -54.38 0.595 40.00 80.00 0.5 Allopurinol

20 mg cmpd I +

300 mg -2.68 -4.24 0.093* -29.72 -43.38 0.132 40.00 80.00 0.5

Allopurinol

40 mg cmpd I +

300 mg -3.54 -4.28 0.446 -39.29 -43.66 0.680 50.00 80.00 0.6

Allopurinol

80 mg cmpd I +

300 mg -4.68 -4.45 0.857 -53.49 -45.37 0.562 64.00 83.33 0.8

Allopurinol

Note: An '*' flags treatments that had a Synergistic Response, defined as a p-value <0.100 and observed value less than the expected value.

[1] Response is defined as a sUA measurement at Day 22 that is less than 6 mg/dL.

[2] P-value is based on T-test statistic comparing expected versus observed.

[3] Ratio is derived as Expected Rate / Observed Rate.

[0161] In conclusion, there is statistical evidence of a synergistic effect for compound I when given in combination with allopurinol. As seen in Table 11, there were 5 treatments

(compound I plus allopurinol combinations) that showed a statistical difference between the mean change from baseline in sUA at Day 22 for the combination treatment when compared to each individual drug. All 5 of these treatments showed a greater reduction from baseline in sUA at Day 22 than expected. In addition, one of the 5 treatments showed a statistical difference between the expected reduction from baseline in sUA at Day 22 and the observed reduction from baseline in sUA at Day 22 indicating synergy.

[0162] Compound I showed statistically significant increases in the proportion of subjects achieving sUA < 6.0 mg/dL (p < 0.001), < 5.0 mg/dL (p < 0.003, and < 4.0 mg/dL (p = 0.01) compared to placebo.

[0163] First dose PK for compound I and the active metabolite of allopurinol, oxypurinol, were roughly dose proportional for C max and AUC(o_24h), and did not indicate a drug-drug PK interaction. Compound I treated subjects showed generally mild reductions in lymphocyte subsets during the drug administration. Adverse event frequency and severity were evenly distributed across all dose groups.

[0164] Conclusion: Compound I combined with allopurinol produces additive or synergistic reductions in sUA in gout patients. Combinations of compound I and allopurinol bring a greater proportion of gout patients to goal sUA levels than allopurinol alone. Three weeks of daily dosing with compound I is generally safe and well tolerated when used in combination with allopurinol.

[0165] Example 3: Evaluation of the safety profile of short term use of compound I.

[0166] Methods: Safety data from Example 1 and Example 2 were combined and

summarized by dose of compound I, with subset analysis by dose of compound I for each allopurinol level (placebo, 100 mg, 200 mg, and 300 mg). All dose groups for compound I were combined and analyzed in total and by dose.

[0167] Results: The 2 randomized trials had a combined safety population (received compound I or placebo) of 186, 141 who received compound I (92 alone and 49 with one of three doses of allopurinol) and 45 who received placebo. Colchicine (0.6 mg/d) or naproxen (250 mg twice daily) was used for gout flare prophylaxis in both studies. Demographics were well balanced between compound I and placebo treated subjects. Mean age was 49 (range 23-69); 97% were male, 67%> white, 10%> black, and 8%> Hispanic. The mean (range) body mass index (BMI) was 34.5 (22.2 - 62.9) and mean (range) sUA at enrollment was 9.8 mg/dL (8 - 15 mg/dL). Adverse events (AEs) occurred in 59%> of subjects receiving compound I and 62% of the placebo group. There were no AE differences in frequency or severity in the subjects receiving concomitant allopurinol and those only receiving compound I. Only one serious AE occurred, a hemorrhoidal bleed in a compound I-treated subject not attributed to study drug. The most common AEs reported in subjects receiving compound I were diarrhea (10%)), headache (10%>), peripheral edema (6%>), lymphocyte count decreased (6%>), and URI (5%>). AE frequency and severity were similar in the placebo and compound I groups, except for peripheral edema and lymphocyte count decrease. Two subjects (4%>) receiving placebo and 7 subjects (5%>) receiving compound I (with and without allopurinol) discontinued therapy due to an AE. All reported AEs with compound I-treated subjects were mild or moderate with the exception of one report each of severe diarrhea and severe diverticulitis. Four subjects receiving placebo experienced 7 severe AEs. There were no opportunistic infections reported, nor was there an imbalance in overall infection rate reported between compound I and placebo treated subjects. Effects on lymphocytes were dose related. A total of 2.1%) of subjects experienced a decrease of one grade or more in absolute lymphocyte count. [0168] Conclusions: Compound I was safe and well tolerated when administered at doses up to 240 mg/day for 21 days. The addition of compound I to allopurinol did not alter the safety profile. The frequency of diarrhea may be explained by the high rate of colchicine use (77%).

[0169] Example 4: Covariate analyses of serum uric acid responses to compound I in two studies in subjects with gout.

[0170] Objective: The goal of the analysis was to examine the contribution of treatment with compound I dose and allopurinol dose, and baseline demographic and laboratory factors to the sUA treatment response. The main analytic endpoint was day 22 (d22) change from baseline in sUA levels.

[0171] Methods: Adult subjects with gout, sUA greater than or equal to 8.0 mg/dL off urate lowering therapy, and Cockcroft-Gault creatinine clearance (CrCL) > 60 mL/min, were enrolled in two phase 2 trials. The monotherapy trial is that of Example 1 above. The combination therapy trial is that of Example 2 above. Drug doses were not adjusted for CrCL or weight. Study drugs were administered for 3 weeks, with weekly assessments of sUA. Data from both studies, which had very similar inclusion criteria and identical outcome assessments, were analyzed separately and combined. Univariate and multivariate regression analyses examined the contributions of baseline sUA, CrCL, age, weight, and race to the change from baseline in sUA.

[0172] Results: One-hundred eighty six (186) subjects (M:F = 180:6, mean [range] age 49 [22-69] yr), including 39 with mild renal impairment (CrCL 60-89 mL/min), were enrolled across the two studies. BL sUA levels, CrCL, age, weight and race were well balanced across the treatment groups. In the combined analyses of the two studies, compound I dose, allopurinol dose, BL sUA (all p<0.001), and BL CrCL (p=0.079) were the statistically significant factors influencing d22 change from baseline in sUA in the multiple regression model. CrCL was not a significant factor for sUA change from baseline in the absence of allopurinol use. Weight, age and race were not significant. In each study analyzed separately, compound I dose and BL values of sUA were consistent significant factors (p<0.002 in all cases). In compound I - allopurinol combination study, allopurinol dose and BL CrCL were also significant factors for d22 change from baseline in sUA.

[0173] Conclusions: Doses of compound I and allopurinol were the most important factors in determining magnitude of sUA change from baseline. Baseline CrCL affects the sUA response to allopurinol. The baseline sUA level influences the magnitude of change in sUA after compound I therapy, when used alone or in combination with allopurinol. Mild renal impairment appears to have no influence in the sUA response to compound I.

[0174] Example 5: Add-on therapy for management of gout in patients failing to achieve sUA goal with allopurinol.

[0175] Compound I is an oral, once-daily, novel purine nucleoside phosphorylase inhibitor in clinical development for the chronic management of gout. Synergistic reduction of sUA when combined with xanthine oxidase inhibition was previously shown herein. In a dose-ranging study of compound I given in combination with allopurinol, there was synergistic reduction in sUA.

[0176] This example assesses the efficacy and safety of compound I at doses of 5, 10, 20, or 40 mg/d when administered as add-on therapy to patients failing to reach sUA goals on 300 mg allopurinol daily.

[0177] The study consisted of screening/baseline, treatment, and follow-up periods. Patients who passed screening (Day -42) were titrated to 300 mg allopurinol daily; after 14 days at this dose, patients qualified for randomization if sUA > 6 mg/dL. Exclusion criteria included low lymphocyte count, gout flare (during screening), moderate-to-severe renal dysfunction, WBC count <3.7 x 10 9 /L, and poorly controlled or unstable comorbid conditions. Adults with gout who met baseline inclusion criteria were randomized to receive 5, 10, 20, or 40 mg/d compound I as add-on therapy. Randomization was stratified according to baseline sUA (<10 and >10 mg/dL). Blinded study drugs were administered for 24 weeks, with biweekly assessments of sUA, safety parameters, and adverse events (AEs) for the first month and monthly assessments thereafter. Patients received colchicine 0.6 mg q.d. or naproxen 220 to 250 mg b.i.d. for gout flare prophylaxis for the study duration. Primary efficacy endpoint was proportion of patients with sUA <6 mg/dL at week 12. Safety was assessed using data on AEs and clinical laboratory measures.

[0178] Of 1106 patients screened, 513 failed screening evaluation. Of the remaining 593 patients who initiated the allopurinol run-in phase, 314 failed study criteria at baseline. A total of 278 patients were randomized. Baseline demographics were similar to those of other gout trials, with a high frequency of obesity, hypertension, hypercholesterolemia, and diabetes (Table 15). A total of 50 (18%) of the 278 patients had mild renal dysfunction.

[0179] Table 15. Baseline Patient Demographics and Clinical Characteristics, by CrCl CrCl 60 to <90 mL/min CrCl >90 mL/min All patients*

Parameter (n=50) (n=226) (N=278)

Age, mean (SD), y 59 (7) 47 (10) 49 (10)

Sex, male:female (%) 86: 14 98:2 96:4

Race, n (%)

White 28 (56) 174 (77) 204 (73)

Black 13 (26) 17 (8) 30 (1 1)

Asian 7 (14) 12 (5) 19 (7)

Other 2 (4) 23 (10) 25 (9)

Weight, mean (SD), kg 93 (21) 1 16 (23) 1 12 (24)

BMI, mean (SD), kg/m 2 32 (6) 38 (25) 37 (23)

Obesity, n (%) 13 (26) 1 17 (52) 130 (47)

Morbid obesity, n (%) 4 (8) 65 (29) 69 (25)

Comorbidities, n (%)

Hypertension 42 (84) 1 19 (53) 161 (58)

Diabetes 11 (22) 31 (14) 42 (15)

Hypercholesterolemia 25 (50) 82 (36) 108 (39)

Coronary heart disease 4 (8) 6 (3) 10 (4)

BMI, body mass index; CrCl, creatinine clearance; SD, standard deviation. *Two patients had missing renal data. BMI >40 kg/m 2 .

[0180] Results: Primary endpoint results show that compound I added to allopurinol doubled the proportion of subjects achieving goal (sUA < 6 mg/dL) at week 12 (Figure 5).

[0181] Secondary endpoint results show that compound I added to allopurinol achieved secondary endpoint of sUA < 5 mg/dL (Table 16).

[0182] Table 16. Secondary Endpoint Results

Allopurinol 300mg +

Compound I

Secondary Placebo 5mg lOmg 20mg 40mg Cmpd I All endpoints (N=56) (N=56) (N=56) (N=56) (N=54) (N=222) sUA < 5mg/dL 0 7 * 3 6* 25 n (%) (0%) (13%) (5%) (16%) (11%) (1 1%)

Gout flares n (%) 3 4 3 4 6 17

(5%) (7%) (5%) (7%) (11%) (8%) sUA change from 0.2 -0.3* -0.1 -0.6* -0.8** -0.4* baseline: mean (1.2) (1.1) (1.3) (1.9) (1.8) (1.6)

(SD)

sUA % change 4.2 -3.5* -0.7 -6.9* - 10.3** -5.3 from baseline: (16.5) (17.9) (20.0) (22.2) (22.7) (20.9) mean (SD)

mITT Population, LOCF: last observation carried forward; *p < 0.05 vs. placebo; ** p < 0.001 vs. placebo.

[0183] No fatal or life -threatening adverse events were observed. No signal for infections was observed. Additionally, adherence to study medications, defined as > 85% drug taken, indicated that adherence to blinded study drug was higher than to allopurinol. Total cholesterol and LDL cholesterol were also significantly reduced, while no significant changes in HDL cholesterol or triglycerides were observed (Figure 6).

[0184] This clinical trial is the first to show the potential benefits of compound I as add-on therapy in patients not achieving therapeutic target on allopurinol 300 mg daily.

[0185] Example 6: Drug-drug interaction profile of compound I.

[0186] Compound I was incubated in gender-pooled human liver microsomes with marker substrates for CYP1A2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, or CYP3A4/5, and the catalytic activity of these isoforms was determined. Compound I was incubated with human primary hepatocytes from 3 donors, and induction of CYP1A2, CYP2B6, CYP2C9, CYP3A4/5, MDR1 (P-glycoprotein or P-gp), and MRP2 was assessed.

[0187] Results: CYP isoforms: No significant inhibition of catalytic activity by compound I was observed for CYP1A2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, or

CYP3A4/5 using gender-pooled human liver microsomal incubations. Additionally, compound I was not shown to be a time-dependent inhibitor of CYP3A4/5. Compound I also did not induce protein synthesis or enzyme activity for CYP1A2, CYP2B6, CYP2C9, and CYP3A4 in primary hepatocyte cultures from human donors. Drug Transporters: Compound I did not induce protein synthesis of MDR1 or MRP2 in primary human hepatocytes, nor did the drug significantly inhibit OAT1 (kidney transport)-mediated uptake of p-aminohippuric acid, a substrate for OAT1. Compound I was not a substrate for the OAT1 transporter.

Compound I was demonstrated to weakly inhibit the kidney transporter OAT2 (11%-24% inhibition at 200 μΜ in CHP-OCT2 cells); however, compound I is unlikely to mediate a drug-drug interaction with compounds that are cleared by OCT2. In a phase 2 trial of compound I administered with allopurinol, a first dose PK assessment revealed no drug-drug interactions with allopurinol or its active metabolite oxypurinol.

[0188] Although the invention has been described and illustrated in the foregoing illustrative embodiments, it is understood that the present disclosure has been made only by way of example, and that numerous changes in the details of implementation of the invention can be made without departing from the spirit and scope of the invention, which is limited only by the claims that follow. Features of the disclosed embodiments can be combined and rearranged in various ways to obtain additional embodiments within the scope and spirit of the invention.