Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CRYSTALLINE FORMS OF KINASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2011/156473
Kind Code:
A1
Abstract:
Salts of N-(4- {4-amino-7-[ 1 -(2-hydroxyethyl)- 1 H-pyrazol-4-yl]thieno[3,2-c]pyridin- 3-yl}phenyl)-N'-(3-fluorophenyl)urea and crystalline forms thereof are suitable pharmaceutical ingredients for pharmaceutical compositions useful in the treatment of disease, for example, cancer.

Inventors:
ZHANG GEOFF G Z (US)
BRACKEMEYER PAUL J (US)
SHEIKH AHMAD Y (US)
Application Number:
PCT/US2011/039603
Publication Date:
December 15, 2011
Filing Date:
June 08, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ABBOTT LAB (US)
ZHANG GEOFF G Z (US)
BRACKEMEYER PAUL J (US)
SHEIKH AHMAD Y (US)
International Classes:
C07D495/04; A61K31/4365; A61P35/00
Domestic Patent References:
WO2010065825A22010-06-10
Foreign References:
US63218309A2009-12-07
Other References:
SUTTON ET AL., J. IMMUNOL., vol. 158, 1997, pages 5783 - 5790
TSE ET AL., CANCER RES., vol. 68, 2008, pages 3421 - 3428
Attorney, Agent or Firm:
STEELE, Susan, L. et al. (100 Abbott Park RoadAP6A-1/D037, Abbott Park Illinois, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea malonate in a solid crystalline form.

2. The compound compound N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonate, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at any one or more ofthe following positions: 6.14, 10.70, 19.54, 21.22, 23.14, 24.00° 2Θ, ± 0.2° 2Θ.

3. The compound of claim 2, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at each of said positions.

4. The compound of claim 2, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at each of said positions: 6.14, 9.36, 10.70, 1 1.68, 12.28, 13.30, 16.32, 16.54, 16.97, 18.47, 19.54, 21.22, 21.50, 23.14, 23.46, 23.68, 24.00° 2Θ, ± 0.2° 2Θ.

5. The compound of any of claims 1-4, wherein the crystalline form is a tetrahydrate.

6. A pharmaceutical composition comprising the compound of any of claims 1-5 and one or more pharmaceutically acceptable excipients.

7. A process for preparing N-(4- {4-amino-7-[ 1 -(2-hydroxyethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonate in a solid crystalline form, wherein the crystalline form is Form II of any of claims 1-5, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2-hydroxyethyl)- 1 H- pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 -fluorophenyl)urea bimalonate solid, water, and tetrahydrofuran; b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonate crystalline Form I to exist in the mixture.

8. The process of claim 7, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea malonate in a solid crystalline form, wherein the crystalline form is Form I.

9. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea malonate in a solid crystalline form, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.14, 10.70, 19.54, 21.22, 23.14, 24.00° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea malonate in a solid crystalline form, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at any one or more ofthe following positions: 6.14, 10.70, 19.54, 21.22, 23.14, 24.00° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

10. The method of claim 9, wherein the crystalline N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonate Form I or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

1 1. The method of claim 9, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

12. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-tartrate in a solid crystalline form.

13. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-tartrate in a solid crystalline form, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at any one or more ofthe following positions: 6.21 , 8.88, 10.28, 10.64, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ.

14. The compound of claim 13, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at each of said positions.

15. The compound of claim 13, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at each of said positions: 6.21 , 8.88, 10.28, 10.64, 1 1.96, 12.44, 12.76, 15.93, 18.48, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ.

16. The compound of any of claims 12-15, wherein the crystalline form is a tetrahydrate.

17. A pharmaceutical composition comprising the compound of any of claims 12-16 and one or more pharmaceutically acceptable excipients.

18. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-tartrate in a solid crystalline form, wherein the crystalline form is Form II of any of claims 12-16, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L- bitartrate solid, water, and tetrahydrofuran; b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-tartrate crystalline Form II to exist in the mixture.

19. The process of claim 20, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- tartrate in a solid crystalline form, wherein the crystalline form is Form II.

20. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- tartrate in a solid crystalline form, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.21 , 8.88, 10.28, 10.64, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- tartrate in a solid crystalline form, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 86.21 , 8.88, 10.28, 10.64, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

21. The method of claim 22, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- tartrate Form II or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

22. The method of claim 22, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

23. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HC1 in a solid crystalline form.

24. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HC1 in a solid crystalline form, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.80, 8.00, 9.71 , 13.63, 16.66, 18.69, 19.49, 22.35° 2Θ, ± 0.2° 2Θ.

25. The compound of claim 24, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at each of said positions.

26. The compound of claim 24, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at each of said positions: 5.80, 6.36, 8.00, 9.71 , 10.22, 13.63, 16.66, 18.69, 19.49, 19.77, 21.59, 22.35, 22.76° 2Θ, ± 0.2° 2Θ.

27. The compound of any of claims 23-26, wherein the crystalline form is an anhydrate.

28. A pharmaceutical composition comprising the compound of any of claims 23-27 and one or more pharmaceutically acceptable excipients.

29. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea HC1 in a solid crystalline form, wherein the crystalline form is Form III of any of claims 23-27, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, water, an alcohol, and hydrochloric acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HC1 crystalline Form III to exist in the mixture.

30. The process of claim 29, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HCl in a solid crystalline form, wherein the crystalline form is Form III.

31. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HCl in a solid crystalline form, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.80, 8.00, 9.71 , 13.63, 16.66, 18.69, 19.49, 22.35° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea HCl in a solid crystalline form, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.80, 8.00, 9.71 , 13.63, 16.66, 18.69, 19.49, 22.35° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

32. The method of claim 31 , wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea HCl Form III or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

33. The method of claim 31 , wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

34. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate in a solid crystalline form.

35. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.37, 7.40, 10.69, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ.

36. The compound of claim 35, wherein the crystalline form is Form IV, characterized at least by a powder X-ray diffraction peak at each of said positions.

37. The compound of claim 35, wherein the crystalline form is Form IV, characterized at least by a powder X-ray diffraction peak at each of said positions: 5.37, 7.40, 10.69, 1 1.92, 14.31 , 16.04, 18.02, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ.

38. The compound of any of claims 34-37, wherein the crystalline form is an anhydrate.

39. A pharmaceutical composition comprising the compound of any of claims 34-38 and one or more pharmaceutically acceptable excipients.

40. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV of any of claims 34-38, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, dimethyl formamide, methansulfonic acid, and acetonitrile;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate crystalline Form IV to exist in the mixture.

41. The process of claim 40, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV.

42. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV of any of claims 34-38, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, methansulfonic acid, and ethyl acetate;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate crystalline Form IV to exist in the mixture.

43. The process of claim 42, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV.

44. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.37, 7.40, 10.69, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea HCl in a solid crystalline form, wherein the crystalline form is Form II, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.37, 7.40, 10.69, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

45. The method of claim 44, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea mesylate Form IV or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

46. The method of claim 44, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

47. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-bitartrate in a solid crystalline form.

48. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-bitartrate in a solid crystalline form, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.04, 3.80, 7.62, 16.12, 23.58° 2Θ, ± 0.2° 2Θ.

49. The compound of claim 48, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at each of said positions.

50. The compound of claim 48, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at each of said positions: 2.70. 3.04, 3.80, 5.64. 6.58. 7.62, 16.12, 16.55, 18.68, 19.10, 19.83, 21.81 , 23.00, 23.58° 2Θ, ± 0.2° 2Θ.

51. The compound of any of claims 47-50, wherein the crystalline form is an anhydrate.

52. A pharmaceutical composition comprising the compound of any of claims 47-51 and one or more pharmaceutically acceptable excipients.

53. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-bitartrate in a solid crystalline form, wherein the crystalline form is Form V of any of claims 47-50, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and L-tartaric acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L-bitartrate crystalline Form V to exist in the mixture.

54. The process of claim 53, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- bitartrate in a solid crystalline form, wherein the crystalline form is Form V.

55. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- bitartrate in a solid crystalline form, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.04, 3.80, 7.62, 16.12, 23.58° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3- fluorophenyl)urea L-bitartrate in a solid crystalline form, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.04, 3.80, 7.62, 16.12, 23.58° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

56. The method of claim 55, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea L- bitartrate Form V or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

57. The method of claim 55, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

58. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea bimalonate in a solid crystalline form.

59. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.10, 8.72, 15.76° 2Θ, ± 0.2° 2Θ.

60. The compound of claim 59, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at each of said positions.

61. The compound of claim 59, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at each of said positions: 6.10, 8.72, 10.67, 1 1.25, 13.08, 15.27, 15.76, 17.46, 18.27, 18.77, 19.42° 2Θ, ± 0.2° 2Θ.

62. The compound of any of claims 58-61 , wherein the crystalline form is an anhydrate.

63. A pharmaceutical composition comprising the compound of any of claims 58-62 and one or more pharmaceutically acceptable excipients.

64. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI of any of claims 58-62, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and malonic acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea bimalonate crystalline Form VI to exist in the mixture.

65. The process of claim 64, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI.

66. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.10, 8.72, 15.76° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4-{4- amino-7- [ 1 -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 - fluorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.10, 8.72, 15.76° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

67. The method of claim 66, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea bimalonate Form VI or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

68. The method of claim 66, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

69. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate in a solid crystalline form.

70. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.20, 16.01 , 17.44, 18.83° 2Θ, ± 0.2° 2Θ.

71. The compound of claim 70, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at each of said positions.

72. The compound of claim 70, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at each of said positions: 4.62, 5.20, 6.99, 9.09, 9.66, 10.40, 13.96, 16.01 , 17.44, 18.83, 20.93, 22.52, 22.80, 24.13° 2Θ, ± 0.2° 2Θ.

73. The compound of any of claims 69-72, wherein the crystalline form is a hydrate.

74. A pharmaceutical composition comprising the compound of any of claims 69-73 and one or more pharmaceutically acceptable excipients.

75. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII of any of claims 69-73, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and ascorbic acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate crystalline Form VII to exist in the mixture.

76. The process of claim 75, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII.

77. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.20, 16.01 , 17.44, 18.83° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3- fluorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.20, 16.01 , 17.44, 18.83° 2Θ, ± 0.2° 2Θ and one or more

pharmaceutically acceptable excipients.

78. The method of claim 77, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea ascorbate Form VII or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

79. The method of claim 77, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

80. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea maleate in a solid crystalline form.

81. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.00, 15.23, 19.79, 23.63° 2Θ, ± 0.2° 2Θ.

82. The compound of claim 81 , wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at each of said positions.

83. The compound of claim 81 , wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at each of said positions: 5.00, 8.10, 9.30, 10.00, 1 1.1 1 , 12.22, 13.67, 15.23, 16.85, 18.37, 19.79, 23.63° 2Θ, ± 0.2° 2Θ.

84. The compound of any of claims 80-83, wherein the crystalline form is a hydrate.

85. A pharmaceutical composition comprising the compound of any of claims 80-84 and one or more pharmaceutically acceptable excipients.

86. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII of any of claims 80-84, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, dimethyl formamide, and maleic acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea maleate crystalline Form VIII to exist in the mixture.

87. The process of claim 86, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII.

88. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.00, 15.23, 19.79, 23.63° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4-{4- amino-7- [ 1 -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 - fluorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.00, 15.23, 19.79, 23.63° 2Θ, ± 0.2° 2Θ and one or more

pharmaceutically acceptable excipients.

89. The method of claim 88, wherein the crystalline N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea maleate Form VIII or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

90. The method of claim 88, wherein the cancer is myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

91. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate in a solid crystalline form.

92. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.85, 7.67, 1 1.23, 16.87° 2Θ, ± 0.2° 2Θ.

93. The compound of claim 92, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at each of said positions.

94. The compound of claim 92, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at each of said positions: 3.85, 5.62, 6.70, 6.91 , 7.67, 1 1.23, 1 1.53, 12.99, 16.87, 17.42° 2Θ, ± 0.2° 2Θ.

95. The compound of any of claims 91-94 wherein the crystalline form is a hydrate.

96. A pharmaceutical composition comprising the compound of any of claims 91-95 and one or more pharmaceutically acceptable excipients.

97. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX of any of claims 91-94, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, propanol, and sulfuric acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate crystalline Form IX to exist in the mixture.

98. The process of claim 97, further comprising isolating N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX.

99. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.85, 7.67, 1 1.23, 16.87° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4-{4- amino-7- [ 1 -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 - fluorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.85, 7.67, 1 1.23, 16.87° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

100. The method of claim 99, wherein the crystalline N-(4- {4-amino-7-[ 1 -(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea sulfate Form IX or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

101. The method of claim 99, wherein the cancer is myelodysplasia syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

102. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea phosphate in a solid crystalline form.

103. The compound N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.85, 6.57, 6.91 , 20.85° 2Θ, ± 0.2° 2Θ.

104. The compound of claim 103, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at each of said positions.

105. The compound of claim 103, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at each of said positions: 5.85, 6.57, 6.91 , 13.83, 17.65, 18.74, 19.12, 20.85° 2Θ, ± 0.2° 2Θ.

106. The compound of any of claims 102-105 wherein the crystalline form is a hydrate.

107. A pharmaceutical composition comprising the compound of any of claims 102- 106 and one or more pharmaceutically acceptable excipients.

108. A process for preparing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form IX of any of claims 102-105, comprising: a) providing a mixture comprising (i) N-(4- {4-amino-7- [ 1 -(2 -hydroxy ethyl)- 1 H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, propanol, and phosphoric acid;

b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea phosphate crystalline Form X to exist in the mixture.

109. The process of claim 108, further comprising isolating N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form X.

1 10. A method for treating cancer in a mammal comprising administering to a subject having the disease a therapeutically effective amount of (a) N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.85, 6.57, 6.91 , 20.85° 2Θ, ± 0.2° 2Θ or (b) a pharmaceutical composition comprising N-(4-{4- amino-7- [ 1 -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 - fluorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.85, 6.57, 6.91 , 20.85° 2Θ, ± 0.2° 2Θ and one or more pharmaceutically acceptable excipients.

1 1 1. The method of claim 1 10, wherein the crystalline N-(4- {4-amino-7-[ 1 -(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea phosphate Form X or pharmaceutical composition is administered by an oral, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial, or intra-articular route.

112. The method of claim 110, wherein the cancer is myelodysplasia syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

Description:
CRYSTALLINE FORMS OF KINASE INHIBITORS

CROSS-REFERENCE TO RELATED APPLICATIONS This applications claims priority to United States Provisional Application Serial No. 61/353,170 filed June 09, 2010, which is incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention relates to crystalline forms of salts of N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea, processes for preparing the crystalline forms, pharmaceutical formulations thereof, and methods of treating cancer.

BACKGROUND OF THE INVENTION

Mitosis is a process by which a complete copy of a duplicated genome is segregated by the microtuble spindle apparatus into two daughter cells. Aurora-kinases, key mitotic regulators required for genome stability, have been found to be overexpressed in human tumors. There is therefore an existing need in the therapeutic arts for compounds which inhibit Aurora-kinases, compositions comprising the inhibitors and methods of treating diseases during which Aurora-kinases are unregulated or overexpressed.

The reversible phosphorylation of proteins is one of the primary biochemical mechanisms mediating eukaryotic cell signaling. This reaction is catalyzed by protein kinases that transfer the g-phosphate group of ATP to hydroxyl groups on target proteins. 518 such enzymes exist in the human genome of which ~90 selectively catalyze the phosphorylation of tyrosine hydroxyl groups. Cytosolic tyrosine kinases reside intracellularly whereas receptor tyrosine kinases (RTKs) possess both extracellular and intracellular domains and function as membrane spanning cell surface receptors. As such, RTKs mediate the cellular responses to environmental signals and facilitate a broad range of cellular processes including

proliferation, migration and survival.

RTK signaling pathways are normally highly regulated, yet their over-activation has been shown to promote the growth, survival and metastasis of cancer cells. Dysregulated RTK signaling occurs through gene over-expression or mutation and has been correlated with the progression of various human cancers. The VEGF receptor (VEGFR) family consists of three RTKs, KDR (kinase insert domain-containing receptor; VEGFR2), FLTl (Fms-like tyrosine kinase; VEGFR1), and FLT4 (VEGFR3). These receptors mediate the biological function of the vascular endothelial growth factors (VEGF-A, -B, -C, -D, -E and placenta growth factor (P1GF)), a family of homodimeric glycoproteins that bind the VEGF receptors with varying affinities.

KDR is the major mediator of the mitogenic, angiogenic and permeability-enhancing effects of VEGF-A, hereafter referred to as VEGF. Many different cell types are able to produce VEGF, yet its biological activity is limited predominately to the vasculature by way of the endothelial cell-selective expression of KDR. Not surprisingly, the VEGF/KDR axis is a primary mediator of angiogenesis, the means by which new blood vessels are formed from preexisting vessels.

FLTl binds VEGF, VEGF-B and placental growth factor. FLTl is expressed on the surface of smooth muscle cells, monocytes and hematopoietic stems cells in addition to endothelial cells. Activation of FLTl signaling results in the mobilization of marrow-derived endothelial progenitor cells that are recruited to tumors where they contribute to new blood vessel formation.

FLT4 mediates the signaling of VEGF-C and VEGF-D, which mediate formation of tumor-associated lymphatic vessels (lymphangiogenesis). Lymphatic vessels are one of the routes by which cancer cells disseminate from solid tumors during metastasis.

The PDGF receptor (PDGFR) family consists of five RTK's, PDGFR-a and -b, CSF1R, KIT, and FLT3.

CSF-1R is encoded by the cellular homo log of the retroviral oncogene v-fms and is a major regulator of macrophage development. Macrophages are frequent components of tumor stroma and have been shown to modify the extracellular matrix in a manner beneficial to tumor growth and metastasis.

KIT is expressed by hematopoietic progenitor cells, mast cells, germ cells and by pacemaker cells in the gut (interstitial cells of Cajal). It contributes to tumor progression by two general mechanisms namely autocrine stimulation by its ligand, stem cell factor (SCF), and through mutations that result in ligand-independent kinase activity.

FLT3 is normally expressed on hematopoietic stem cells where its interaction with FLT3 ligand (FL) stimulates stem cell survival, proliferation and differentiation. In addition to being over-expressed in various leukemia cells, FLT3 is frequently mutated in hematological malignancies with approximately one-third of patients with acute myeloid leukemia (AML) harboring activating mutations.

The identification of effective small compounds which specifically inhibit signal transduction and cellular proliferation by modulating the activity of tyrosine kinases to regulate and modulate abnormal or inappropriate cell proliferation, differentiation, or metabolism is therefore desirable. In particular, the identification of methods and compounds that specifically inhibit the function of a tyrosine kinase which is essential for angiogenic processes or the formation of vascular hyperpermeability leading to edema, ascites, effusions, exudates, and macromolecular extravasation and matrix deposition as well as associated disorders would be beneficial.

Compounds that inhibit protein kinases such as Aurora-kinases and the VEGFR and PDGFR families of kinases have been identified, including N-(4-{4-amino-7-[l-(2- hydroxyethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea. These compounds, and methods to make them, are disclosed in U.S. Patent Application No. 12/632183 (hereinafter "the Ί83 application"), incorporated by reference herein in its entirety.

It now has been found that salts of N-(4-{4-amino-7-[ 1 -(2 -hydroxy ethyl)- lH-pyrazol- 4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)ur ea can be converted into crystalline forms, which can advantageously be used as an active pharmaceutical ingredient in cancer therapy. To that purpose, these crystalline forms are converted into pharmaceutical formulations.

A crystalline form is the form in which the position of the molecules relative to one another is organized according to a three-dimensional lattice structure. Polymorphs are different crystalline forms of the same compound resulting from a different arrangement of the molecules in the solid state. Polymorphs differ from each other in their physical properties but not their chemical composition.

Polymorphism is of particular interest in the development of suitable pharmaceutical dosage forms. Certain polymorphic forms may exhibit superior stability and storability, resulting in enhanced shelf-life of the pharmaceutical product. In addition, certain polymorphic forms are more readily manufactured in high purity in large quantities. Critically, polymorphs of an active pharmaceutical ingredient can have different aqueous solubility and dissolution rates, which may have therapeutic consequences due to the potential differences in bioavailability between polymorphs of the same compound.

The present invention provides crystalline forms of salts of N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea having beneficial properties in one or more of the following characteristics: the ability to be formulated in a pharmaceutical dosage form, adequate shelf-life in a pharmaceutical dosage form, and/or ability to be effectively administered in a pharmaceutical dosage form.

SUMMARY OF THE INVENTION

In one embodiment, the invention provides N-(4- {4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea malonate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea L-tartrate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea HC1 in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea mesylate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea bitartrate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea bimalonate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea ascorbate in solid crystalline form. In one embodiment, the invention provides N-(4- {4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea maleate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea sulfate in solid crystalline form.

In one embodiment, the invention provides N-(4-{4-amino-7-[l -(2-hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea phosphate in solid crystalline form.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea malonate characterized herein and designated Form I. In a further embodiment, the invention provides hydrated forms of Form I, including the tetrahydrate form.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea L-tartrate characterized herein and designated Form II. In a further embodiment, the invention provides hydrated forms of Form II, including the tetrahydrate form.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea HC1 characterized herein and designated Form III. In a further

embodiment, the invention provides an anhydrate form of Form III.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea mesylate characterized herein and designated Form IV. In a further embodiment, the invention provides an anhydrate form of Form IV.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea bitartrate characterized herein and designated Form V. In a further embodiment, the invention provides an anhydrate form of Form V. In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea bimalonate characterized herein and designated Form VI. In a further embodiment, the invention provides an anhydrate form of Form VI.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea ascorbate characterized herein and designated Form VII. In a further embodiment, the invention provides an anhydrate form of Form VII.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea malonate characterized herein and designated Form VIII. In a further embodiment, the invention provides a hydrate form of Form VIII.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea sulfate characterized herein and designated Form IX. In a further embodiment, the invention provides a hydrate form of Form IX.

In a further embodiment, the invention provides a crystal polymorph of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea phosphate characterized herein and designated Form X. In a further embodiment, the invention provides a hydrate form of Form X.

There is further provided a pharmaceutical composition comprising Form I, Form II, Form III, Form IV, Form V, Form VI, Form VII, Form VIII, Form IX, or Form X and one or more pharmaceutically acceptable excipients.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea malonate in a solid crystalline form, wherein the crystalline form is Form I, comprising: a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea bimalonate solid, water, and tetrahydrofuran; and b) causing N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonate crystalline Form I to exist in the mixture. There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fluorophenyl)urea L- tartrate in a solid crystalline form, wherein the crystalline form is Form II comprising:

a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-bitartrate solid, water, and tetrahydrofuran; b) causing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-tartrate crystalline Form II to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea HC1 in a solid crystalline form, wherein the crystalline form is Form III comprising:

a)providing a mixture comprising (i) N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, water, an alcohol, and hydrochloric acid; and b) causing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea HC1 crystalline Form III to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV of any of claims 34-38, comprising: a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea free base solid, dimethyl formamide, methansulfonic acid, and acetonitrile; b) causing N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c] pyridin-3-yl}phenyl)-N'-(3- fluorophenyl)urea mesylate crystalline Form IV to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fluorophenyl)urea L- bitartrate in a solid crystalline form, wherein the crystalline form is Form V, comprising: a)providing a mixture comprising (i) N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and L-tartaric acid; b)causing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-bitartrate crystalline Form V to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI, comprising a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and malonic acid; b) causing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea bimalonate crystalline Form VI to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII, comprising: a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, tetrahydrofuran, water, and ascorbic acid; b) causing N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea ascorbate crystalline Form VII to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII, comprising: a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, dimethyl formamide, and maleic acid; b) causing N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea maleate crystalline Form VIII to exist in the mixture.

There is further provided a process for preparing N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX, comprising: a) providing a mixture comprising (i) N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, propanol, and sulfuric acid; b) causing N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea sulfate crystalline Form IX to exist in the mixture.

There is further provided a process for preparing N-(4- {4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea phosphate in a solid crystalline form, wherein the crystalline form is Form X, comprising: a)providing a mixture comprising (i) N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid, propanol, and phosphoric acid; b) causing N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea phosphate crystalline Form X to exist in the mixture.

In a further embodiment, the invention provides a method for treating cancer in a mammal comprising administering to a subject having the disease therapeutically effective amount of salts of N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2- c]pyridin-3-yl}phenyl)-N'-(3-fiuorophenyl)urea in a solid crystalline form, wherein the crystalline form is Form I, II, III, IV, V, VI, VII, VIII, IX, or X or (b) a pharmaceutical composition comprising salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea in a solid crystalline form, wherein the crystalline form is Form I, II, III, IV, V, VI, VII, VIII, IX, or X, one or more pharmaceutically acceptable excipients. Examples of such cancers include myelodysplastic syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

Additional embodiments of the invention, including more particular aspects of those provided above, will be found in, or will be evident from, the detailed description that follows.

BRIEF DESCRIPTION OF THE DRAWING

Fig. 1 is a PXRD scan of crystal polymorph Form I N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea dihydrogen malonate. Fig. 2 is a PXRD scan of crystal polymorph Form II N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea hydrogen L-tartrate.

Fig. 3 is a PXRD scan of crystal polymorph Form III N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea HC1.

Fig. 4 is a PXRD scan of crystal polymorph Form IV N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea mesylate.

Fig. 5 is a PXRD scan of crystal polymorph Form V N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea L- bitartrate.

Fig. 6 is a PXRD scan of crystal polymorph Form VI N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea bimalonate.

Fig. 7 is a PXRD scan of crystal polymorph Form VII N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea ascorbate.

Fig. 8 is a PXRD scan of crystal polymorph Form VIII N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea maleate.

Fig. 9 is a PXRD scan of crystal polymorph Form IX N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea sulfate.

Fig. 10 is a PXRD scan of crystal polymorph Form X N-(4- {4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea phosphate.

DETAILED DESCRIPTION

The invention encompasses crystalline forms of salts of N-(4-{4-amino-7-[l-(2- hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phen yl)-N'-(3-fiuorophenyl)urea. N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c] pyridin-3-yl}phenyl)-N'- (3-fluorophenyl)urea free base is prepared, illustratively, as described in Example 1 of above- cited U.S. Patent Application No. 12/632183, the entire disclosure of which is incorporated by reference herein. The term "free base" is used for convenience herein to refer to N-(4-{4- amino-7- [ 1 -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 - fluorophenyl)urea parent compound as distinct from any salt thereof.

Those skilled in the art will also understand that the term "monohydrate" when referring to N-(4- {4-amino-7-[l -(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea means that there is one water molecule for every molecule of N-(4- {4-amino-7-[ 1 -(2-hydroxy ethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea. Those skilled in the art will also understand that the term "tetrahydrate" when referring to N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea means that there is four water molecule for every molecule of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yljthieno [3 ,2-c]pyridin-3 -yl} phenyl)-N"-(3 -fluorophenyl)urea.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea malonate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea malonate in a solid crystalline form, wherein the crystalline form is Form I, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.14, 10.70, 19.54, 21.22, 23.14, 24.00° 2Θ, ± 0.2° 2Θ, herein defined as Form I. Alternately, the invention encompasses Form I characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form I characterized at least by a powder X-ray diffraction peak at each of said positions: 6.14, 9.36, 10.70, 1 1.68, 12.28, 13.30, 16.32, 16.54, 16.97, 18.47, 19.54, 21.22, 21.50, 23.14, 23.46, 23.68, 24.00° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea L-tartrate in a solid crystalline form. In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea L-tartrate in a solid crystalline form, wherein the crystalline form is Form II,

characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.21 , 8.88, 10.28, 10.64, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ, herein defined as Form II. Alternately, the invention

encompasses Form II characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form II characterized at least by a powder X-ray diffraction peak at each of said positions: 6.21 , 8.88, 10.28, 10.64, 1 1.96, 12.44, 12.76, 15.93, 18.48, 19.18, 20.58, 21.16, 21.38, 21.75, 22.50, 22.86, 23.86, 24.50, 24.92° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea HCl in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea HCl in a solid crystalline form, wherein the crystalline form is Form III, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.80, 6.36, 8.00, 9.71 , 10.22, 13.63, 16.66, 18.69, 19.49, 19.77, 21.59, 22.35, 22.76° 2Θ, ± 0.2° 2Θ, herein defined as Form IV. Alternately, the invention encompasses Form IV characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another

embodiment, the invention encompasses Form IV characterized at least by a powder X-ray diffraction peak at each of said positions: 5.37, 7.40, 10.69, 1 1.92, 14.31 , 16.04, 18.02, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea mesylate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea mesylate in a solid crystalline form, wherein the crystalline form is Form IV,

characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.37, 7.40, 10.69, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ, herein defined as Form IV. Alternately, the invention encompasses Form IV characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form IV characterized at least by a powder X-ray diffraction peak at each of said positions: 5.37, 7.40, 10.69, 1 1.92, 14.31 , 16.04, 18.02, 18.39, 18.78, 20.42, 21.20° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea L-bitartrate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fiuorophenyl) urea L-bitartrate in a solid crystalline form, wherein the crystalline form is Form V, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.04, 3.80, 7.62, 16.12, 23.58° 2Θ, ± 0.2° 2Θ, herein defined as Form V.

Alternately, the invention encompasses Form V characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form V characterized at least by a powder X-ray diffraction peak at each of said positions: 2.70. 3.04, 3.80, 5.64. 6.58. 7.62, 16.12, 16.55, 18.68, 19.10, 19.83, 21.81 , 23.00, 23.58° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea bimalonate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fiuorophenyl) urea bimalonate in a solid crystalline form, wherein the crystalline form is Form VI, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 6.10, 8.72, 15.76° 2Θ, ± 0.2° 2Θ, herein defined as Form VI. Alternately, the invention encompasses Form VI characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form VI characterized at least by a powder X-ray diffraction peak at each of said positions: 6.10, 8.72, 10.67, 1 1.25, 13.08, 15.27, 15.76, 17.46, 18.27, 18.77, 19.42° 2Θ, ± 0.2° 2Θ. In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea ascorbate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea ascorbate in a solid crystalline form, wherein the crystalline form is Form VII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.20, 16.01 , 17.44, 18.83° 2Θ, ± 0.2° 2Θ, herein defined as Form VII. Alternately, the invention encompasses Form VII characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form

VII characterized at least by a powder X-ray diffraction peak at each of said positions: 4.62, 5.20, 6.99, 9.09, 9.66, 10.40, 13.96, 16.01 , 17.44, 18.83, 20.93, 22.52, 22.80, 24.13° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea maleate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea maleate in a solid crystalline form, wherein the crystalline form is Form VIII, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.00, 15.23, 19.79, 23.63° 2Θ, ± 0.2° 2Θ, herein defined as Form VIII. Alternately, the invention encompasses Form VIII characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form

VIII characterized at least by a powder X-ray diffraction peak at each of said positions: 5.00, 8.10, 9.30, 10.00, 1 1.1 1 , 12.22, 13.67, 15.23, 16.85, 18.37, 19.79, 23.63° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea sulfate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea sulfate in a solid crystalline form, wherein the crystalline form is Form IX, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 3.85, 7.67, 1 1.23, 16.87° 2Θ, ± 0.2° 2Θ, herein defined as Form IX. Alternately, the invention encompasses Form IX characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form IX characterized at least by a powder X-ray diffraction peak at each of said positions: 3.85, 5.62, 6.70, 6.91 , 7.67, 1 1.23, 1 1.53, 12.99, 16.87, 17.42° 2Θ, ± 0.2° 2Θ.

In one embodiment, the invention encompasses N-(4- {4-amino-7-[l-(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea phosphate in a solid crystalline form.

In one embodiment the invention encompasses a crystalline form of N-(4-{4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3-fluorophenyl) urea phosphate in a solid crystalline form, wherein the crystalline form is Form X, characterized at least by a powder X-ray diffraction peak at any one or more of the following positions: 5.85, 6.57, 6.91 , 20.85° 2Θ, ± 0.2° 2Θ, herein defined as Form X. Alternately, the invention encompasses Form X characterized at least by a powder X-ray diffraction peak at each of said positions. In yet another embodiment, the invention encompasses Form X characterized at least by a powder X-ray diffraction peak at each of said positions: 5.85, 6.57, 6.91 , 13.83, 17.65, 18.74, 19.12, 20.85° 2Θ, ± 0.2° 2Θ.

The crystalline form of salts of N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea , for example Form I, II, III, IV, V, VI, VII, VIII, IX, or X, can be useful as API for the preparation of pharmaceutical compositions suitable for any route of administration, including oral, to a subject in need thereof. Other routes of administration include, without limitation, parenteral, sublingual, buccal, intranasal, pulmonary, topical, transdermal, intradermal, ocular, otic, rectal, vaginal, intragastric, intracranial, intrasynovial and intra-articular routes.

Where it is desired to provide salts of N-(4-{4-amino-7-[ 1 -(2 -hydroxy ethyl)- 1H- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea, for example Form I, II, III, IV, V, VI, VII, VIII, IX, or X, in solution form, for example in a liquid formulation for oral or parenteral administration, the citrate salts of N-(4- {4-amino-7-[l -(2-hydroxyethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea will not, of course, be present in such a formulation in crystalline form; indeed, the presence of crystals is generally undesired in such a formulation. However, crystalline salts o fN-(4-{4-amino-7-[l- (2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea of the present invention can nonetheless be important as API in a process for preparing such a formulation.

Even where the desired formulation is one containing salts of N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea in amorphous form, for example a solid dispersion formulation, crystalline salts of N-(4-{4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea can still be useful as API in a process for preparing such a formulation.

As API, a crystalline form of salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH- pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorop henyl)urea, such as Form I, II, III, IV, V, VI, VII, VIII, IX, or X, has advantages over the amorphous form. For example, purification of API to the high degree of purity required by most regulatory authorities is more efficient and therefore costs less where the API is crystalline as opposed to amorphous form. Physical and chemical stability, and therefore shelf-life of the API solid, is also typically better for crystalline than amorphous forms. Ease of handling is improved over the amorphous form, which tends to be oily or sticky. Drying ismore straightforward and more easily controlled in the case of the crystalline material, which has a well-defined drying or desolvation temperature, than in the case of the amorphous material, which has greater affinity for organic solvents and no well-defined drying temperature. Downstream processing using crystalline API permits enhanced process control. These advantages are illustrative a non-limiting.

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea is present in a pharmaceutical composition of the invention in an amount that can be therapeutically effective when the composition is administered to a subject in need thereof according to an appropriate regimen. Typically, a unit dose (the amount administered at a single time), which can be administered at an appropriate frequency, e.g., twice daily to once weekly, is about 10 to about 1,000 mg, depending on the compound in question. Where frequency of administration is once daily (q.d.), unit dose and daily dose are the same. Illustratively, the unit dose is typically about 25 to about 1,000 mg, more typically about 50 to about 500 mg, for example about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 450 or about 500 mg.

Excipients include but are not limited to, for example, encapsulating materials and additives such as absorption accelerators, antioxidants, binders, buffers, carriers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, glidants, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents, mixtures thereof and the like.

Excipients for preparation of formulations comprising or made with crystalline salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3-yl}phenyl)-N'- (3-fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X to be administered orally in solid dosage form include, for example, agar, alginic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, 1,3-butylene glycol, carbomers, castor oil, cellulose, cellulose acetate, cocoa butter, copovidone, corn starch, corn oil, cottonseed oil, cross-povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, groundnut oil, hydroxypropylmethyl cellulose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mannitol, monoglycerides, olive oil, povidone, peanut oil, potassium phosphate salts, potato starch, povidone, propylene glycol, Ringer's solution, safflower oil, sesame oil, silicon dioxide, sodium carboxymethyl cellulose, sodium phosphate salts, sodium lauryl sulfate, sodium sorbitol, sodium stearylfumarate, soybean oil, stearic acids, stearyl fumarate, sucrose, surfactants, talc, tragacanth, tetrahydrofurfuryl alcohol, triglycerides, vitamin E and derivatives thereof, water, mixtures thereof and the like.

Excipients for preparation of compositions comprising or made with crystalline salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3-yl}phenyl)- N * -(3-fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X to be administered ophthalmically or orally in liquid dosage forms include, for example, 1,3-butylene glycol, castor oil, corn oil, cottonseed oil, ethanol, fatty acid esters of sorbitan, germ oil, groundnut oil, glycerol, isopropanol, olive oil, polyethylene glycols, propylene glycol, sesame oil, water, mixtures thereof and the like. Excipients for preparation of compositions comprising or made with crystalline salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3-yl}phenyl)- N * -(3-fiuorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X to be administered osmotically include, for example, chlorofluorohydrocarbons, ethanol, water, mixtures thereof and the like.

Excipients for preparation of compositions comprising or made with crystalline salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3-yl}phenyl)- N * -(3-fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X to be administered parenterally include, for example, 1,3-butanediol, castor oil, corn oil, cottonseed oil, dextrose, germ oil, groundnut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, U.S. P. or isotonic sodium chloride solution, water, mixtures thereof and the like.

Excipients for preparation of compositions comprising or made with crystalline salts of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3-yl}phenyl)- N * -(3-fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X to be administered rectally or vaginally include, but are not limited to, cocoa butter, polyethylene glycol, wax, mixtures thereof and the like.

The composition is normally administered in an amount providing a therapeutically effective daily dose of the drug. The term "daily dose" herein means the amount of drug administered per day, regardless of the frequency of administration. For example, if the subject receives a unit dose of 150 mg twice daily, the daily dose is 300 mg. Use of the term "daily dose" will be understood not to imply that the specified dosage amount is necessarily administered once daily. However, in a particular embodiment the dosing frequency is once daily (q.d.), and the daily dose and unit dose are in this embodiment the same thing.

What constitutes a therapeutically effective dose depends on the particular compound, the subject (including species and body weight of the subject), the disease (e.g., the particular type of cancer) to be treated, the stage and/or severity of the disease, the individual subject's tolerance of the compound, whether the compound is administered in monotherapy or in combination with one or more other drugs, e.g. , other chemotherapeutics for treatment of cancer, and other factors. Thus the daily dose can vary within wide margins, for example from about 10 to about 1,000 mg. Greater or lesser daily doses can be appropriate in specific situations. It will be understood that recitation herein of a "therapeutically effective" dose herein does not necessarily require that the drug be therapeutically effective if only a single such dose is administered; typically therapeutic efficacy depends on the composition being administered repeatedly according to a regimen involving appropriate frequency and duration of administration. It is strongly preferred that, while the daily dose selected is sufficient to provide benefit in terms of treating the cancer, it should not be sufficient to provoke an adverse side-effect to an unacceptable or intolerable degree. A suitable therapeutically effective dose can be selected by the physician of ordinary skill without undue

experimentation based on the disclosure herein and on art cited herein, taking into account factors such as those mentioned above. The physician may, for example, start a cancer patient on a course of therapy with a relatively low daily dose and titrate the dose upwards over a period of days or weeks, to reduce risk of adverse side-effects.

Illustratively, suitable doses ofN-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea are generally about 10 to about 1 ,000 mg/day, more typically about 50 to about 500 mg/day or about 200 to about 400 mg/day, for example about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 450 or about 500 mg/day, administered at an average dosage interval of 3 to 10 days, or about 4 to 8 days, or about 7 days.

A composition comprising crystalline salts of N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API) of the invention are suitable for use in monotherapy or in combination therapy, for example with other chemotherapeutics or with ionizing radiation.

A composition comprising crystalline salts of N-(4- {4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API), can be administered in combination therapy with one or more therapeutic agents that include, but are not limited to, alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotics, antiproliferatives, antivirals, aurora kinase inhibitors, other apoptosis promoters (for example, Bcl-xL, Bcl-w and Bfl-1 inhibitors), activators of a death receptor pathway, Bcr-Abl kinase inhibitors, BiTE (bi-specific T-cell engager) antibodies, antibody-drug conjugates, biological response modifiers, cyclin-dependent kinase (CDK) inhibitors, cell cycle inhibitors, cyclooxygenase-2 (COX-2) inhibitors, dual variable domain binding proteins (DVDs), human epidermal growth factor receptor 2 (ErbB2 or HER/2neu) receptor inhibitors, growth factor inhibitors, heat shock protein (HSP)-90 inhibitors, histone deacetylase (HDAC) inhibitors, hormonal therapies, immunologicals, inhibitors of apoptosis proteins (IAPs), intercalating antibiotics, kinase inhibitors, kinesin inhibitors, JAK2 inhibitors, mammalian target of rapamycin (mTOR) inhibitors, microR As, mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors, multivalent binding proteins, non-steroidal anti-inflammatory drugs (NSAIDs), poly-ADP (adenosine diphosphate)-ribose polymerase (PARP) inhibitors, platinum chemotherapeutics, polo-like kinase (Plk) inhibitors, phosphoinositide-3 kinase (PI3K) inhibitors, proteasome inhibitors, purine analogs, pyrimidine analogs, receptor tyrosine kinase inhibitors, retinoids, deltoids, plant alkaloids, small inhibitory ribonucleic acids (siRNAs), topoisomerase inhibitors, ubiquitin ligase inhibitors, and the like.

BiTE antibodies are bi-specific antibodies that direct T-cells to attack cancer cells by simultaneously binding the two cells. The T-cell then attacks the target cancer cell.

Examples of BiTE antibodies include, but are not limited to, adecatumumab (Micromet MT201), blinatumomab (Micromet MT103) and the like. Without being limited by theory, one of the mechanisms by which T-cells elicit apoptosis of the target cancer cell is by exocytosis of cytolytic granule components, which include perforin and granzyme B. In this regard, Bcl-2 has been shown to attenuate the induction of apoptosis by both perforin and granzyme B. These data suggest that inhibition of Bcl-2 could enhance the cytotoxic effects elicited by T-cells when targeted to cancer cells (Sutton et al. (1997) J. Immunol. 158:5783-5790).

SiRNAs are molecules having endogenous RNA bases or chemically modified nucleotides. The modifications do not abolish cellular activity, but rather impart increased stability and/or increased cellular potency. Examples of chemical modifications include phosphorothioate groups, 2'-deoxynucleotide, 2'-OCH 3 -containing ribonucleotides, 2'-F- ribonucleotides, 2'-methoxyethyl ribonucleotides, combinations thereof and the like. The siRNA can have varying lengths {e.g., 10-200 bps) and structures {e.g., hairpins,

single/double strands, bulges, nicks/gaps, mismatches) and are processed in cells to provide active gene silencing. A double-stranded siRNA (dsRNA) can have the same number of nucleotides on each strand (blunt ends) or asymmetric ends (overhangs). The overhang of 1- 2 nucleotides can be present on the sense and/or the antisense strand, as well as present on the 5'- and/ or the 3 '-ends of a given strand. For example, siRNAs targeting Mcl-1 have been shown to enhance the activity of ABT-263 or ABT-737 in various tumor cell lines (Tse et al. (2008) Cancer Res. 68:3421-3428 and references therein).

Multivalent binding proteins are binding proteins comprising two or more antigen binding sites. Multivalent binding proteins are engineered to have the three or more antigen binding sites and are generally not naturally occurring antibodies. The term "multispecific binding protein" means a binding protein capable of binding two or more related or unrelated targets. Dual variable domain (DVD) binding proteins are tetravalent or multivalent binding proteins binding proteins comprising two or more antigen binding sites. Such DVDs may be monospecific (i.e., capable of binding one antigen) or multispecific (i.e., capable of binding two or more antigens). DVD binding proteins comprising two heavy-chain DVD

polypeptides and two light-chain DVD polypeptides are referred to as DVD Ig's. Each half of a DVD Ig comprises a heavy-chain DVD polypeptide, a light-chain DVD polypeptide, and two antigen binding sites. Each binding site comprises a heavy -chain variable domain and a light-chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.

Alkylating agents include altretamine, AMD-473, AP-5280, apaziquone,

bendamustine, brostallicin, busulfan, carboquone, carmustine (BCNU), chlorambucil, Cloretazine™ (laromustine, VNP 40101M), cyclophosphamide, dacarbazine, estramustine, fotemustine, glufosfamide, ifosfamide, KW-2170, lomustine (CCNU), mafosfamide, melphalan, mitobronitol, mitolactol, nimustine, nitrogen mustard N-oxide, ranimustine, temozolomide, thiotepa, treosulfan, trofosfamide and the like.

Angiogenesis inhibitors include epidermal growth factor receptor (EGFR) inhibitors, endothelial-specific receptor tyrosine kinase (Tie-2) inhibitors, insulin growth factor-2 receptor (IGFR-2) inhibitors, matrix metalloproteinase-2 (MMP-2) inhibitors, matrix metalloproteinase-9 (MMP-9) inhibitors, platelet-derived growth factor receptor (PDGFR) inhibitors, thrombospondin analogs, vascular endothelial growth factor receptor tyrosine kinase (VEGFR) inhibitors and the like.

Antimetabolites include Alimta™ (pemetrexed disodium, LY231514, MTA),

5-azacitidine, Xeloda™ (capecitabine), carmofur, Leustat™ (cladribine), clofarabine, cytarabine, cytarabine ocfosfate, cytosine arabinoside, decitabine, deferoxamine,

doxifluridine, eflornithine, EICAR (5-ethynyl-l- -D-ribofuranosylimidazole-4-carboxamide), enocitabine, ethenylcytidine, fludarabine, 5-fluorouracil (5-FU) alone or in combination with leucovorin, Gemzar™ (gemcitabine), hydroxyurea, Alkeran™ (melphalan), mercaptopurine, 6-mercaptopurine riboside, methotrexate, mycophenolic acid, nelarabine, nolatrexed, ocfosfate, pelitrexol, pentostatin, raltitrexed, ribavirin, S-l, triapine, trimetrexate, TS-1, tiazofurin, tegafur, vidarabine, UFT and the like.

Antivirals include ritonavir, hydroxychloroquine and the like.

Aurora kinase inhibitors include ABT-348, AZD-1152, MLN-8054, VX-680, aurora A-specific kinase inhibitors, aurora B-specific kinase inhibitors, pan-aurora kinase inhibitors and the like.

Bcl-2 family protein inhibitors other than ABT-263 or compounds of Formula I herein include AT-101 ((-)gossypol), Genasense™ Bcl-2 -targeting antisense oligonucleotide (G3139 or oblimersen), IPI-194, IPI-565, N-(4-(4-((4'-chloro(l,r-biphenyl)-2-yl)methyl) piperazin- 1 -yl)benzoyl)-4-((( 1 R)-3 -(dimethylamino)- 1 -((phenylsulfanyl)methyl)propyl)amino)- 3-nitrobenzenesulfonamide) (ABT-737), GX-070 (obatoclax) and the like.

Bcr-Abl kinase inhibitors include dasatinib (BMS-354825), Gleevec™ (imatinib) and the like.

CDK inhibitors include AZD-5438, BMI-1040, BMS-387032, CVT-2584, flavopyridol, GPC- 286199, MCS-5A, PD0332991, PHA-690509, seliciclib (CYC-202 or R-roscovitine), ZK- 304709 and the like.

COX-2 inhibitors include ABT-963, Arcoxia™ (etoricoxib), Bextra™ (valdecoxib), BMS-347070, Celebrex™ (celecoxib), COX-189 (lumiracoxib), CT-3, Deramaxx™

(deracoxib), JTE-522, 4-methyl-2-(3 ,4-dimethylphenyl)- 1 -(4-sulfamoylphenyl)- 1 H-pyrrole, MK-663 (etoricoxib), NS-398, parecoxib, RS-57067, SC-58125, SD-8381, SVT-2016, S- 2474, T-614, Vioxx™ (rofecoxib) and the like.

EGFR inhibitors include ABX-EGF, anti-EGFR immunoliposomes, EGF-vaccine, EMD-7200, Erbitux™ (cetuximab), HR3, IgA antibodies, Iressa™ (gefitinib), Tarceva™ (erlotinib or OSI-774), TP-38, EGFR fusion protein, Tykerb™ (lapatinib) and the like.

ErbB2 receptor inhibitors include CP-724714, CI-1033 (canertinib), Herceptin™ (trastuzumab), Tykerb™ (lapatinib), Omnitarg™ (2C4, petuzumab), TAK-165, GW-572016 (ionafamib), GW-282974, EKB-569, PI- 166, dHER2 (HER2 vaccine), APC-8024 (HER2 vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HEPv2 trifunctional bispecific antibodies, mAB AR-209, mAB 2B-1 and the like.

Histone deacetylase inhibitors include depsipeptide, LAQ-824, MS-275, trapoxin, suberoylanilide hydroxamic acid (SAHA), TSA, valproic acid and the like.

HSP-90 inhibitors include 17AAG, CNF-101 , CNF-1010, CNF-2024, 17-DMAG, geldanamycin, IPI-504, KOS-953, Mycograb™ (human recombinant antibody to HSP-90), nab-17AAG, NCS-683664, PU24FC1, PU-3, radicicol, SNX-21 12, STA-9090, VER-49009 and the like.

Inhibitors of apoptosis proteins include HGS-1029, GDC-0145, GDC-0152, LCL-161 , LBW-242 and the like.

Antibody-drug conjugates include anti-CD22-MC-MMAF, anti-CD22-MC-MMAE, anti-CD22-MCC-DMl , CR-01 1-vcMMAE, PSMA-ADC, MEDI-547, SGN-19A, SGN-35, SGN-75 and the like.

Activators of death receptor pathway include TRAIL and antibodies or other agents that target TRAIL or death receptors (e.g., DR4 and DR5) such as apomab, conatumumab, ETR2-ST01 , GDC0145 (lexatumumab), HGS-1029, LBY-135, PRO-1762, trastuzumab and the like.

Kinesin inhibitors include Eg5 inhibitors such as AZD-4877 and ARRY-520, CENPE inhibitors such as GSK-923295A, and the like.

JAK2 inhibitors include CEP-701 (lesaurtinib), XL019, INCB-018424 and the like.

MEK inhibitors include ARRY-142886, ARRY-438162, PD-325901 , PD-98059 and the like. mTOR inhibitors include AP-23573, CCI-779, everolimus, RAD-001 , rapamycin,

temsirolimus, ATP-competitive TORC1/TORC2 inhibitors, including PI-103, PP242, PP30 and Torin 1 , and the like.

Non-steroidal anti-inflammatory drugs include Amigesic™ (salsalate), Dolobid™ (diflunisal), Motrin™ (ibuprofen), Orudis™ (ketoprofen), Relafen™ (nabumetone),

Feldene™ (piroxicam), ibuprofen cream, Aleve™ and Naprosyn™ (naproxen), Voltaren™ (diclofenac), Indocin™ (indomethacin), Clinoril™ (sulindac), Tolectin™ (tolmetin),

Lodine™ (etodolac), Toradol™ (ketorolac), Daypro™ (oxaprozin) and the like.

PDGFR inhibitors include CP-673451 , CP-868596 and the like. Platinum chemotherapeutics include cisplatin, Eloxatin™ (oxaliplatin), eptaplatin, lobaplatin, nedaplatin, Paraplatin™ (carboplatin), picoplatin, satraplatin and the like.

Polo-like kinase inhibitors include BI-2536 and the like.

Phosphoinositide-3 kinase inhibitors include wortmannin, LY -294002, XL- 147, CAL- 120, ONC-21, AEZS-127, ETP-45658, PX-866, GDC-0941, BGT226, BEZ235, XL765 and the like.

Thrombospondin analogs include ABT-510, ABT-567, ABT-898, TSP-1 and the like.

VEGFR inhibitors include Avastin™ (bevacizumab), ABT-869, AEE-788,

Angiozyme™ (a ribozyme that inhibits angiogenesis (Ribozyme Pharmaceuticals (Boulder, CO) and Chiron (Emeryville, CA)), axitinib (AG- 13736), AZD-2171, CP-547632, IM-862, Macugen™ (pegaptanib), Nexavar™ (sorafenib, BAY43-9006), pazopanib (GW-786034), vatalanib (PTK-787 or ZK-222584), Sutent™ (sunitinib or SU-11248), VEGF trap,

Zactima™ (vandetanib or ZD-6474) and the like.

Antibiotics include intercalating antibiotics such as aclarubicin, actinomycin D, amrubicin, annamycin, Adriamycin™ (doxorubicin), Blenoxane™ (bleomycin),

daunorubicin, Caelyx™ and Myocet™ (liposomal doxorubicin), elsamitrucin, epirubicin, glarubicin, idarubicin, mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, Valstar™ (valrubicin), zinostatin and the like.

Topoisomerase inhibitors include aclarubicin, 9-aminocamptothecin, amonafide, amsacrine, becatecarin, belotecan, BN-80915, Camptosar™ (irinotecan hydrochloride), camptothecin, Cardioxane™ (dexrazoxane), diflomotecan, edotecarin, Ellence™ and

Pharmorubicin™ (epirubicin), etoposide, exatecan, 10-hydroxycamptothecin, gimatecan, lurtotecan, mitoxantrone, orathecin, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide, topotecan and the like.

Antibodies include Avastin™ (bevacizumab), CD40-specific antibodies, chTNT-l/B, denosumab, Erbitux™ (cetuximab), Humax-CD4™ (zanolimumab), IGFlR-specific antibodies, lintuzumab, Panorex™ (edrecolomab), Rencarex™ (WX G250), Rituxan™ (rituximab), ticilimumab, trastuzumab, CD20 antibodies types I and II and the like.

Hormonal therapies include Arimidex™ (anastrozole), Aromasin™ (exemestane), arzoxifene, Casodex™ (bicalutamide), Cetrotide™ (cetrorelix), degarelix, deslorelin, Desopan™

(trilostane), dexamethasone, Drogenil™ (flutamide), Evista™ (raloxifene), Afema™ (fadrozole), Fareston™ (toremifene), Faslodex™ (fulvestrant), Femara™ (letrozole), formestane, glucocorticoids, Hectorol™ (doxercalciferol), Renagel™ (sevelamer carbonate), lasofoxifene, leuprolide acetate, Megace™ (megestrol), Mifeprex™ (mifepristone),

Nilandron™ (nilutamide), tamoxifen including Nolvadex™ (tamoxifen citrate), Plenaxis™ (abarelix), prednisone, Propecia™ (finasteride), rilostane, Suprefact™ (buserelin), luteinizing hormone releasing hormone (LHRH) including Trelstar™ (triptorelin), histrelin including Vantas™ (histrelin implant), Modrastane™ (trilostane), Zoladex™ (goserelin) and the like. Deltoids and retinoids include seocalcitol (EB1089 or CB1093), lexacalcitol (KH1060), fenretinide, Panretin™ (alitretinoin), tretinoin including Atragen™ (liposomal tretinoin), Targretin™ (bexarotene), LGD-1550 and the like.

PARP inhibitors include ABT-888, olaparib, KU-59436, AZD-2281, AG-014699, BSI-201, BGP-15, INO-1001, ONO-2231 and the like.

Plant alkaloids include vincristine, vinblastine, vindesine, vinorelbine and the like.

Proteasome inhibitors include Velcade™ (bortezomib), MG132, NPI-0052, PR-171 and the like.

Examples of immunologicals include interferons and other immune-enhancing agents. Interferons include interferon alpha, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma- la, Actimmune™ (interferon gamma- lb), interferon gamma-nl, combinations thereof and the like. Other agents include Alfaferone (IFN-a), BAM-002 (oxidized glutathione), Beromun™ (tasonermin), Bexxar™ (tositumomab), Campath™ (alemtuzumab), CTLA4 (cytotoxic lymphocyte antigen 4), dacarbazine, denileukin, epratuzumab, Granocyte™ (lenograstim), lentinan, leukocyte alpha interferon, imiquimod, MDX-010 (anti-CTLA-4), melanoma vaccine, mitumomab, molgramostim, Mylotarg™ (gemtuzumab ozogamicin), Neupogen™ (filgrastim), OncoVAC-CL, Ovarex™

(oregovomab), pemtumomab (Y-muHMFGl), Provenge™ (sipuleucel-T), sargaramostim, sizofiran, teceleukin, Theracys™ (BCG or Bacillus Calmette-Guerin), ubenimex, Virulizin™ (immunotherapeutic, Lorus Pharmaceuticals), Z-100 (Specific Substance of Maruyama or SSM), WF-10 (tetrachlorodecaoxide or TCDO), Proleukin™ (aldesleukin), Zadaxin™

(thymalfasin), Zenapax™ (daclizumab), Zevalin™ (90Y-ibritumomab tiuxetan) and the like. Biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth or differentiation of tissue cells to direct them to have anti-tumor activity, and include krestin, lentinan, sizofiran, picibanil, PF- 3512676 (CpG-8954), ubenimex and the like.

Pyrimidine analogs include cytarabine (cytosine arabinoside, ara C or arabinoside C), doxifluridine, Fludara™ (fludarabine), 5-FU (5-fluorouracil), floxuridine, Gemzar™

(gemcitabine), Tomudex™ (raltitrexed), triacetyluridine, Troxatyl™ (troxacitabine) and the like.

Purine analogs include Lanvis™ (thioguanine), Purinethol™ (mercaptopurine) and the like.

Antimitotic agents include batabulin, epothilone D (KOS-862), N-(2-((4- hydroxyphenyl)amino)pyridin-3-yl)-4-methoxybenzenesulfonamid e, ixabepilone (BMS- 247550), paclitaxel, Taxotere™ (docetaxel), larotaxel (PNU-100940, RPR-109881 or XRP- 9881), patupilone, vinflunine, ZK-EPO (synthetic epothilone) and the like.

Ubiquitin ligase inhibitors include MDM2 inhibitors such as nutlins, NEDD8 inhibitors such as MLN4924, and the like.

A composition comprising crystalline salts of N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluo rophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API) can also be used as radiosensitizers that enhance the efficacy of radiotherapy. Examples of radiotherapy include, but are not limited to, external beam radiotherapy (XBRT), teletherapy, brachytherapy, sealed-source radiotherapy, unsealed-source radiotherapy and the like.

Additionally or alternatively, a composition comprising crystalline salts of N-(4- {4- amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyr idin-3-yl}phenyl)-N'-(3- fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API) can be administered in combination therapy with one or more antitumor or chemotherapeutic agents selected from Abraxane™ (ABI-007), ABT-100 (farnesyl transferase inhibitor), Advexin™ (Ad5CMV-p53 vaccine or contusugene ladenovec), Altocor™ or Mevacor™ (lovastatin), Ampligen™ (poly(I)-poly(C12U), a synthetic RNA), Aptosyn™ (exisulind), Aredia™ (pamidronic acid), arglabin, L-asparaginase, atamestane (l-methyl-3,17-dione- androsta-l,4-diene), Avage™ (tazarotene), AVE-8062 (combretastatin derivative), BEC2 (mitumomab), cachectin or cachexin (tumor necrosis factor), Canvaxin™ (melanoma vaccine), CeaVac™ (cancer vaccine), Celeuk™ (celmoleukin), histamine including Ceplene™ (histamine dihydrochloride), Cervarix™ (AS04 adjuvant-adsorbed human papilloma virus (HPV) vaccine), CHOP (Cytoxan™ (cyclophosphamide) + Adriamycin™ (doxorubicin) + Oncovin™ (vincristine) + prednisone), combretastatin A4P, Cypat™

(cyproterone), DAB(389)EGF (catalytic and translocation domains of diphtheria toxin fused via a His-Ala linker to human epidermal growth factor), dacarbazine, dactinomycin,

Dimericine™ (T4N5 liposome lotion), 5,6-dimethylxanthenone-4-acetic acid (DMXAA), discodermolide, DX-8951f (exatecan mesylate), eniluracil (ethynyluracil), squalamine including Evizon™ (squalamine lactate), enzastaurin, EPO-906 (epothilone B), Gardasil™ (quadrivalent human papilloma virus (Types 6, 11, 16, 18) recombinant vaccine),

Gastrimmune™, Genasense™ (oblimersen), GMK (ganglioside conjugate vaccine), GVAX™ (prostate cancer vaccine), halofuginone, histerelin, hydroxycarbamide, ibandronic acid, IGN- 101, IL-13-PE38, IL-13-PE38QQR (cintredekin besudotox), IL-13-pseudomonas exotoxin, interferon-a, interferon-γ, Junovan™ and Mepact™ (mifamurtide), lonafarnib, 5,10- methylenetetrahydro folate, miltefosine (hexadecylphosphocholine), Neovastat™ (AE-941), Neutrexin™ (trimetrexate glucuronate), Nipent™ (pentostatin), Onconase™ (ranpirnase, a ribonuclease enzyme), Oncophage™ (vitespen, melanoma vaccine treatment), OncoVAX™ (IL-2 vaccine), Orathecin™ (rubitecan), Osidem™ (antibody-based cell drug), Ovarex™ MAb (murine monoclonal antibody), paclitaxel albumin-stabilized nanoparticle, paclitaxel, Pandimex™ (aglycone saponins from ginseng comprising 20(S)-protopanaxadiol (aPPD) and 20(S)-protopanaxatriol (aPPT)), panitumumab, Panvac™-VF (investigational cancer vaccine), pegaspargase, peginterferon alfa (PEG interferon A), phenoxodiol, procarbazine, rebimastat, Removab™ (catumaxomab), Revlimid™ (lenalidomide), RSR13 (efaproxiral), Somatuline™ LA (lanreotide), Soriatane™ (acitretin), staurosporine (Streptomyces staurospores), talabostat (PT100), Targretin™ (bexarotene), Taxoprexin™ (docosahexaenoic acid (DHA) + paclitaxel), Telcyta™ (canfosfamide, TLK-286), Temodar™ (temozolomide), tesmilifene, tetrandrine, thalidomide, Theratope™ (STn-KLH vaccine), Thymitaq™

(nolatrexed dihydrochloride), TNFerade™ (adenovector: DNA carrier containing the gene for tumor necrosis factor-a), Tracleer™ or Zavesca™ (bosentan), TransMID-107R™ (KSB-311, diphtheria toxins), tretinoin (retin-A), Trisenox™ (arsenic trioxide), Ukrain™ (derivative of alkaloids from the greater celandine plant), Virulizin™, Vitaxin™ (anti-avP3 antibody), Xcytrin™ (motexafm gadolinium), Xinlay™ (atrasentan), Xyotax™ (paclitaxel poliglumex), Yondelis™ (trabectedin), ZD-6126 (N-acetylcolchinol-O-phosphate), Zinecard™

(dexrazoxane), zoledronic acid, zorubicin and the like.

In one embodiment, a composition comprising crystalline salts of N-(4- {4-amino-7- [l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c]pyridin-3-y l}phenyl)-N'-(3- fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API), is administered in a therapeutically effective amount to a subject in need thereof to treat cancer.

Examples include, but are not limited to, acoustic neuroma, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (monocytic, myeloblastic,

adenocarcinoma, angiosarcoma, astrocytoma, myelomonocytic and promyelocytic), acute t- cell leukemia, basal cell carcinoma, bile duct carcinoma, bladder cancer, brain cancer, breast cancer, bronchogenic carcinoma, cervical cancer, chondrosarcoma, chordoma,

choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelocytic (granulocytic) leukemia, chronic myleogeneous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, dysproliferative changes (dysplasias and metaplasias), embryonal carcinoma, endometrial cancer,

endotheliosarcoma, ependymoma, epithelial carcinoma, erythroleukemia, esophageal cancer, estrogen-receptor positive breast cancer, essential thrombocythemia, Ewing's tumor, fibrosarcoma, follicular lymphoma, germ cell testicular cancer, glioma, heavy chain disease, hemangioblastoma, hepatoma, hepatocellular cancer, hormone insensitive prostate cancer, leiomyosarcoma, liposarcoma, lung cancer, lymphagioendotheliosarcoma,

lymphangiosarcoma, lymphoblastic leukemia, lymphoma (Hodgkin's and non-Hodgkin's), malignancies and hyperproliferative disorders of the bladder, breast, colon, lung, ovaries, pancreas, prostate, skin and uterus, lymphoid malignancies of T-cell or B-cell origin, leukemia, lymphoma, medullary carcinoma, medulloblastoma, melanoma, meningioma, mesothelioma, multiple myeloma, myelogenous leukemia, myeloma, myxosarcoma, neuroblastoma, non-small cell lung cancer, oligodendroglioma, oral cancer, osteogenic sarcoma, ovarian cancer, pancreatic cancer, papillary adenocarcinomas, papillary carcinoma, pinealoma, polycythemia vera, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, sarcoma, sebaceous gland carcinoma, seminoma, skin cancer, small cell lung carcinoma, solid tumors (carcinomas and sarcomas), small cell lung cancer, stomach cancer, squamous cell carcinoma, synovioma, sweat gland carcinoma, thyroid cancer, Waldenstrom's macroglobulinemia, testicular tumors, uterine cancer and Wilms' tumor in a mammal,

In a more particular embodiment, a composition comprising crystalline salts of N-(4- {4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3,2-c] pyridin-3-yl}phenyl)-N'-(3- fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X (or prepared using as API), is administered in a therapeutically effective amount to a subject in need thereof to treat myelodysplasia syndrome, acute myeloid leukemia, colorectal cancer, non-small cell lung cancer, and ovarian cancer.

In still further embodiments of the invention, there is provided a method for treating cancer in a mammal comprising dissolving crystalline salts of N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X in a pharmaceutically acceptable solvent or mixture of solvents, and administering the resulting solution in a therapeutically effective amount to subject having the disease.

In still further embodiments of the invention, there is provided a method for treating cancer in a mammal comprising dispersing crystalline salts of N-(4-{4-amino-7-[l-(2- hydroxy ethyl)- 1 H-pyrazol-4-yl]thieno [3 ,2-c]pyridin-3 -yl} phenyl)-N'-(3 -fluorophenyl)urea Form I, II, III, IV, V, VI, VII, VIII, IX, or X in a pharmaceutically acceptable polymeric carrier, and administering the resulting solid dispersion in a therapeutically effective amount to subject having the disease.

EXAMPLES

The following examples are merely illustrative, and do not limit this disclosure in any way.

PXRD data were collected using a G3000 diffractometer (Inel Corp., Artenay, France) equipped with a curved position sensitive detector and parallel beam optics. The

diffractometer was operated with a copper anode tube (1.5 kW fine focus) at 40 kV and 30 mA. An incident beam germanium monochrometer provided monochromatic radiation. The diffractometer was calibrated using the attenuated direct beam at one-degree intervals. Calibration was checked using a silicon powder line position reference standard (NIST 640c). The instrument was computer controlled using the Symphonix software (Inel Corp., Artenay, France) and the data was analyzed using the Jade software (version 6.5, Materials Data, Inc., Livermore, CA). The sample was loaded onto an aluminum sample holder and leveled with a glass slide.

Example 1

Preparation of N-(4-{4-amino-7-[ l-(2-hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3,2- clpyridin-3-yl|phenyl)-N'-(3-fluorophenyl)urea malonate Form I N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solids (100 mg) were suspended in THF/water mixture (80/20 v/v, 500 μί). Solids were collected after three days equilibration at ambient conditions by centrifuge filtration.

Table 1. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea malonte Form I

Peak Position (° 2Θ)

6.136

9.363

10.696

11.684

12.280

13.297

16.321

16.543

16.966

18.474

19.538

21.217

21.498

22.177

23.137

23.456

23.684

Example 2

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yllphenvD-N'-O-fluorophenvDurea dihydrogen L-tartrate Form II N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea L-bitartrate solid (30 mg) was dissolved in THF/water mixture (80/20 v/v, 500 μί). Single crystals were observed after equilibrating at ambient conditions for six weeks.

Table 2. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea L-tartrate Form II

Peak Position (° 2Θ)

6.205

8.879

10.277

10.644

11.958

12.436

12.763

15.933

18.479

19.181

20.581

21.155

21.382

21.745

22.504

22.861

23.855 24.500

24.916

Example 3

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea HC1 Form III

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (27 mg) was suspended in 2-propanol (750 μΐ,) at 40 °C with magnetic stirring. Hydrochloric acid (60 μί, 1 N) was diluted with 2- propanol (250 μί). Hydrochloric acid solution was then slowly added to the A-968660 free base suspension at 40 °C with magnetic stirring. Crystallization was observed shortly after the complete addition of the HC1 solution. Solid was collected by centrifuge filtration.

Example 4

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yllphenvD-N'-O-fluorophenvDurea HC1 Form III

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (126 mg) was suspended in 2-propanol (2.0 mL) at 40 °C with magnetic stirring. Hydrochloric acid (284 μΕ, 1 N) was diluted with 2-propanol (216 μί). Hydrochloric acid solution was then slowly added to A-968660 free base suspension at 40 °C with magnetic stirring. Crystallization was observed shortly after the complete addition of the HC1 solution. Solid was collected by centrifuge filtration.

Example 5

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea HC1 Form III

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (89 mg) was suspended in ethanol (2.5 mL) at 40 °C with magnetic stirring. Hydrochloric acid (380 μΐ,, 1 N) was diluted with water (620 μί). Hydrochloric acid solution was slowly added to A-968660 free base suspension at 40 °C with magnetic stirring. Crystallization was observed shortly after the complete addition of the HC1 solution. Solid was collected by centrifuge filtration. Table 3. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea HC1 Form III

Peak Position (° 2Θ)

5.804

6.358

7.995

9.714

10.222

13.630

16.664

18.691

19.493

19.766

21.590

22.345

22.755

Example 6

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea mesylate Form IV

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (26 mg) was suspended in

dimethylformamide (125 μί) at 40 °C with magnetic stirring. Methanesulfonic acid (59 μί, 1 M) was slowly added to A-968660 free base suspension at 40 °C with magnetic stirring. Clear solution obtained after addition of methanesulfonic acid. Acetonitrile (150 μί) was then added to clear solution at 40 °C with magnetic stirring. Amorphous precipitation was observed shortly after the complete addition of the acid solution. The amorphous suspension was heated to 80 °C to obtain a clear solution, and then cooled spontaneously to ambient temperatures. Crystallization was observed upon cooling. Solid was collected by centrifuge filtration.

Example 7

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea mesylate Form IV

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (26 mg) was suspended in ethyl acetate (100 μί) at 40 °C with magnetic stirring. Methanesulfonic acid (6.6 μί, 4 M) was slowly added to A-968660 free base suspension at 40 °C with magnetic stirring. Amorphous material precipitated upon addition of the acid solution. Additional ethyl acetate (250 μί) was added and the suspension was stirred at 40 °C for 30 minutes. Solid was collected by centrifuge filtration.

Example 8

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea mesylate Form IV

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (71 mg) was suspended in ethyl acetate (1.0 mL) at 45 °C with magnetic stirring. Methanesulfonic acid (36 μΐ ^ , 4 M) was slowly added to A-968660 free base suspension at 40 °C with magnetic stirring. Amorphous material precipitated upon addition of the acid solution. The suspension was stirred at 40 °C overnight. Solid was collected by centrifuge filtration.

Table 4. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea mesylate Form IV

Peak Position (° 2Θ)

5.37

7.399

10.687

11.921 14.311

16.043

18.017

18.385

18.784

20.416

21.195

Example 9

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluoropheny0urea bitartrate Form V

A saturated solution of 1-tartaric acid was prepared at 25 C by dissolving tartaric acid in a 85/15 v/v THF/water mixture. This solution (2 mL) was heated 2 to 65 C.

Approximately 200 mg of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid was then added, which readily dissolved. N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea L-bitartrate crystallized within 30 minutes from this clear solution. The slurry was then diluted with the saturated solution (at 25 °C) of 1-tartaric acid and cooled to ambient temperatures. The slurry was continue to be stirred overnight and the solids were isolated via filtration. The filtered solids were washed with 85/15 v/v mixture of THF/water to remove excess 1-tartaric acid and then dried in the oven at 45 °C.

Table 5. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea mesylate Form V

Peak Position (° 2Θ)

2.696

3.038

3.796

6.579 7.615

16.124

16.553

18.678

19.095

19.826

21.807

22.998

23.576

Example 10

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluoropheny0urea bimalonate Form VI

A saturated solution of malonic acid was prepared at 25 °C by dissolving malonic acid in a 85/15 v/v THF/water mixture. This solution (2 mL) was heated to 65 °C. Approximately 250 mg of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid was then added. A clear solution formed within 10 minutes. The solution was cooled to ambient temperatures and continued to be stirred overnight. Solids crystallized during the overnight hold. They were isolated via filtration and then washed with 85/15 v/v mixture of THF/water to remove excess malonic acid and then dried in the oven at 45 °C.

Table 6. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea bimalonate Form VI

Peak Position (° 2Θ)

6.096

8.716

10.674

11.250

13.084 15.273

15.755

17.461

18.269

18.770

19.416

Example 11

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluoropheny0urea ascorbate Form VII

A saturated solution of ascorbic acid was prepared at 25 °C by dissolving ascorbic acid in a 85/15 v/v THF/water mixture. This solution (2 mL) was heated to 65 °C.

Approximately 250 mg of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]thieno[3 ,2- c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea free base solid was added. A clear solution formed within 10 minutes. The solution was cooled to ambient temperatures. Solids began to crystallize at 45 °C during the cooling process. The suspension was stirred overnight before isolating the solids via filtration. They were washed with 85/15 v/v mixture of THF/water to remove excess ascorbic acid and then dried in the oven at 45 °C.

Table 7. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea ascorbate Form VIII

17.435

18.833

20.930

22.521

22.797

24.128

Example 12

Preparation of N-(4-{4-amino-7-[l-(2-hydroxyethyl)-lH-pyrazol-4-yllthieno[3 ,2-clpyridin-3- yl|phenyl)-N'-(3-fluoropheny0urea maleate Form VIII

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (27 mg) was suspended in

dimethylformamide (125 μί) at 40 °C with magnetic stirring. Maleic acid solution (120 μί, 0.5 M) was then slowly added to A-968660 free base suspension at 50 °C with magnetic stirring. Crystallization was observed upon complete addition of the acid solution. Solid was collected by centrifuge filtration.

Table 8. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea maleate Form VIII

19.790

23.634

Example 13

Preparation of N-(4-{4-amino-7-ri-(2-hvdroxyethyl)-lH-pyrazol-4-yl1thienor3 ,2-c1pyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea sulfate Form IX

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (15 mg) was suspended in 2-propanol (150 μί). Sulfuric acid solution (98%, 4 μΐ,) was added to the suspension. The suspension was stirred at ambient temperatures for 10 days. Solid was collected by centrifuge filtration.

Table 9. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea sulfate Form IX

Peak Position (° 2Θ)

3.848

5.619

6.704

6.906

7.673

11.230

11.533

12.986

16.868

17.418

Example 14

Preparation of N-(4-{4-amino-7-ri-(2-hvdroxyethyl)-lH-pyrazol-4-yl1thienor3 ,2-c1pyridin-3- yl|phenyl)-N'-(3-fluorophenyl)urea phophate Form X

N-(4-{4-amino-7-[l -(2 -hydroxy ethyl)- lH-pyrazol-4-yl]thieno [3, 2-c]pyridin-3- yl}phenyl)-N'-(3-fluorophenyl)urea free base solid (18 mg) was suspended in 2-propanol (150 μΐ,). Phosphoric acid solution (85%, 5 uL) was added to the suspension. The suspension was stirred at ambient temperatures for 10 days. Solid was collected by centrifuge filtration.

Table 10. PXRD Peak Listing: N-(4-{4-amino-7-[l-(2-hydroxy ethyl)- lH-pyrazol-4- yl]thieno[3,2-c]pyridin-3-yl}phenyl)-N'-(3-fluorophenyl)urea phophate Form X.