Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR IMPROVING COGNITIVE FUNCTION
Document Type and Number:
WIPO Patent Application WO/2012/109491
Kind Code:
A1
Abstract:
This invention relates to methods and compositions for treating central nervous system (CNS) disorders with cognitive impairment. In particular, it relates to the use of inhibitors of synaptic vesicle glycoprotein 2A (SV2A), alone or in combination with valproate, in treating central nervous system (CNS) disorders with cognitive impairment in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCl ), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer's Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, amyotrphic lateral sclerosis and cancer-therapy-related cognitive impairment.

Inventors:
GALLAGHER MICHELA (US)
HABERMAN REBECCA (US)
KOH MING TENG (US)
Application Number:
PCT/US2012/024556
Publication Date:
August 16, 2012
Filing Date:
February 09, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV JOHNS HOPKINS (US)
GALLAGHER MICHELA (US)
HABERMAN REBECCA (US)
KOH MING TENG (US)
International Classes:
A01N43/36
Foreign References:
US20100099735A12010-04-22
US20080167291A12008-07-10
Other References:
See also references of EP 2672822A4
Attorney, Agent or Firm:
HALEY, James, F., Jr. (1211 Avenue of the AmericasNew York, NY, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for treating a central nervous system (CNS) disorder with cognitive impairment in a subject in need or at risk thereof, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function in the subject, the method comprising the step of

administering to said subject a therapeutically effective amount of a synaptic vesicle protein 2 A (SV2A) inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

2. The method of claim 1, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is selected from the group of SV2A inhibitors referred to in International Patent Application PCT/US2009/005647; International Patent Application Publications

WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595;

WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658;

WO2002/094787; WO2001/062726; U.S. Patents 7,465,549; 7,244,747;

5,334,720; 4,696,943; 4,696,942; U.S. Patent Applications 12/580,464;

61/105,847; 61 /152,631; and 61/175,536; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692; or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof.

3. The method of claim 1 , wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is selected from the group consisting of ievetiracetam, seietracetam, and brivaraeetam or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof.

4. The method of claim 2, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is

Ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

5. The method of claim 2, wherein the SV2A inhibitor or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

6. The method of claim 2, wherein the SV2A. inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

7. The method of any one of claims 1 to 6, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.1 mg/kg to 5 mg/kg.

8. The method of claim 7, wherein the daily dose is 0.1 - 0.1 mg/kg.

9. The method of any one of claims 1 to 6, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.01 mg/kg to 2,5 mg/kg,

10, The method of claim 9 , wherein the daily dose is 0.1 - 2.5 mg/kg.

The method of claim 9 . wherein the daily dose is 0.4 - 2.5 mg/kg.

12. The method of claim 9 , wherein the daily dose is 0,6 - 1 ,8 mg/kg.

The method of claim 9 , wherein the daily dose is 0,04 mg/kg,

'he method of claim 9 , wherein the daily dose is 0.06 - mg/k

15. The method of any one of claims 1 to 6, wherein the SV2A inhibitor or a harmaceuticallv acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 2 mg/kg to 4 mg/kg,

16. The method of claim 15 , wherein the daily dose is 2 - 3 mg/kg.

17. The method of claim 15 , wherein the daily dose is 3 - 4 mg/kg.

18. The method of any one of claims 1 to 6, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a dai ly dose of 0.2 mg/kg to 0.4 mg/kg.

19. The method of claim 18 , wherein the daily dose is 0.2 - 0.3 mg/kg.

20. The method of claim 18 , wherein the daily dose is 0.3 - 0.4 mg/kg.

21. The method of any one of claims 1 to 6, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0,001 - 5 mg/kg.

The method of claim 21, wherein the daily dose is 0.001 - 0.5 mg/kg.

13. The method of claim 21, wherein the daily dose is 0.01 - 0.5 mg/kg.

24. A method for treating a central nervous system (CNS) disorder with cognitive impairment in a subject in need or at risk thereof, delaying or slowing the progression of cognitive impairment in the subject, or reducing the rate of decline of cognitive function in the subject, the method comprising the step of administering to said subject an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in combination with valproate or a pharmaceutically acceptable salt thereof.

25. The method of claim 24, wherein the valproate or a pharmaceutically acceptable salt thereof is administered at a daily dose such that the subject maintains a blood total valproate level of 0.5 to 5 μ /ηιΙ plasma, and wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered at a daily dose of 0.01 to 1 mg/kg.

26. The method of claim 24, wherein the valproate or a pharmaceutically acceptable salt thereof is administered at a daily dose such that the subject maintains a blood total valproate level of 0.5 to 5 μ /η ί plasma, and wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered at a daily dose of 0.001 to 1 mg/kg.

27. The method of claim 24, wherein the valproate or a pharmaceutically acceptable salt thereof is administered at a dai ly dose such that the subject maintains a blood total valproate level of 0.5 to 5 μ¾/ηι! plasma, and wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered at a dai ly dose of 0.1 to 5 mg/kg.

28. The method of claim 24, wherein the valproate or a pharmaceutically acceptable salt thereof is administered at a daily dose such that the subject maintains a blood total valproate level of 0.5 to 5 μg/ml plasma, and wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered at a daily dose of 0,05 to 0,5 mg/kg.

29. The method of any one of claims 24 to 28, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is selected from the group of SV2A inhibitors referred to in International Patent Application PCT/US2QQ9/Q05647; International Patent Application Publications WO2010/144712; WO2010/002869; WO2008/132139;

WO2007/065595; WO2006/128693; WO2006/128692; WO2005/05 188;

WO2004/087658; WO2002/094787; WO2001/062726; U.S. Patents 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. Patent Applications 12/580,464; 61/105,847; 61/152,631; and 61/175,536; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,1 13; and 1,309,692; or pharmaceutically acceptable salts, hydrates, solvates or polymorphs thereof.

30. The method of any one of claims 24 to 28, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is selected from the group consisting of levetiracetam, seletracetam, and brivaracetam or pharmaceutically acceptable salts, hydrates, solvates or

polymorphs thereof.

31. The method of claim 30, wherein the S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is levetiracetam or a pharmaceutically acceptable salt, hydrate, sol vate or polymorph thereof.

32. The method of claim 30, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

33. The method of claim 30, wherein the SV2 A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

34. The method of any one of claims 24 to 33, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and the valproate or a pharmaceutically acceptable salt thereof are administered simultaneously.

35. The method of any one of claims 24 to 33, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and the valproate or a pharmaceutically acceptable salt thereof are administered in a single formulation.

36. The method of any one of claims 24 to 33, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and the valproate or a pharmaceutically acceptable salt thereof are administered sequentially.

37. A pharmaceutical composition comprising a S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polvmoiph thereof being present in an amount of 5 - 140 nig.

38. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 0.7 - 180 mg.

39. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof, the SV2A. inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 0.07 - 350 mg.

40. A pharmaceutical composition comprising a S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polvmoiph thereof being present in an amount of 50 - 250 mg.

41. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 3 - 50 mg.

42. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 0.05 - 35 mg.

43. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and valproate or a pharmaceutically acceptable salt thereof, the SV2 A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 3 - 50 mg.

44. A pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and valproate or a pharmaceutically acceptable salt thereof, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof being present in an amount of 0.07 - 50 mg.

45. A pharmaceutical composition comprising an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof and valproate or a pharmaceutically acceptable salt thereof,

46. The composition of claim 45, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the composition is present in an amount of 0,05 - 35 mg.

47. The composition of claim 45, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the composition is present in an amount of 0.07 - 350 mg.

48. The composition of claim 45, wherein the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof in the composition is present in an amount of 50 - 250 mg,

49. The composition of claim 45, wherein the S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is present in an amount less than 350 mg, less than 250 mg, less than 200 mg, less than 1 0 mg, less than 100 mg, less than 50 mg, less than 35 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0, 1 mg, less than 0.07 nig, or less than 0.05 mg.

50. A method for treating a central nervous system (C S) disorder with cognitive impairment in a subject in need or at risk thereof, delaying or slowing the progression of cognitive impairment in the subject, or reducing the rate of decline of cognitive function in the subject, the method comprising the step of administering to said subject a therapeutically effective amount of levetiracelam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

51 . The method of claim 50, wherein the ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 1 - 2 nig/kg.

52. The method of claim 50, wherein the Ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered ever}' 12 or 24 hours at a daily dose of 70 - 140 mg.

53. The method of claim 50, wherein the ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.1 - 2,5 rng/kg.

54. The method of claim 50, wherein the Ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 7 - 180 mg.

55. The method of claim 50, wherein the ievetiracetam or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0,4 - 2,5 mg/'kg.

56. The method of claim 50, wherein the Ievetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 25 - 180 mg.

57. The method of claim 50, wherein the levetiracetani or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0,6 - 1 ,8 mg/kg.

58. The method of claim 50, wherein the levetiracetani or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 40 - 130 mg.

59. The method of claim 50, wherein the levetiracetani or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered ever}' 12 or 24 hours at a daily dose of 2.0 - 4.0 mg/kg.

60. The method of claim 50, wherein the levetiracetani or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 140 - 300 mg.

61. The method of claim 50, wherein the levetiracetani or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 3.0 - 4.0 mg/kg,

62. The method of claim 50, wherein the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 200 - 300 mg.

63. The method of claim 50, wherein the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 2.0 - 3.0 mg kg.

64. The method of claim 50, wherein the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 140 - 200 mg.

65. The method of claim 50, wherein the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.1 - 5 mg/kg.

66. The method of claim 50, wherein the levetiracetam or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 7 - 350 nig/kg,

67. The method of claim 50, wherein the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to Table 1 or Table 2.

68. A method for treating a central nervous system (CNS) disorder with cognitive impairment in a subject in need or at risk thereof, delaying or slowing the progression of cognitive impairment in the subject, or reducing the rate of decline of cognitive function in the subject, the method comprising the step of administering to said subject a therapeutically effective amount of brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

69. The method of claim 68, wherein the brivaracetam or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0, 1 - 0.2 mg/kg.

70. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 7 - 15 mg.

71. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered ever}' 12 or 24 hours at a daily dose of 0.01 - 2.5 mg/kg.

72. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0,7 - 180 mg.

73. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.04 - 2,5 mg/kg.

74. The method of claim 68, wherein the brivaracetam or a pharmaceutical ly acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 2.5 - 180 nig.

75. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.06 - 1.8 mg/kg.

76. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered ever}' 12 or 24 hours at a daily dose of 4.0 - 130 mg.

77. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.2 - 0.4 mg/kg.

78. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 14 - 30 mg.

79. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of 0.1 - 35 mg or 0,001 ~ 0.5 mg/kg.

80. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered ever}' 12 or 24 hours at a dai ly dose of at least 0.1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 mg; and at a daily dose of no more than 2.5 mg mg, 10 mg, 1 mg, 20 mg, 25 mg, 30 mg, or 35 mg.

81. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg kg, 0.0075 mg/kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg; and at a d dose of no more than 0.5 mg/kg, 0,4 mg/kg. 0.3 mg/kg, 0.2 mg/kg. 0.15 mg/kg, 0.1 nig/kg, 0.05 mg/kg, or 0.04 mg/kg.

82. The method of claim 68, wherein the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to Table 3 or Table 4.

83. A. method for treating a central nervous system (CNS) disorder with cognitive impairment in a subject in need or at risk thereof, delaying or slowing the progression of cognitive impairment in the subject, or reducing the rate of decline of cognitive function in the subject, the method comprising the step of administering to said subject a therapeutically effective amount of seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof.

84. The method of claim 83, wherein the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every' 12 or 24 hours at a daily dose of 0.1 - 35 mg or 0.0015 - 0.5 mg/kg.

85. The method of claim 83, wherein the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1 ,5 mg, or 2.0 mg; and at a daily dose of no more than 2,5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, or 35 mg.

86. The method of claim 83, wherein the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg; and at a daily dose of no more than 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0,2 mg/kg, 0.15 mg/kg, 0, 1 mg/kg, 0.05 mg/kg, or 0.04 mg/kg.

87. The method of claim 83, wherein the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to Table 5 or Table 6.

88. The method of any one of claims 1, 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is age-related cognitive impairment.

89. The method of claim 88, wherein the age-related cognitive impairment is Mild Cognitive Impairment,

90. The method of claim 89, wherein the Mild Cognitive

Impairment is amnestic Mild Cognitive Impairment.

91. The method of any one of claims 1, 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is dementia.

92. The method of claim 91 , wherein the dementia is Alzheimer's disease.

93. The method of any one of claims 1 , 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is schizophrenia.

94. The method of any one of claims 1, 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is amyotrophic lateral sclerosis.

95. The method of any one of claims 1, 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is post traumatic stress disorder,

96. The method of any one of claims 1 , 24, 50, 68 and 83, wherein the CNS disorder with cognitive impairment is associated with cancer therapy.

Description:
[0001] This application claims priority and benefit from U.S. Provisional Patent Application 61/441 ,251 , filed February 9, 2011, the contents and disclosures of which are incorporated herein by reference in their entirety,

Field of the Invention

[0002] This invention relates to methods and compositions for treating central nervous system (CNS) disorders with cognitive impairment. In particular, it relates to the use of inhibitors of synaptic vesicle glycoprotein 2A (SV2A). alone or in combination with valproate, in treating central nervous system (CNS) disorders with cognitive impairment in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment ( MCI ), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (AR(. ' D). dementia,

Alzheimer's DiseasefAD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, amyotrphic lateral sclerosis and cancer-therapy-related cognitive impairment. Background! of the Invention

[0003] Cogniti ve ability may decline as a normal consequence of aging or as a consequence of a CNS disorder.

[0004] A significant population of elderly adults experiences a decline in cognitive ability that exceeds what is typical in normal aging. Such age-related loss of cognitive function is characterized clinically by progressive loss of memory, cognition, reasoning, and judgment. Mild Cognitive Impairment (MCI), Age-Associated Memory Impairment (AAMI), Age-Related Cognitive Decline (ARCD) or similar clinical groupings are among those related to such age-related loss of cognitive function. According to some estimates, there are more than 16 million people with AAM I in the U.S. alone (Barker et al, 1995), and MCI is estimated to affect 5.5 - 7 million in the U.S. over the age of 65 (Plassman et al., 2008). [0005] Other centra] nervous system (CNS) disorders, such as dementia,

Alzheimer's Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, amyotrophic lateral sclerosis (ALS) and cancer-therapy-related cognitive impairment, are also associated with cognitive impairment.

10006] There is, therefore, a need for effective treatment for centra! nervous system (CNS) disorders with cognitive impairment and to improve cognitive function in patients diagnosed with age-related cognitive impairment, MCI, amnestic MCI, AAMI, ARCD, dementia, AD, prodromal AD, PTSD,

schizophrenia, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, and similar central nervous system (CNS) disorders with cognitive impairment or at risk of developing them,

Summary of the Invention

[0007] In accordance with a first aspect of the present invention, there is provided a method for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a central nervous system (CNS) disorder with cognitive impairment, or at risk thereof, the method comprising the step of administering to said subject a therapeutically effecti ve amount of an S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In some embodiments of this aspect of the invention, the methods improve or treat cognitive function in said subject. In some embodiments of this aspect of the invention, the methods delay or slow the progression of cogniti ve impairment in said subject, in some embodiments of this aspect of the invention, the methods reduce the rate of decline of cognitive function in said subject. In some embodiments of this aspect of the invention, the methods prevent or slow the progression of said CNS disorder with cognitive impairment in said subject. In other embodiments of this aspect of the invention, the methods alle viate, ameliorate, or slow the progression, of one or more symptoms associated with said CNS disorder with cognitive impairment in said subject,

[0008] In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is age-related cognitive impairment, such as Mild

Cognitive Impairment ( MCI ). Age-Associated Memory Impairment (AAMI), Age R elated Cognitive Decline (ARCD). In one embodiment of this aspect of the invention, the MCI is amnestic MCI. In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is dementia, Alzheimer's Disease( AD), prodromal AD, post traumatic stress disorder (PTSD),

schizophrenia, amyotrophic lateral sclerosis (ALS) or cancer-therapy-related cognitive impairment. In one embodiment of this aspect of the invention, the subject that suffers such a CNS disorder or cognitive impairment is a human patient. [0009] The SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this aspect of the invention include those disclosed in, for example, United States (U.S.) Patent Application 12/580,464, International Patent Application

PCT/US20Q9/QQ5647, U.S. Patent Application 61/105,847, U.S. Patent

Application 61/152,631, and U.S. Patent Application 61/175,536. However, any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of this aspect of the invention. In other embodiments, the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in International Patent Applications

WO2010/144712; WO2010/002869; WO2008/132139; WO2007/065595;

WO2006/128693; WO2006/128692; WO2005/054188; WO2004/087658;

WO2002/094787; WO2001/062726; U.S. Patents 7,465,549; 7,244,747;

5,334,720; 4,696,943; 4,696,942; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,113; and 1,309,692 or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs. In other embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof. In other embodiments, the SV2A inhibitor is levetiracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is brivaracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is seletracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. 10010] In other embodiments of this aspect of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof can be administered at doses as disclosed, for example, in U.S. Patent Application 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61 /105,847, U.S. Patent Application 61/152,631, U.S. Patent

Application 61/175,536, and U.S. Patent Application 61/441,251. In other embodiments of this aspect of the in vention, the SV2A inhibitor or a

pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0.2 nig/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 2.0 to 4.0 mg/kg, or about 2.0 to 3.0 mg kg, or about 3.0 to 4.0 mg/kg, or about 0.2 to 0.4 mg/kg, or about 0.2 to 0.3 mg/kg, or about 0.3 to 0.4 mg/kg, or about 0.001 - 5 mg/kg, or about 0.001 - 0.5 mg/kg, or about 0.01 - 0.5 mg/kg.

[0011] In accordance with a second aspect of the present invention, there is provided a method for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a central nervous system (CNS) disorder with cognitive impairment, or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph in combination with valproate or an analog, derivative or pharmaceutically acceptable salt thereof, In some embodiments of this aspect of the invention, the methods improve or treat cognitive function in said subject. In some embodiments of this aspect of the invention, the methods delay or slow the progression of cognitive impairment in said subject. In some embodiments of this aspect of the invention, the methods reduce the rate of decline of cognitive function in said subject. In some embodiments of this aspect of th e invention, th e methods prevent or slow the progression of said CNS disorder with cognitive impairment in said subject. In other embodiments of this aspect of the invention, the methods alleviate, ameliorate, or slo the progression, of one or more symptoms associated with said CNS disorder with cognitive impairment in said subject.

[0012] In some embodiments of this aspect of the invention, the SV2A inhibitor and/or valproate are administered at doses that are subtherapeutic as compared to the doses at which they are therapeutically effective when administered in the absence of the other. [0013] In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is age-related cognitive impairment, such as Mild Cognitive Impairment (MCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD). In one embodiment of this aspect of the invention, the MCI is amnestic MCI, In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is dementia, Alzheimer's Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia or cancer-therapy-related cognitive impairment. In one embodiment, the subject that suffers such cognitive impairment is a human patient.

[0014] The SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is usef l in the methods and compositions of this aspect of the invention include those disclosed in, for example, United States (U.S.) Patent Application 12/580,464, International Patent Application

PCT US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent

Application 61/152,631, U.S. Patent Application 61/175,536, and U.S. Patent Application 61/441 ,251. However, any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of this aspect of the invention. In other embodiments, the SV2A inhibitor is selected from the group of SV2A inhibitors referred to in Internationa! Patent Applications WO2010/144712; WO2010/002869; WO2008/132139;

WO2007/065595; WO2006/128693; WO2006/128692; WO2005/054188;

WO2004/087658; WO2002/094787; WO2001/062726; U.S. Patents 7,465,549; 7,244,747; 5,334,720; 4,696,943; 4,696,942; U.S. Patent Application Publication Numbers 20090312333; 20090018148; 20080081832; 2006258704; and UK Patent Numbers 1,039,1 13; and 1,309,692 or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs. In other embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, hydrates, solvates, or polymorphs thereof. In other embodiments, the SV2A inhibitor is levetiracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2A inhibitor is brivaracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof. In other embodiments, the SV2 A inhibitor is seletracetam or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.

[0015] in other embodiments of this aspect of the in vention, the S V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is administered in combination with valproate or its analog, derivative or

pharmaceutically acceptable salt can be administered at doses as disclosed, for example, in U.S. Patent Application 12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent

Application 61/152,631, U.S. Patent Application 61/175,536, and U.S. Patent

Application 61/441 ,251 , In other embodiments of this aspect of the invention, the SV2A. inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is administered in combination with valproate or its analog, derivative or pharmaceutically acceptable salt is administered every 12 or 24 hours at a daily dose of about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.1 mg/kg to 5 mg/kg, or about 0.05 mg/kg to 0.5 mg/kg. [0016] In certain embodiments of this a spect of the invention, va lproate or an analog, derivative or pharmaceutically acceptable salt thereof that is administered in combination with the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered at a daily dose such that the subject maintains a blood total valproate level of 0.5 to 5 μ^ηιΐ plasma.

|0017] In other embodiments of this aspect of the invention, the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph and the valproate or its analog, derivative or pharmaceutically acceptable salt are administered simultaneously, or sequentially, or in a single formulation or in separate formulations packaged together. In other embodiments of this aspect of the invention, the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph and the valproate or its analog, derivative or

pharmaceutically acceptable salt are administered via different routes. As used herein, "combination" includes administration by any of these formulations or routes of admini stration .

[0018] In accordance with a third aspect of the present invention, there is provided a pharmaceutical composition comprising a SV2A inhibitor or a pharmaceutically acceptable salt thereof. In certain embodiments of this aspect of the invention, the SV2A inhibitor is present in an amount of 0.07 - 60 mg, 0,07 - 350 mg, 25 - 60 mg, 25 - 125 mg, 50 - 250 mg, 5 - 140 mg, 0,7 - 180 mg, 125 - 240 mg, 3 - 50 mg, or 3 - 60 mg. . In other embodiments of this aspect of the invention, the SV2A inhibitor is present in an amount of 0.05 - 35 mg.

[0019] In accordance with a fourth aspect of this invention, there is provided a pharmaceutical composition comprising an S V2A inhibitor or a pharmaceutically acceptable salt thereof in combination with valproate or an analog, derivative or pharmaceutically acceptabl e salt thereof. In some embodiments of this aspect of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt thereof is present in an amount of 0.05 - 35 mg, 0.07 - 60 mg, 0.07 - 350 mg, 25 - 60 mg, 25 - 125 mg, 50 - 250 mg, 5 - 15 mg, 5 - 30 mg, 5 - 140 mg, 0.7 - 180 mg, 125 240 mg, 3 - 50 mg, or 0.07 - 50 mg, or 3 - 60 mg, , In other embodiments, the amount of the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is less than 350 mg, less than 250 mg, less than 200 mg, less than 1 0 mg, less than 100 mg, less than 50 mg, less than 35 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0,5 mg, less than 0.1 mg, less than 0.07 mg, or less than 0.05 mg. [0020] In accordance with a fift aspect of the present invention, there is provided a method for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a central nervous system (CNS) disorder with cognitive impairment, or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of levetiracetam or a pharmaceutically acceptable salt thereof. In some embodiments of this aspect of the invention, the methods improve or treat cognitive function in said subject. In some embodiments of this aspect of the invention, the methods dela y or slow the progression of cognitive impairment in said subject. In some embodiments of this aspect of the invention, the methods reduce the rate of decline of cognitive function in said subjectin some embodiments of this aspect of the invention, the methods prevent or slow the progression of said CNS disorder with cognitive impairment in said subject. In other embodiments of this aspect of invention, the methods alleviate, ameliorate, or slo the progression, of one or more symptoms associated with said CNS disorder with cognitive impairment in said subject,

[0021] In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is age-related cognitive impairment, such as Mild Cognitive Impairment (MCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (AR.CD). In one embodiment of this aspect of the invention, the MCI is amnestic MCI. In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is dementia, Alzheimer's Disease (AD), prodromal AD, post traumatic stress disorder (PTSD),

schizophrenia or cancer-therapy-related cognitive impairment, In one

embodiment, the subject that suffers such cognitive impairment is a human patient. f 0022] in certain embodiments of this aspect of the invention, the levetiracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours a t a daily dose of about 1 to 2 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg kg, or about 2.0 to 3.0 mg/kg, or about 3.0 to 4.0 mg/kg, or about 2.0 to 4.0 mg/kg, or about 0.1 - 5 mg/kg, or about 70 to 140 mg, or about 7 to 1 80 mg, or about 25 - 180 mg, or about 40 to 130 mg, or about 140 to 300 mg, or about 200 to 300 mg, or about 140 to 200 mg, or about 7 - 350 mg.

[0023] In certain embodiments of this aspect of the invention, the levetiraeetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to one of the daily dose ranges indicated as "+" listed in Table 1 or Table 2.

[0O24J In accordance with a sixth aspect of the present in vention, there is provided a method for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a central nervous system (CNS) disorder with cognitive impairment, or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of brivaracetam or a pharmaceutically acceptable salt thereof. In some embodiments of this aspect of the invention, the methods improve or treat cognitive function in said subject. In some embodiments of this aspect of the invention, the methods delay or slo w the progression of cogniti ve impairment in said subject. In some embodiments of this aspect of the invention, the methods reduce the rate of decline of cognitive function in said subject.In some embodiments of this aspect of the invention, the methods prevent or slow the progression of said CNS disorder with cognitive impairment in said subject. In other embodiments of this aspect of invention, the methods alleviate, ameliorate, or slow the progression, of one or more symptoms associated with said CNS disorder with cognitive impairment in said subject.

[0025] In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is age-related cognitive impairment, such as Mild Cognitive Impairment (MCI), Age- Associated Memor Impairment (AAMI), Age Related Cognitive Decline (ARCD). In one embodiment of this aspect of the invention, the MCI is amnestic MCI. In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is dementia, Alzheimer's Disease (AD), prodromal AD, post traumatic stress disorder (PTSD),

schizophrenia or cancer-therapy-related cognitive impairment. In one

embodiment, the subject that suffers such cognitive impairment is a human patient.

[0026] In certain embodiments of this aspect of the invention, the brivaracetam or the pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0.2 mg/ ' kg, or about 0.01 to 2.5 mg/kg, or about 0.04 to 2.5 mg/ ' kg, or about 0.06 to 1.8 nig/kg, or about 0.2 to 0.4 mg/kg, or about 7 to 15 mg, or about 0.7 to 180 mg, or about 2.5 to 180 mg, or about 4.0 to 130 mg, or about 14 to 30 mg.

[0027] In certain embodiments of this aspect of the invention, the brivaracetam or the pharmaceutically acceptable salt, hydrate, sol vate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 mg; but no more than a daily dose of 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, or 35 mg. In other embodiments, the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/ ' kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/ ' kg; but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.1 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.04 mg/ ' kg.

[0028] In certain embodiments of this aspect of the invention, the bri varacetam or a pharmaceutically acceptable salt, hydrate, sol vate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to one of the daily dose ranges indicated as listed in Table 3 or Table 4. For example, the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be administered every 12 or 24 hours at a daily dose of 0.1 - 35 mg, 0.5 - 35 mg, 0.75 - 35 mg, 1.0 - 35 mg, 1.5 - 35 mg, 2.0 - 35 mg, 0.1 - 30 mg, 0.1 - 25 mg, 0.1 - 20 mg, 0.1 - 15 mg, 0.1 - 10 mg, 0.1 - 5 mg, 0.1 - 2.5 mg, 0.0015 - 0.5 mg/kg, 0.0075 - 0.5 mg/kg, 0.01 - 0.5 mg/kg, 0.015 - 0.5 mg/kg, 0.02 - 0.5 mg/kg, 0.03 - 0.5 mg/kg, 0.0015 - 0.4 mg/ ' kg, 0.0015 - 0.3 mg/kg, 0.0015 - 0.2 mg/kg, 0.0015 - 0.15 nig/kg, 0.0015 - 0.1 mg/kg, 0.0015 - 0.05 mg/kg, or 0.0015 - 0.04 mg kg.

[0029] in accordance with a seventh aspect of the present invention, there is provided a method for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a central nervous system (CNS) disorder with cognitive impairment, or at risk thereof, the method comprising the step of administering to said subject a therapeutically effective amount of selectracetam or a pharmaceutically acceptable salt thereof. In some embodiments of this aspect of the invention, the methods improve or treat cognitive function in said subject. In some embodiments of this aspect of the invention, the methods delay or slo the progression of cognitive impairment in said subject. In some embodiments of this aspect of the invention, the methods reduce the rate of decline of cogniti ve function in said subject. In some embodiments of this aspect of the invention, the methods prevent or slow the progression of said CNS disorder with cognitive impairment in said subject. In other embodiments of this aspect of invention, the methods alleviate, ameliorate, or slow the progression, of one or more symptoms associated with said CNS disorder with cognitive impairment in said subject. [0030 J In some embodiments of this aspect of the in vention, the CN S disorder with cognitive impairment is age-related cognitive impairment, such as Mild Cognitive Impairment (MCI), Age-Associated Memory Impairment (ΑΑΜΪ), Age Rela ted Cognitive Decline (ARCD). In one embodiment of this aspect of the invention, the MCI is amnestic MCI, In some embodiments of this aspect of the invention, the CNS disorder with cognitive impairment is dementia, Alzheimer's Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia or cancer-therapy-related cognitive impairment. In one embodiment, the subject that suffers such cognitive impairment is a human patient.

[0031] In certain embodiments of this aspect of the invention, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.1 nig, 0.5 mg, 0.75 nig, 1.0 mg, 1.5 nig, or 2.0 mg; but no more than a daily dose of 2.5 nig, 5 nig, 10 nig, 15 nig, 20 nig, 25 mg, 30 mg, or 35 mg. In other embodiments, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 nig/kg, 0.01 mg/ ' kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg kg: but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.15 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.04 mg kg.

[0032 j In certain embodiments of this aspect of the invention, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose according to one of the daily dose ranges indicated as "+" listed in Table 5 or Table 6. For example, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be administered every 12 or 24 hours at a daily dose of 0.1 - 35 mg, 0.5 - 35 mg, 0.75 - 35 mg, 1 .0 - 35 mg, 1 .5 - 35 mg, 2.0 - 35 mg, 0.1 - 30 mg, 0.1 - 25 mg, 0.1 - 20 mg, 0.1 - 15 mg, 0.1 - 10 mg, 0.1 - 5 mg, 0.1 - 2.5 mg, 0.0015 - 0.5 mg/kg, 0.0075 - 0.5 mg/kg, 0.01 - 0.5 mg/kg, 0.015 - 0.5 mg/kg, 0.02 - 0.5 mg/kg, 0.03 - 0.5 mg/kg, 0.0015 - 0.4 mg/kg, 0.0015 - 0.3 mg/kg, 0.0015 - 0.2 mg/kg, 0.0015 - 0,1 mg/kg, 0.0015 - 0.1 mg kg, 0.0015 - 0.05 mg/kg, or 0.0015 - 0.04

Brief Description of the Drawings

[0033] FIG. 1 depicts increased mRNA expression of the gene encoding SV2A in the dentate gyrus of the hippocampus of aged-impaired rats (AI) as compared to young rats (Y) and aged-unimpaired rats (AU). Normalized Affymetrix GeneChip probe set signal values (Y-axis), as a measure of mRNA expression, are plotted against learning indices of different rats, as a measure of cogniti ve impairment.

[0034] FIG, 2 depicts the effects of administering levetiracetam on the spatial memory retention of six aged-impaired rats (AI) in a M orris Water Maze (MWM) test. Three treatment conditions were employed: vehicle control, levetiracetam (5 mg kg day) and levetiracetam (10 nig/kg/day), The AI rats were trained for two consecutive days, with a one-time treatment prior to the training trials per day. 24 hours later, the AI rats were tested. The time the AI rats, 24 hours after treatment with the different conditions and two days of training, spent swimming in the target quadrant or the target annulus in a memory retention trial is used as a measure of spatial memory retention. The target quadrant refers to the quadrant of the maze (which is a circular pool) where the escape platform is placed during the training trials, The target annulus refers to the exact location of the escape platform during the training trials. [0035 J FIG. 3 depicts the effects of administering levetiracetam on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test. Six treatment conditions were employed: vehicle control, levetiracetam (1.25 mg kg), levetiracetam (2.5 mg/kg), levetiracetam (5 mg/ ' kg), levetiracetam (10 mg/kg) and levetiracetam (20 mg/kg). In the RAM task used, there w r as a one-hour delay between presentation of a subset of arms (5 arms a vail able and 3 arms blocked) and completion of the eight-arm win-shift task (eight arms available). Rats were pre -treated 30 - 40 minutes before daily trials with a one-time drug/control treatment. The number of errors made by the rats after the delay was used as a measure of spatial memory retention. Errors were defined as instances when rats entered an arm from which food had already been retrieved in the pre-del ay component of the trial or when rats re- visited an arm in the post-delay session that had already been visited. Paired t- tests were used to compare the number of errors between different doses of levetiracetam and vehicle control. [0036] FIG. 4 depicts the effects of administering levetiracetam or valproate separately on the spatial memory retention often aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test.

[0037] FIG. 5 depicts the effects of administering levetiracetam or valproate in combination on the spatial memory retention of ten aged-impaired rats (AI) in an eight-arm Radial Arm Maze (RAM) test. [0038] FIG. 6 shows an isobologram plotting levetiracetam dose against valproate dose. The diagonal straight line is the line of additivity, anchored on each axis by the lowest effective doses of valproate and levetiracetam when assessed individually. [0039] FIG, 7 depicts the experimental design of the human trials for levetiracetam treatment.

[0040] FIG. 8A depicts the average activity in the left CA3 of aMCI subjects with placebo treatment and age-matched control subjects with placebo treatment during the presentation of Sure stimuli that the subject correctly identified as "similar."

[0041] FIG. 8B depicts the average activity in the left CA3 of aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as "similar." [0042] FIG, 8C is a table of the data represented in FIGS. 8A and 8B.

[0043] FIG, 9A depicts the average activity in the left entorhinal corte of age- matched control subjects with placebo treatment and aMCI subjects with placebo treatment during the presentation of lure stimuli that the subject correctly identified as "similar," [0044] FIG. 9B depicts the average activity in the left entorhinal cortex of the same aMCI subjects with placebo treatment or levetiracetam treatment (125 mg twice a day for two weeks) during the presentation of lure stimuli that the subject correctly identified as "similar."

[0045] FIG. 9C is a table of the data represented in FIGS. 9A and 9B. [0046] FIG. 10A depicts an example of the sequence of images shown to subjects in the explicit 3-al tentative forced choice task described in Example 2,

[0047] FIG, 10B shows sample pairs of similar ("lure") images. [0048] FIG. 11 shows the difference between the aMCI (placebo) subjects and age-matched control (placebo) subjects in their performance of the explicit 3- ai tentative forced choice task described in Example 2. Each bar represents the proportion of the subject responses (old, similar, or new) when presented with a lure image.

[0049] FIG. 12 shows the difference between the same aMCI subjects with placebo treatment or with levetiracetam treatment (125 mg twice a day for two weeks) in their performance of the explicit 3 -alternative forced choice task described in Example 2. Each bar represents the proportion of the subjects responses (old, similar, or new) when presented with a lure image.

[0050] FIG. 13 is a table of the data represented in FIGS. 11 and 12.

[0051] FIG. 14 A shows the difference between the age-matched control (placebo) subjects and the aMCI subjects treated with placebo or with

le vetiracetam (125 mg twice a day for two weeks ) in their performance of the Bushke Selective reminding Test - Delayed Recall.

[0052] FIG. 14B is a table of the data represented in FIG. 14A.

[0053] FIG. 15A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Benton Visual Retention Test. [0054] FIG. 15B is a table of the data represented in FIG. I SA,

[0055] FIG. 16 A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates l est - Recognition. [0056] FIG. 16B is a table of the data represented in FIG. 16A.

[0057] FIG. 17A shows the difference between the control (placebo) subjects and the aMCI subjects treated with placebo or with levetiracetam (125 mg twice a day for two weeks) in their performance of the Verbal Paired Associates Test - Delayed Recall.

10058] FIG. 17B is a table of the data represented in FIG. 17A.

10059] FIG. 18A is a table showing the subject selection process for the human levetiracetam trial described in Example 2.

[0060] FIG. 18B is a table showing the characteristics of the subjects selected for the human levetiracetam trial described in Example 2,

Detailed Description of the Invention

[0061] Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurocheniistry, virology, immunology, microbiology,

pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art,

[0062] The methods and techniques of the present invention are generally performed, unless otherwise indicated, according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout this specification. See, e.g. "Principles of Neural Science", McGraw-Hill Medical, New York, N.Y. (2000); Motulsky, "Intuitive Biostatistics", Oxford University Press, Inc. (1995); Lodish et al., "Molecular Cell Biology, 4th ed.", W. H. Freeman & Co., New York (2000);

Griffiths et al., "Introduction to Genetic Analysis, 7th ed.", W. H. Freeman & Co., N.Y. (1999); Gilbert et al,, "Developmental Biology, 6th ed.", Sinauer Associates, Inc., Sunderland, MA (2000).

[0063 ] Chemistry terms used herein are used according to conventional usage in the art, as exemplified by "The McGraw-Hill Dictionary of Chemical Terms", Parker S., Ed., McGraw-Hill, San Francisco, C.A. (1985). [0064] All of the above , and any other publications, patents and published patent applications referred to in this application are specifically incorporated by reference herein, In case of conflict, the present specification, including its specific definitions, will control. [0065] Throughout this specification, the word "comprise" or variations such as "comprises" or "comprising" will be understood to imply the inclusion of a stated integer (or components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or

components). [0066] The singular forms "a." "an," and "the" include the plurals unless the context clearly dictates otherwise.

[0067] The term "including" is used to mean "including but not limited to". "Including" and "including but not limited to" are used interchangeably.

[0068] The term "agent" is used herein to denote a chemical compound (such as an organic or inorganic compound, a mixture of chemical compounds), a biological macromolecule (such as a nucleic acid, an antibody, including parts thereof as well as humanized, chimeric and huma antibodies and monoclonal antibodies, a protein or portion thereof, e.g., a peptide, a lipid, a carbohydrate), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. Agents include, for example, agents which are known with respect to stmcture, and those which are not known with respect to structure. The SV2A inhibitory activity of such agents may render them suitable as "therapeutic agen ts" in the methods and composit ions of this invent ion.

[0069] A "patient", "subject", or "individual" are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovmes, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats).

[0070] "Cognitive function" or "cognitive status" refers to any higher order intellectual brain process or brain state, respectively, involved in learning and/or memory including, but not limited to, attention, information acquisition, information processing, working memory, short-term memory ' , long-term memory, anterograde memory, retrograde memory, memory retrieval, discrimination learning, decision-making, inhibitor response control, attentional set-shifting, delayed reinforcement learning, reversal learning, the temporal integration of voluntary behavior, and expressing an interest in one's surroundings and self-care.

[0071] In humans, cognitive function may be measured, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test (Buschke and Puld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual

Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R)

(Wechsler, 1997); the Benton Visual Retention Test; or the explicit 3-alternative forced choice task. See Folstein et al, J Psychiatric Res 12: 189-98, (1975);

Robbins et al, Dementia 5: 266-81, (1994); Rey, L'examen clinique en

psychologic, (1964); Kluger et al, J Geriatr Psychiatry Neurol 12: 168-79, (1999); Marquis et al, 2002 and Masur et al., 1994. [0072] in animal model systems, cognitive function may be measured in various conventional ways known in the art, including using a Morris Water Maze

( MWM), Barnes circular maze, elevated radial arm maze, T maze or any other mazes in which the animals use spatial information. Other tests known in the art may also be used to assess cognitive function, such as novel object recognition and odor recognition tasks.

[0073] Cognitive function may also be measured using imaging techniques such as Positron Emission Tomography (PET), functional magnetic resonance imaging (fMRI), Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function. In animals, cognitive function may also be measured with electrophysiological techniques. [0074] "Promoting' ' ' cognitive function refers to affecting impaired cognitive function so that it more closely resembles the function of a normal, unimpaired subject. Cognitive function may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to cany ' out daily activities of normal l ife at the same level of proficiency as a normal, unimpaired subject,

[0075] In some cases, "promoting" cognitive function in a subject affected by age-related cognitive refers to affecting impaired cognitive function so that it more closely resembles the function of an aged-matched normal, unimpaired subject, or the function of a young adult subject. Cognitive function of that subject may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to cany' out daily activities of normal life at the same level of proficiency as an aged-matched normal, unimpaired subject or as a young adult subject. [0076] "Preserving" cognitive function refers to affecting normal or impaired cognitive function such that it does not decline or does not fall below that observed in the subject upon first presentation or diagnosis, or delays such decline.

[0077] "Improving" cognitive function includes promoting cognitive function and/or preserving cognitive function in a subject. [0078] "Cognitive impairment" refers to cognitive function in subjects that is not as robust as that expected in a normal, unimpaired subject. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in a normal, unimpairmed subject. In some cases, "cognitive impairment" in subjects affected by aged-related cognitive impairment refers to cognitive function in subjects that is not as robust as that expected in an aged-matched normal, unimpaired subject, or the function of a young adult subject (i.e. subjects with mean scores for a given age in a cognitive test). [0079] "Age-related cognitive impairment" refers to cognitive impairment in aged subjects, wherein their cognitive function is not as robust as that expected in an age-matched normal subject or as that expected in young adult subjects, In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in an age-matched normal subject. In some cases, cognitive function is as expected in an age-matched normal subject, but reduced by about 5%, about 10%, about 30%, about 50% or more, compared to cognitive function expected in a young adult subject. Age-related impaired cognitive function may be associated with Mild Cognitive Impairment (MCI) (including amestic MCI and non-amnestic MCI), Age-Associated Memory Impairment (AAMI), and Age-related Cognitive Decline (A CD).

10080] "Cognitive impairment" associated with AD or related to AD or in AD refers to cognitive function in subjects that is not as robust as that expected in subjects who have not been diagnosed AD using conventional methodologies and standards.

[0081] "Mild Cognitive Impairment" or "MCI" refers to a condition

characterized by isolated memory impairment unaccompanied other cognitive abnormalities and relatively normal functional abilities. One set of criteria for a clinical characterization of MCI specifies the following characteristics: (1) memory complaint (as reported by patient, informant, or physician), (2) normal activities of daily living (ADLs), (3) normal global cognitive function, (4) abnormal memory for age (defined as scoring more than 1.5 standard deviations belo the mean for a given age), and (5) absence of indicators of dementia (as defined by DSM-IV guidelines). Petersen et al, Srch. Neurol. 56: 303-308 (1999); Petersen, "Mild cognitive impairment: Aging to Alzheimer's Disease." Oxford University Press, N.Y. (2003).

[0082] Diagnosis of MCI usually entails an objective assessment of cognitive impairment, which can be garnered through the use of well-established

neuropsychological tests, including the Mini Mental State Examination (MMSE), the Cambridge Neuropsychological Test Automated Battery (CANTA B) and individual tests such as Rey Auditory Verbal Learning Test (AVLT), Logical Memory Subtest of the revised Wechsler Memory Scale (WMS-R) and the New York University (NYU) Paragraph Recall Test. See Folstein et a!., J Psychiatric Res 12: 189-98 (1975); Robbins et al, Dementia 5: 266-81 (1994): Kluger et al, J Geriatric Psychiatry Neurol 12: 168-79 (1999). [0083] "Age-Associate Memory Impairment (AAMI)" refers to a decline in memory due to aging. A patient may be considered to have AAMI if he or she is at least 50 years old and meets all of the following criteria: a) The patient has noticed a decline in memory performance, b) The patient performs worse on a standard test of memory compared to young adults, c) All other obvious causes of memory decline, except normal aging, have been ruled out (in other words, the memory decline cannot be attributed to other causes such as a recent heart attack or head injury, depression, adverse reactions to medication, Alzheimer's disease, etc.).

10084] "Age-Related Cognitive Decline (ARCD)" refers to declines in memory and cognitive abilities that are a normal consequence of aging in humans (e.g., Craik & Salthouse, 1992). This is also true in virtually all mammalian species. Age- Associated Memory Impairment refers to older persons with objective memory declines relative to their younger years, but cognitive functioning that is normal relative to their age peers (Crook et al., 1986). Age-Consistent Memory Decline, is a less pejorative label which emphasizes that these are normal developmental changes (Crook, 1993; Larrabee, 1996), are not pathophysiological (Smith et al., 1991), and rarely progress to overt dementia (Youngjohn & Crook, 1993). The DSM-IV (1994) has codified the diagnostic classification of ARCD.

10085] Alzheimer's disease (AD) is characterized by memory deficits in its earl - phase. Later symptoms include impaired judgment, disorientation, confusion, behavior changes, trouble speaking, and motor deficits. Histologically, AD is characterized by beta-amyloid plaques and tangles of protein tau.

[0086] Vascular dementia is caused by strokes. Symptoms overlap with those of AD, but without the focus on memory impairment. [0087] Dementia with Lewy bodies is characterized by abnormal deposits of alpha-synuciein that form inside neurons in the brain. Cognitive impairment may be similar to AD, including impairments in memory and judgment and behavior changes. [0088] Frontotemporal dementia is characterized by gliosis, neuronal loss, superficial spongiform degeneration in the frontal cortex and/or anterior temporal lobes, and Picks' bodies. Symptoms include changes in personality and behavior, including a decline in social skills and language expression/comprehension. f 0089] "Post traumatic stress disorder (PTSD)" refers to an anxiety disorder characterized by an immediate or delayed response to a catastrophic event, characterized by re-experiencing the trauma, psychic numbing or avoidance of stimuli associated with the trauma, and increased arousal. Re -experiencing phenomena include intrusive memories, flashbacks, nightmares, and psychological or physiological distress in response to trauma reminders. Such responses produce anxiety and can have significant impact, both chronic and acute, on a patient's quality of life and physical and emotional health, PTSD is also associated with impaired cognitive performance, and older individuals with PTSD have greater decline in cognitive performance relative to control patients,

10090] "Schizophrenia" refers to a chronic debilitating disorder, characterized by a spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations (e.g., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in patients,

[0091 ] "Amyotrophic lateral sclerosis," also known as ALS, refers to a progressive, fatal, neurodegenerative disease characterized by a degeneration of motor neurons, the nerve cells in the central nervous system that control voluntar muscle movement. ALS is also characterized by neuronal degeneration in the entorhinal cortex and hippocampus, memory deficits, and neuronal

hyperexcitability in different brain areas such as the cortex. f 0092] "Cancer therapy-related cognitive impairment" refers to cognitive impairment that develops in subjects that are treated with cancer therapies such as chemotherapy and radiation. Cytotoxicity and other adverse side-effects on the brain of cancer therapies result in cognitive impairment in such functions as memory, learning and attention.

[0093] "Treating" a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results. Beneficial or desired clinical results include, but are not limited to, improving cognitive function, delaying or slowing the progression of cogniti ve impairment, reducing the rate of decline of cognitive function, preventing or slowing the progression of the disease or disorder, or alleviation, amelioration, or slowing the progression, of one or more symptoms associated with CNS disorders with cognitive impairment, such as age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI, dementia, Alzheimer's Disease (AD), prodromal AD, PTSD, schizophrenia, amyotrophic lateral sclerosis (ALS) or cancer therapy-related cognitive

impairment. Treating age-related cognitive impairment further comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI, ARCD and AAMI) into dementia (e.g. , AD).

[0094J "Treating cognitive impairment" refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. Treatment of cognitive impairment in humans may improve cognitive function to any detectable degree, but is preferably improved sufficiently to allow the impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject. In some cases, "treating cognitive impairment" refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject's performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. In some cases, "treating cognitive impairment" in a subject affecting by age-related cognitive impairment refers to takings steps to improve cognitive function in the subject so that the subject's cognitive function, after treatment of cognitive impairment, more closely resembles the function of an age- matched normal, unimpaired subject, or the function of a young adult subject. In some cases, "treating cognitive impairment" in a subject refers to taking steps to delay or slow the progression of cognitive impairment in a subject with cognitive impairment. In some cases, "treating cognitive impairment" in a subject refers to taking steps to reduce the rate of decline of cognitive function in a subject with cognitive impairment.

10095] "Administering" or "administration of a substance, a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art. For example, a compound or an agent can be administered, intravenous iy, arteriaily, intradermally, intramuscularly, intraperitonealy, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasal!}' (by inhalation), intraspinaliy, intracerebral!y, and transdermal!y (by absorbtion, e.g., through a skin duct). A compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slo w or controlled release of the compound or agent. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is

administering the drag to the patient. [0096] Appropriate methods of administering a substance, a compound or an agent to a subject will also depend, for example, on the age of the subject, whether the subject is active or inactive at the time of administering, whether the subject is cognitively impaired at the time of administering, the extent of the impairment, and the chemical and biological properties of the compound or agent (e.g. solubility, digestibility, bioavailability, stability and toxicity). In some embodiments, a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously, e.g., to a subject by injection. In some embodiments, the orally administered compound or agent is in an extended release or slow release

formulation, or administered using a device for such slow or extended release.

[0097] As used herein, administration of an SV2A inhibitor or a

pharmaceutical ly acceptable salt, hydrate, solvate, or polymorph thereof and valproate or an analog, derivative or pharmaceutically acceptable salt thereof "in combination" or "together" includes simultaneous administration and/or

administration at different times, such as sequential administration, it also includes administration in a single formulation or in separate formulation packaged

together,

[0098] The term "simultaneous administration," as used herein, means that the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph and valproate or its analog, derivative or pharmaceutically acceptable salt, are administered with a time separation of no more than about 15 minutes, and in some embodiments no more than about 10 minutes. When the drags are administered simultaneously, the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph and valproate or its analog, derivative or pharmaceutically acceptable salt,, may be contained in the same dosage (e.g., a unit dosage form comprising both the SV2A inhibitor and the valproate) or in discrete dosages (e.g., the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph, is contained in one dosage form and the valproate or its analog, derivative or pharmaceutically acceptable salt, is contained in another dosage form). [0099J The term "sequential administration" as used herein means that the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph are valproate or its analog, derivative or pharmaceutically acceptable salt, are

administered with a time separation of more than about 15 minutes, and in some embodiments more than about one hour, or up to 12 hours. Either the SV2A inhibitor or the valproate may be administered first. For sequential administration, he SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate, or polymorph, and valproate or its analog, derivative or pharmaceutically acceptable salt, may be contained in discrete dosage forms, optionally contained in the same container or package.

[0100] A "therapeutically effective amount" of a drug or agent is an amount of a drug or an agent that, when administered to a subject will have the intended therapeutic effect, e.g. improving cognitive function, or delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function in a subject, e.g., a patient having a CNS disorder with cognitive

impairment. The full therapeutic effect does not necessarily occur by

administration of one dose, and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations. The precise effective amount needed for a subject will depend upon, for example, the subject's size, health and age, the nature and extent of the cognitive impairment, and the therapeutics or combination of therapeutics selected for administra tion, and the mode of administration. The skilled worker can readily determine the effective amount for a given situation by routine

experimentation.

[0101] "Subtherapeutic amount" refers to an amount administered of an agent or compound of the invention that is less than the therapeutic amount, that is, less than the amount normally used when said agent or compound is administered alone (i.e., individually and in the absence of other therapeutic agents or compounds) to treat disorders involving cognitive dysfunction.

[0102] "Analog" is used herein to refer to a compound which functionally resembles another chemical entity, but does not share the identical chemical structure. For example, an analog is sufficiently similar to a base or parent compound such that it can substitute for the base compound in therapeutic applications, despite minor structural differences.

[0103] "Derivative" is used herein to refer to the chemical modification of a compound. Chemical modifications of a compound can include, for example, replacement of hydrogen by an alkyl, acyl, or amino group. Many other

modifications are also possible.

[0104] The term "prodrug" is art-recognized and is intended to encompass compounds or agents which, under physiological conditions, are converted into an 8V2A inhibitor or valproate. A common method for making a prodrug is to select moieties which are hydrolyzed or metabolized under physiological conditions to provide the desired compound or agent. In other embodiments, the prodrug is con verted by an enzymatic activity of the host animal to an inhibitor of SV2A or valproate.

[0105] "Pharmaceutically acceptable salts" is used herein to refer to an agent or a compound according to the invention that is a therapeutically active, non-toxic base and acid salt form of the compounds. The acid addition salt form of a compound that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrohrornic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hvdroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, metiianesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclic, salicylic, p- aminosalicylic, pamoic and the like. See, e.g., WO 01/062726.

Description of Methods of the Invention

[0106] The methods of this invention comprise administration of an SV2A inhibitor or a pharmaceutically acceptable salt thereof. The methods of this invention further comprise administration of an SV2A inhibitor or a pharmaceutically acceptable salt thereof in combination with administration of valproate or a pharmaceutically

acceptable salt thereof. The agents or compounds of the SV2A inhibitor or the valproate and their pharmaceutically acceptable salts also include hydrates, solvates, polymorphs, and prodrugs of those agents, compounds, and salts. Methods of Assessing Cognitive Impairment

[0107] Animal models serve as an important resource for developing and evaluating treatments for CNS disorders with cognitive impairment. Features that characterize cognitive impairment in animal models typically extend to cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans. The extent of cognitive impairment in an animal model for a CNS disorder, and the efficac of a method of treatment for said CNS disorder may be tested and confirmed with the use of a variety of cogniti ve tests. |0108] A Radial Arm Maze (RAM) behavioral task is one example of a cognitive test, specifically testing spacial memory (Chappeli et al. Neuropharmacology 37: 481-487, 1998). The RAM apparatus consists of, e.g., eight equidistantly spaced arms. A maze arm projects from each facet of a center platform. A food well is located at the distal end of each arm. Food is used as a reward. Blocks can be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus may also be provided. After habituation and training phases, spatial memory of the subjects may be tested in the RAM under control or test compound- treated conditions. As a part of the test, subjects are pretreated before trial s with a vehicle control or one of a range of dosages of the test compound. At the beginning of each trial, a subset of the arms of the eight-arm maze is blocked. Subjects are allowed to obtain food on the unblocked arms to which access is permitted during this initial "information phase" of the trial. Subjects are then removed from the maze for a delay period, e.g., a 60 second delay, a 15 minute delay, a one-hour delay, a two-hour delay, a six hour delay, a 24 hour delay, or longer) between the information phase and the subsequent "retention test," during which the barriers on the maze are removed, thus allowing access to all eight arms. After the delay period, subjects are placed back onto the center platform (with the barriers to the previously blocked arms removed) and allowed to obtain the remaining food rewards during this retention test phase of the trial. The identity and configuration of the blocked arms vary across trials. The number of "errors" the subjects make during the retention test phase is tracked. An error occurs in the trial if the subjects entered an arm from which food had already been retrieved in the pre-deiay component of the trial, or if it re- visits an arm in the post-delay session that had already been visited. A fewer number of errors would indicate better spatial memory. The number of errors made by the test subject, under various test compound treatment regimes, can then be compared for efficacy of the test compound in treating CNS disorders with cognitive impairment,

[0109] Another cognitive test that may be used to assess the effects of a test compound on the cognitive impairment of a CNS disorder model animal is the Morris water maze. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampal-dependent (de Hoz et al, Eur. J. Neurosci., 22:745-54, 2005; Steele and Morris, Hippocampus 9: 118-36, 1999). The subject is trained to locate a submerged escape platform hidden underneath the surface of the pool. During the training trial, a subject is released in the maze (pool) from random starting positions around the perimeter of the pool. The starting position varies from trial to trial. If the subject does not locate the escape platform within a set time, the experimenter guides and places the subject on the platform to "teach" the location of the platform. After a delay period following the last training trial, a retention test in the absence of the escape platform is given to assess spatial memory. The subject's level of preference for the location of the (now absent) escape platform, as measured by, e.g., the time spent in that location or the number of crossings of that location made by the mouse, indicates better spatial memory, i.e., treatment of cognitive impairment. The preference for the location of the escape platform under different treatment conditions, can then be compared for efficacy of the test compound in treating CNS disorders with cognitive impairment,

[0110 j There are various tests known in the art for assessing cognitive function in humans, for example and without limitation, the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test ( Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest: the Visual Reproduction subtest of the Wechsler Memory Scale- Revised (WMS-R) ( Wechsler, 1997); or the Benton Visual Retention Test. See Folstein et al., J Psychiatric Res 12: 189-98, (1975); Robbins et al., Dementia 5: 266-81 , (1994); Rey, L'examen clinique en psychologie, (1964); luger et al., J Geriair Psychiatry Neurol 12:168-79, (1999); Marquis et al, 2002 and Masur et al,, 1994. Another example of a cognitive test in humans is the explicit 3- a!ternative forced choice task. In this test, subjects are presented with color photographs of common objects consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. The second of the pair of similar objects is referred to as the "lure". These image pairs are fully randomized and presented indi vidually as a series of images. Subjects are instructed to make a judgment as to whether the objects seen are new, old or similar. A "similar" response to the presentation of a lure stimulus indicates successful memory retrieval by the subject. By contrast, calling the lure stimulus "old" or "new" indicates that correct memory retrieval did not occur.

[0111] In addition to assessing cognitive performance, the progression of age- related cognitive impairment and dementia, as well as the conversion of age- related cognitive impairment into dementia, may be monitored by assessing surrogate changes in the brain of the subject. Surrogate changes include, without limitation, changes in regional brain volumes, perforant path degradation, and changes seen in brain function through resting state fMRI (R-fMRj ) and fhiorodeoxyghicose positron emission tomography (FDG-PET). Examples of regional brain volumes useful in monitoring the progression of age-related cognitive impairment and dementia include reduction of hippocampal volume and reduction in volume or thickness of entorhinal cortex. These volumes may be measured in a subject by, for example, MR1. Aisen et al., Alzheimer's &

Dementia 6:239-246 (2010). Perforant path degradation has been shown to be linked to age, as well as reduced cognitive function. For example, older adults with more perforant path degradation tend to perform worse in hippocampus- dependent memor tests. Perforant path degradation may be monitored in subjects through ultrahigh-resolution diffusion tensor imaging (DTI). Yassa et al.. PNAS 107: 12687-12691 (2010). Resting-state f I ( R-fM R l ) involves imaging the brain during rest, and recording large-amplitude spontaneous low- frequency (<0.1 Hz) fluctuations in the fMRI signal that are temporally correlated across functionally related areas. Seed-based functional connectivity, independent component analyses, and/or frequency-domain analyses of the signals are used to reveal functional connectivity between brain areas, particularly those areas whose connectivity increase or decrease with age, as well as the extent of cognitive impairment and/or dementia. FDG-PET uses the uptake of FDG as a measure of regional metabolic activity in the brain. Decline of FDG uptake in regions such as the posterior cingulated cortex, temporoparietal cortex, and prefrontal association cortex has been shown to relate to the extent of cognitive decline and dementia. Aisen et al., Alzheimer's & Dementia 6:239-246 (2010), Herholz et al.,

Neurolmage 17:302-316 (2002). Age-Related Cognitive Impairment

[0112] This invention provides methods and compositions for treating age-related cognitive impairment or the risk thereof using an S V2A inhibitor or a

pharmaceutically acceptable salt, hydrate, solvate or polymorth thereof, alone or in combination with valproate or an alalog, derivative or pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises improving cognitive function in patients with age-related cognitive impairment. In certain

embodiments, treatment comprises slowing or delaying the progression of age- related cognitive impairment. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with age-related cognitive impairment. In certain embodiments, treatment comprises preventing or slowing the progression, of age-related cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with age-related cognitive impairment. In certain embodiments, treatment of age-related cogniti ve impairment comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI, A RCD and AAMI) into dementia (e.g., AD). The methods and compositions may be used for human patients in clinical applications in the treating age-related cognitive impairment in conditions such as MCI, ARCD and AAMI or for the risk thereof. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.

[0113] In some embodiments, a subject to be treated by the methods and compositions of this invention exhibits age-related cognitive impairment or is at risk of such impairment. In some embodiments, the age-related cognitive impairment includes, without limitation, Age-Associated Memory Impairment (AAMI), Mild Cognitive impairment (MCI) and Age-related Cognitive Decline (ARCD).

[0114] Animal models serve as an important resource for developing and evaluating treatments for such age-related cognitive impairments. Features that characterize age-related cognitive impairment in animal models typically extend to age-related cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans.

[0115] Various animal models of age-related cognitive impairment are known in the art. For example, extensive behavioral characterization has identified a naturally occurring form of cognitive impairment in an outbred strain of aged Long-Evans rats (Charles River Laboratories; Gallagher et ai, Behav. Neurosci. 107:618-626, (1993 )). In a behavioral assessment with the Morris Water Maze (MWM), rats learn and remember the location of an escape platform guided by a configuration of spatial cues surrounding the maze. The cognitive basis of performance is tested in probe trials using measures of the animal's spatial bias in searching for the location of the escape platform. Aged rats in the study population have no difficulty swimming to a visible platform, but an age-dependent impairment is detected when the platform is camouflaged, requiring the use of spatial information. Performance for individual aged rats in the outbred Long- Evans strain varies greatly. For example, a proportion of those rats perform on a par with young adults. However, approximately 40-50% fall outside the range of young performance. This variability among aged rats reflects reliable individual differences. Thus, within the aged population some animals are cognitively impaired and designated aged-impaired (A!) and other animals are not impaired and are designated aged-unimpaired (AU). See, e.g., Colombo et al, Proc. Natl. Acad. Set. 94: 14195-14199, (1997); Gallagher and Burwell, Neurobiol. Aging 10: 691-708, (1989); Gallagher et al Behav. Neurosci. 107:618-626, (1993); Rapp and Gallagher, Proc. Natl. Acad. Sci. 93: 9926-9930, (1996); Nicolle et ai,

Neuroscience 74: 74 1 -756. (1996); Nicoile et al., J. Neurosci. 19: 9604-9610, (1999); International Patent Publication WO2007/019312 and International Patent Publication WO 2004/048551 . Such an animal model of age-related cognitive impairment may be used to assay the effecti veness of the methods and

compositions this invention in treating age-related cognitive impairment.

|0116] The efficacy of the methods and compositions of this invention in treating age-related cognitive impairment may be assessed using a variety of cognitive tests, including the Morris water maze and the radial arm maze, as discussed above.

Dementia

[0117] This invention also provides methods and compositions for treating dementia using an 8V2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorth thereof, alone or in combination with valproate or an alalog, derivative or pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises improving cognitive function in patients with dementia. In certain embodiments, treatment comprises slowing or delaying the progression of dementia. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with dementia. In certain embodiments, treatment comprises preventing or slowing the progression, of dementia. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with dementia. In certain embodiments, the symptom to be treated is cognitive impairment, in certain embodiments, the dementia is Alzheimer's disease (AD), vascular dementia, dementia with Lewy bodies, or frontotemporal dementia. The methods and compositions may be used for human patients in clinical applications in trea ting dementia. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications. jOl 18] Animal models serve as an important resource for developing and evaluating treatments for dementia. Features that characterize dementia in animal models typically extend to dementia in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of dementia are known in the art, such as the PD APP, Tg2576, APP23, TgCRNDS, J20, hPS2 Tg, and APP + PS1 transgenic mice. Sankaranarayanan, Curr. Top. Medicinal Chem. 6: 609-627, 2006; Kobayashi et al. Genes Brain Behav. 4: 173- 196. 2005; Ashe and Zahns, Neuron. 66: 631-45, 2010. Such animal models of dementia may be used to assay the effectiveness of the methods and compositions of this invention of the invention in treating dementia.

|0119] The efficacy of the methods and compositions of this in vention in treating dementia, or cognitive impairment associated with dementia, may be assessed in animals models of dementia, as well as human subjects with dementia, using a variety of cognitive tests known in the art, as discussed above.

Post Traumatic Stress Disorder

[0120] This invention also provides methods and compositions for treating post traumatic stress disorder (PTSD) using an SV2 A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymoxth thereof, alone or in combination with valproate or an aialog, derivative or pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises improving cognitive function in patients with PTSD. In certain embodiments, treatment comprises slowing or delaying the progression of PTSD. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with PTSD. In certain

embodiments, treatment comprises preventing or slowing the progression, of PTSD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with PTSD. In certain embodiments, the symptom to be treated is cognitive impairment. The methods and compositions may be used for human patients in clinical applications in treating PTSD. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.

|0121] Patients with PTSD (and, to a lesser degree trauma-exposed patients without PTSD) have smaller hippocampal volumes (Woon et ai. , Prog. Neuro- Psychopharm, & Biological Psych. 34, 1 181-1188; Wang et al., Arch, Gen.

Psychiatry 67:296-303, 2010). PTSD is also associated with impaired cognitive performance. Older individuals with PTSD have greater declines in cognitive performance relative to control patients (Yehuda et al., Bio. Psych. 60: 714-721, 2006) and have a greater likelihood of developing dementia (Yaffe et al., Arch. Gen. Psych. 678: 608-613, 2010).

[0122 J Animal models serve as an important resource for developing and evaluating treatments for PTSD. Features that characterize PTSD in animal models typically extend to PTSD in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of PTSD are known in the art.

[0123] One rat model of PTSD is Time-dependent sensitization (TDS). TDS involves exposure of the animal to a severely stressful event followed by a situational reminder of the prior stress. The following is an example of TDS. Rats are placed in a restrainer, then placed in a swim tank and made to swim for a period of time, e.g., 20 mill. Following this, each rat is then immediately exposed to a gaseous anesthetic until loss of consciousness, and finally dried. The animals are left undisturbed for a number of days, e.g. , one week. The rats are then exposed to a "restress" session consisting of an initial stressor, e.g., a swimming session in the swim tank (Liberzon et al., Psychomuroendocrinology 22: 443-453, 1997; Harvery et al., Psychopharmacology 175:494-502, 2004). TDS results in an enhancement of the acoustic startle response (ASR) in the rat, which is comparable to the exaggerated acoustic startle that is a prominent symptom of PTSD (Khan and Liberzon, Psychopharmacology 172: 225-229, 2004). Such animal models of PTSD may be used to assay the effecti veness of the methods and compositions of this invention of the invention in treating PTSD. [0124] The efficacy of the methods and compositions of this invention in treating PTSD, or cognitive impairment associated with PTSD, may also be assessed in animals models of PTSD, as well as human subjects with PTSD, using a variety of cognitive tests known in the art, as discussed above.

[0125] This invention additionally provides methods and compositions for treating schizophrenia using an SV2A inhibitor or a pharmaceutically acceptable sail, hydrate, solvate or polymorih thereof alone or in combination with valproate or an alalog, derivative or pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises improving cognitive function in patients with schizophrenia. In certain embodiments, treatment comprises slowing or delaying the progression of schizophrenia. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with schizophrenia. In certain embodiments, treatment comprises preventing or slowing the progression, of schizophrenia. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with schizophrenia. In certain embodiments, the symptom to be treated is cognitive impairment. The methods and compositions may be used for human patients in clinical applications in treating schizophrenia. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.

[0126] Cognitive impairments are also associated with schizophrenia. They precede the onset of psychosis and are present in non-affected relatives. The cognitive impairments associated with schizophrenia constitute a good predictor for functional outcome and are a core feature of the disorder. Cognitive features in schizophrenia reflect dysfunction in frontal cortical and hippocampal circuits. Patients with schizophrenia also present hippocampal pathologies such as reductions in hippocampal volume, reductions in neuronal size and dysfunctional hyperactivity. An imbalance in excitation and inhibition in these brain regions has also been documented in schizophrenic patients suggesting that drugs targeting inhibitory mechanisms could be therapeutic. See, e.g., Guidotti et a!,, Psychopharmacology 180: 191-205, 2005: Zierhut, Psych. Res. Neuroimag.

183: 187-194, 2010; Wood et ah, Neurolmage 52:62-63, 2010; Vinkers et ah, Expert Opin. Investig. Drugs 19: 1217-1233, 2009; Young et al, Pharmacol. Ther. 122: 150-202, 2009. [0127] Animal models serve as an important resource for developing and evaluating treatments for schizophrenia. Features that characterize schizophrenia in animal models typically extend to schizophrenia in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. V arious animal models of schizophrenia are known in the art. [0128] One animal model of schizophrenia is protracted treatment with methionine. Methionine-treated mice exhibit deficient expression of GAD67 in frontal cortex and hippocampus, similar to those reported in the brain of postmortem schizophrenia patients. They also exhibit prepulse inhibition of startle and social interaction deficits (Tremoniizzo et al, PNAS, 99: 17095-17100, 2002). Another animal model of schizophrenia is methylaoxymethanol acetate

(MAM)-treatment in rats. Pregnant female rats are administered MAM (20 mg/kg, intraperitoneal) on gestational day 17. MAM-treatment recapitulate a

pathodevelopmental process to schizophrenia-like phenotypes in the offspring, including anatomical changes, behavioral deficits and altered neuronal information processing. More specifically, MAM-treated rats display a decreased density of parvalbumin-positive GABAergic interneurons in portions of the prefrontal cortex and hippocampus. In behavioral tests, MAM-treated rats display reduced latent inhibition. Latent inhibition is a behavioral phenomenon where there is reduced learning about a stimulus to which there has been prior exposure with any consequence. This tendency to disregard previously benign stimuli, and reduce the formation of association with such stimuli is believed to prevent sensory overload. Low latent inhibition is indicative of psychosis. Latent inhibition may be tested in rats in the following manner. Rats are divided into two groups. One group is pre- exposed to a tone over multiple trials. The other group has no tone presentation. Both groups are then exposed to an auditory fear conditioning procedure, in which the same tone is presented concurrently with a noxious stimulus, e.g. an electric shock to the foot. Subsequently, both groups are presented with the tone, and the rats' change in locomotor activity during tone presentation is monitored. After the fear conditioning the rats respond to the tone presentation by strongly reducing locomotor activity. However, the group that has been exposed to the tone before the conditioning period displays robust latent inhibition: the suppression of locomotor activity in response to tone presentation is reduced. MAM -treated rats, by contrast show impaired latent inhibition. That is, exposure to the tone previous to the fear conditioning procedure has no significant effect in suppressing the fear conditioning, (see Lodge et at, J. Neurosci., 29:2344-2354, 2009) Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia.

|0129] The efficacy of the methods and compositions of this invention in treating schizophrenia, or cognitive impairment associated with schizophrenia, may also be assessed in animal models of schizophrenia, as wel l as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as discussed above.

Amyotrophic Lateral Sclerosis (ALS)

[0130] This invention additionally provides methods and compositions for treating ALS using an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorth thereof alone or in combination with valproate or a an ala!og, derivative or pharmaceutically acceptable salt thereof. In certain embodiments, treatment comprises impro ving cognitive function in patients with ALS. In certain embodiments, treatment comprises slowing or delaying the progression of ALS. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with ALS. In certain embodiments, treatment comprises preventing or slowing the progression, of ALS. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with ALS. In certain

embodiments, the symptom to be treated is cognitive impairment. The methods and compositions may be used for human patients in clinical applications in treating ALS. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.

|0131] In addition to the degeneration of motor neurons, ALS is characterized by neuronal degeneration in the entorhinal cortex and hippocampus, memory deficits, and neuronal hyperexcitability in different brain areas such as the cortex.

[0132] The efficacy of the methods and compositions of this invention in treating ALS, or cognitive impairment associated with ALS, may also be assessed in animal model s of ALS, as well as human subjects wit ALS, using a variety of cognitive tests known in the art, as discussed above. Casicer therapy-related cognitive impairment

|0133] This invention additionally provides methods and compositions for treating cancer therapy-related cognitive impairment using an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorth thereof, alone or in combination with valproate or an alalog, derivative or pharmaceutically acceptable salt thereof, In certain embodiments, treatment comprises improving cognitive function in patients with cancer therapy-related cognitive impairment. In certain embodiments, treatment comprises slowing or delaying the progression of cancer therapy-related cognitive impairment. In certain embodiments, treatment comprises reducing the rate of decline of cognitive function associated with cancer therapy-related cognitive impairment. In certain embodiments, treatment comprises preventing or slowing the progression, of cancer therapy-related cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with cancer therapy-related cognitive impairment, The methods and compositions may be used for human patients in clinical applications in treating cancer therapy-related cognitive impairment, The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those

applications. [0134] Therapies that are used in cancer treatment, including chemotherapy, radiation, or combinations thereof, can cause cognitive impairment in patients, in such functions as memoiy, learning and attention. Cytotoxicity and other adverse side-effects on the brain of cancer therapies are the basis for this form of cogniti ve impairment, which can persist for decades. (Dietrich et al, Oncologist 13: 1285-95, 2008; Soussain et al, Lancet 374: 1639- 1 , 2009).

[0135] Cognitive impairment following cancer therapies reflects dysfunction in frontal cortical and hippocampal circuits that are essential for normal cognition. In animal models, exposure to either chemotherapy or radiation adversely affects performance on tests of cognition specifically dependent on these brain systems, especially the hippocampus (Kim et al., J. Radiat. Res. 49:517-526, 2008: Yang et al., Neurobiol. Learning and Mem. 93:487-494, 2010). Thus, drugs targeting these cortical and hippocampal systems could be neuroprotective in patients receiving cancer therapies and efficacious in treating symptoms of cognitive impairment that may last beyond the interventions used as cancer therapies.

[0136] Animal models serve as an important resource for developing and evaluating treatments for cancer therapy-related cognitive impairment. Features that characterize cancer therapy-related cognitive impairment in animal models typically extend to cancer therapy-related cognitive impairment in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of cancer therapy-related cognitive impairment are known in the art.

[0137] Examples of animal models of cancer therapy-related cognitive impairment include treating animals with anti-neoplastic agents such as cyclophosphamide (CYP) or with radiation, e.g., b0 Co gamma-rays. (Kim et al., J. Radiat. Res. 49:517-526, 2008; Yang et al., Neurobiol. Learning arid Mem.

93:487-494, 2010). The cognitive function of animal models of cancer therapy- related cognitive impairment may then be tested with cognitive tests to assay the effectiveness of the methods and compositions of the invention in treating cancer therapy-related cognitive impairment. The efficacy of the methods and

compositions of this invention in treating cancer therapy-related cognitive impairment, as well as human subjects with cancer therapy-related cognitive impairment, using a variety of cognitive tests known in the art, as discussed abo ve.

SV2A Inhibitor

| 138] "Synaptic vesicle protein-2 (SV2)" is a family of synaptic vesicle proteins, which consists of three members, designated SV2A, SV2B, and SV2C. SV2A is the most widely distributed family member, being expressed ubiquitously in the brain. The proteins are integral membrane proteins and have a low-level homology (20-30%) to the twelve transmembrane family of bacterial and fungal transporter proteins that transport sugar, citrate, and xenobiotics (Bajjaiieh et al., Science. 257: 1271-1273. (1992)). SV2 family proteins are present in the brain and endocrine cells, and further are present in all synaptic and endocrine vesicles. SV2 proteins are reported to play a role in normal synaptic function, and functions in a maturation step of primed vesicles that converts the vesicles into a Ca( 2+ )~ and synaptotagmin-responsive state (Sudhof et al, 2009). Functionally, SV2 proteins are reported to enhance synaptic currents and increase the probability of transmitter release by maintaining the size of the readily re leasable pool of vesicles (Custer et al, 2006).

[0139] "SV2A inhibitor" refers to any agent, substance or compound that binds to SV2A and reduces synaptic function by reducing pre-synaptic vesicle release (See, e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006; Custer et al, 2006; Smedt et al, 2007; Yang et al, 2007; Meehan,

"Levetiracetam has an activity-dependent effect on inhibitory transmission," Epilepsia, 2012 J an 31; and Example 8 of WO 2001 /62726, all of which are specifically incorporated herein by reference.) A substance, or a compound or an agent is an SV2A inhibitor even if it does not itself bind to SV2A, as long as it causes, or affects the ability of, another compound or agent to bind SV2A or reduce synaptic function by reducing pre-synaptic vesicle release. SV2A inhibitors, as used herein, include pharmaceutically acceptable salts of the inhibitors thereof. They also include hydrates, polymorphs, prodrugs, salts, and sol vates of these inhibitors. [0140] Among the SV2A inhibitors or pharmaceutically acceptable salts, hydrates, solvates and polymorphs thereof that are useful in the methods and compositions of this invention are those disclosed, for example, United States (U.S.) Patent Application 12/580,464, International Patent Application

PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent

Application 61/152,631 , and U.S. Patent Application 61/175,536. However, any SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be used in the methods and compositions of the invention. In some embodiments, the SV2A inhibitor is selected from the group of SV2A. inhibitors referred to in International Patent Applications WO2010/144712;

WO201.0/002869; WO2008/132139; WO2007/065595; WO2006/128693;

WO2006/128692; WO2005/054188; WO2004/087658; WO2002/094787;

WO2001/062726; U.S. Patents 7,465,549; 7,244,747; 5,334,720; 4,696,943;

4,696,942; U.S. Patent Application Publication Numbers 20090312333;

20090018148; 20080081832; 2006258704; and UK Patent Numbers 1 ,039,1 13; and 1,309,692 or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs. Other SV2A inhibitors may also be used in this invention.

Appl icants also refer to methods of preparing these compounds found in the documents cited above. Other synthetic methods may also be used. These methods are well known to those skilled in the ail.

10141] In some embodiments of this in vention, the SV2A inhibitor is selected from the group consisting of levetiracetam, brivaracetam, and seletracetam or derivatives or analogs or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, or prodrugs thereof. [0142] In some embodiments of this invention, the SV2A inhibitor is levetiracetam or salts, solvates, hydrates, polymorphs or prodrugs thereof.

Levetiracetam refers to the International Union of Pure and Applied Chemistry (IUPAC) name of the compound (2S)-2-(2-oxopyrrolidin-l-yl) butanamide). Levetiracetam is a widely used antiepileptic drug. Levetiracetam binds to a specific site in the CNS: the synaptic vesicle protein 2A (SV2A) (See. e.g., Noyer et al. 1995; Fuks et al. 2003; Lynch et al. 2004; Gillard et al. 2006) and has further been shown to directly inhibit synaptic activity and neurotransmission by inhibiting presynaptic neurotransmitter release (Yang et a!., 2007).

|Θ143] Among the SV2A inhibitors useful for the methods and compositions of this invention are the following: i) International Patent Application WO 2001/062726:

A compound having the formula I or a pharmaceutically acceptable salt thereof,

wherein X is-C XR or-CAOR 7 or-CA^R 8 or CN ;

A 1 and A 2 are independently oxygen, sulfur or~NR ' ';

R 1 is hydrogen, alkyl, aryl or-CH 2 -R la wherein R ia is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;

R 2 , R* and R are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative;

R 2 , R Jd and R ½ are the same or different and each is independently hydrogen, halogen, alkyl, alkenyl, alkynyl or aryl;

R 3 , R 6 , R' and R 9 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or a oxy derivative; and R l is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative; with the provisos that at least one of as IV, R\ R 4 , R 2i! , R ' a and R 4a is other than hydrogen; and that when the compound is a mixture of all possible isomers, X is-CONR 3 R 6 , A 2 is oxygen and R 1 is hydrogen, methyl, ethyl or propyl then substitution on the pyrollidine ring is other than mono-, di-, or tri- methyi or mono-ethyl; and that when R 1 , R", R 4 , R 2d , R 3a and R 4d are each hydrogen, A ' " is oxygen and X is CONR 5 R then R J is different from carboxy, ester, amido, substituted oxo-pyrroiidine, hydroxy, oxy derivative, amino, amino derivatives, methyl, naphthyl, phenyl optionally substituted by oxy derivatives or in the para position by an halogen atom.

In the definitions set forth below, unless otherwise stated, R" and R 12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfmyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.

The term "oxy derivative", as used herein is defined as including -0-R 1 ' groups wherein R 11 is as defined above except for "oxy derivative". Non- limiting examples are alkoxy, alkenyl oxy, alkynyloxy, acyloxy, oxyester, oxyamido, aikyisulfonyloxy, alkylsulfinyloxy, arylsuifonyloxy,

arylsuifmyloxy, aryioxy, aralkoxy or heterocyclooxy such as pentyloxy, allyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methyl enedioxy, carbonate.

The term "thio derivative" as used herein, is defined as including-S-R 11 groups wherein R 1 1 is as defined above except for "thio derivative". Non- limiting examples are alkylthio, alkenylihio, alkynylthio and arylthio. The term "amino derivative" as used herein, is defined as including-NHR 11 or -NR^R 1 " groups wherein R J 1 and R 12 are as defined above. Non-limiting examples are mono- or di-alkyl-, aikenyi-, alkynyi- and aryiamino or mixed amino.

The term "acyl derivative" as used herein, represents a radical derived from carboxvlic acid and thus is defined as including groups of the formula R J 1 -CO-, wherein R 11 is as defined above and may also be hydrogen. Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl,

heptanedioyl, cyciohexanecarbonvl, crotonovl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, mcotinoyl, 4-carboxybutanoyl, oxalyS, ethoxalyl, eysteinyS, oxamoyl.

The term "suifonyi derivative" as used herein, is defined as including a group of the formula -SO 2 -R" , wherein R f 1 is as defined above except for "suifonyi derivative". Non-limiting examples are alkylsulfonyl,

alkenylsuifonyl, alkynylsuifonyl and arylsulfonyl.

The term "suffinyl derivative" as used herein, is defined as including a group of the formul a -SO-R 11 , wherein R 11 is as defined above except for "sulfinyl derivative". Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, aikynylsulfinyl and aryl sulfinyl.

The term "a!ky!", as used herein, is defined as including saturated, monovalent hydrocarbon radicals ha ving straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1 -6 carbon atoms for non-cyclic alkyl and 3-6 carbon atoms for cycioalkyl (in these two preferred cases, unless otherwise specified, "lower alkyl"). Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cy ano, thiocyanato, acyl, acyloxy, suifonyi derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cycioalkyl, sulfonic acid, sulfonamide, thio derivative, oxyester, oxyamido, heterocycle, vinyl, Cl-5-alkoxy, C6-10- ai loxy and C6-10-aryl. Preferred alky! groups are methyl, ethyl, propyl, isopropyl, butyl, iso or ter- butyl, and 2,2,2-trimethyiethyl each optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro and cyano, such as trifiuoromethyl, triehloromethyl, 2,2,2- trichloroethyl, l, l-dimethyl-2,2-dibromoethyl, 1,1-dimethy 1-2,2,2- trichloroethyl.

The term "alkenyl" as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond such as ethenyl (= vinyl), 1- methyl- l-etheiiyl, 2,2-dimethyl- 1 -ethenyl, 1- propenyl, 2-propenyl (= ally!), 1 -butenyl, 2-butenyl, 3-butenyl, 4-pentenyl, 1 - methyl-4-pentenyl, 3-methyl-l-pentenyi, l-hexenyl, 2-hexenyl, and the like and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl and heterocycle such as mono- and di-halo vinyl where halo is fluoro, chloro or bromo.

The term "alkynyl" as used herein, is defined as including a monovalent branched or unbranched hydrocarbon radical containing at least one carbon- carbon triple bond, for example ethynyl, 2-propynyl (= propargyl), and the like and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl and heterocycle, such as haloethynyl.

When present as bridging groups, aikyi, alkenyl and alkynyl represent straight- or branched chains, Cl-12, preferably Cl-4-alkylene or C2-12-, preferably C2-4-alkenylene or -alkynylene moieties respectively, Groups where branched derivatives are conventionally qualified by prefixes such as "n", "sec", "iso" and the like (e.g., "n-propyl", "sec-butyl") are in the n- form unless otherwise stated.

The term "aryl" as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6- 30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfinyl, alkylamino, earboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyi, alkylsulfinyl, alkylthio, oxyester, oxyamido, aryl, C I -6-alkoxy, C6~10-aryloxy, Cl-6-alkyl, Cl-6-haloalkyl. Aryl radicals are preferably monocyclic containing 6-10 carbon atoms, Preferred aryl groups are phenyl and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, nitro, amino, azido, Cl.-6-alkoxy, CI -6- alkylthio, CI -6 alkyl, Cl-6-haloalkyl and phenyl,

The term "halogen", as used herein, includes an atom of C , Br, F, I.

The term "hydroxy", as used herein, represents a group of the formula -OH.

The term "thiol", as used herein, represents a group of the formula -SFL

The term "cyano", as used herein, represents a group of the formula -CN.

The term "nitro", as used herein, represents a group of the formula -NO?.

The term "nitrooxy", as used herein, represents a group of the formula - ON0 2 ,

The term "amino", as used herein, represents a group of the formula -NH 2 .

The term "azido", as used herein, represents a group of the formula -N 3 .

The term "earboxy", as used herein, represents a group of the formula - COOH.

The term "sulfonic acid", as used herein, represents a group of the formula S0 3 H.

The term "sulfonamide", as used herein, represents a group of the formula - SO -Ni !;:. The term "ester", as used herein is defined as including a group of formula - COO-R 11 wherein R f 1 is as defined above except oxy derivative, thio derivative or amino derivative.

The term "ether" is defined as including a group selected from Cl-50- straight or branched alkyl, or C2-50- straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.

The term "amido" is defined as including a group of formula -CONH 2 or- CONHR 11 or -CONR n R 12 wherein R 11 and R 12 are as defined above.

The term "heterocycle", as used herein is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl. Non-limiting examples of aromatic

heterocycies are pyridyi, furyl, pyrrolyl, thienyi, isothiazolyl, imidazolyl, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolyl, isoquinoiyl, isobenzofuranyl, benzothienyl, pyrazolyl, indolyl, indolizinyl, purinyl, isoindolyl, carbazolyl, thiazolyl, 1 , 2, 4-thiadiazolyl, thieno (2,3 -b) furanyl, furopyranyl,

benzofuranyl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl,

benzothiazolyl, or benzoxazolyl, cimiolinyl, phthalazinyl, quinoxalmyl.

phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyS, phenothiazinyl, furazanyl, isochromanyl. indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5- azacytidinyi, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl,

pyrrolopyrimidinyl, and pyrazolopyrimidinyl optionally substituted by alkyl or as described above for the alkyl groups. Non-limiting examples of non aromatic heterocycies are tetrahydro furanyl, tetrahydropyranyl, piperidinyl, piperidyi, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yI, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) or the same which can optionally be substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. The term "heterocycle" also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a eyciohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1)heptanyl, 7- oxabicyclo (2,2.1) heptanyl, 8- azabieyclo (3.2, l )ocianyl.

In the above definitions it is to be understood that when a substituent such as R 2 , R 3 , R 4 , R 2a , R 3a , R 4a , R 5 , R 6 , R 7 , R 8 is attached to the rest of the molecule via a heteroatom or a carbonvl, a straight- or branched chain, C1-12-, preferably Cl-4-alkylene or C2-12, preferably C2-4-alkenylene or-alkynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule.

Preferred examples of X are -COO R' or -CONR 3 R 6 , wherein R " R 5 and R' are preferably hydrogen, Cl-4-alkyl, phenyl or alkylphenyl.

Preferably X is carboxy or -CONR 5 R 6 , wherein R 5 and R 6 are preferably hydrogen, Cl-4-alkyl, phenyl or alkylphenyl, especially ~CONH 2 .

Preferably A 1 and A? are each oxygen.

Preferably R 1 is hydrogen, alkyl, especially Cl-12 alkyl, particularly lower alkyl or aryl especially phenyl.

Examples of preferred R ] groups are methyl, ethyl, propyl, isopropyi, butyl, iso- or ter-butyl. 2,2,2-trimethylethyl each optionally attached via a methylene bridge or the same substituted by at least one halogen atom such as

trifluoromethyl, trichloromethyl, 2,2,2-trichloroethyl, 1,1-dimethy 1-2,2- dibromoethyl, l,l-dimethyl-2,2,2~trichloroethyl,

R 1 as ethyl is especially preferred. Preferably R z and R a are independently hydrogen, halogen or alkyl, especially lower alkyl.

Exampl es of preferred " and R 2a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2- trimethylethyl or the same substituted by at least one halogen atom such as trifluoromethyl, trichloromethyl, 2,2,2-trichioroethyl, 1,1-dimethy 1-2,2- dibromoethyl, l,l~dimethyl-2,2,2~trichloroethyi,

Especiailv at least one and most preferably both of R i and R a are hydrogen.

Preferably R 3a , R " and R 4 are independently hydrogen, alkyl, especially methyl or ethyl or aryi especially phenyl or araikyi, especially benzyl.

Examples of preferred R ja , R 4 and R ',a groups are independently hydrogen, halogen or methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2- trimethylethyl or the same substituted by at least one halogen atom such as trill uororneihyl, trichioromeihyl, 2,2,2-trichloroethyl, l ,l-dimethyl-2, 2- dibromoethyl , 1 , 1 -dimethyl-2,2,2-trichloroethy 1.

Especially at least one and most preferably both of R 4 and R 4a are hydrogen.

R 3a is particularly hydrogen or alkyl, especially lower alkyl and is most preferably hydrogen.

Preferably R' is hydrogen, Cl -12-alkyl, especially Cl-6-alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, sulfinyl, sulfonyl, carbonyl or oxycarbonyl group and optionally, a Cl-4-alkylene bridge, particularly methylene ; C2~6-alkenyl or -alkynyl, especially C2-3-alkenyl or-aikynyl each optionally substituted by one or more halogens ; azido ; cyano ; ami do ; carboxy ; triazolyi, tetrazolyl, pyrrolidinyl, pyridyl, 1- oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl or piperazinyl each optionally substituted by one or more substituents selected from halogen, C1-6- alkyl and phenyl and attached to the ring either directly or via a carbonyi group or a CI -4-alkylene bridge, particularly methylene ; naphthyl ; or phenyl, phenylalkyl or phenylalkenyl each optionally substituted by one or more substituents selected from halogen, Cl-6-alkyl, CI -6 haloalkyl, Cl ~6~aikoxy, C l-6-alkylthio, amino, azido, phenyl and nitro and each attached to the ring either directly or via an oxy, sulfonyl, sulfonyloxy, carbonyi or carbonyioxy group and optionally additionally a C 1 -4-alkylene bridge, particularly methylene.

Also, preferably, R J is Cl-6-alkyi optionally substituted by one or more substituents selected from halogen, thiocyanato, azido, alkoxy, alkylthio, phenyisulfonyl ; nitrooxy ; C2-3- aikenyl or-alkynyl each optionally substituted by one or more halogens or by acetyl ; tetrazolyl, pyridyi, furyl, pyrrolyl, thiazolyl or thienyl ; or phenyl or phenylalkyl each optionally substituted by one or more substituents selected from halogen, Cl-6-alkyl, CI -6 haloalkyl, Cl-6-alkoxy, amino, azido, phenyl and nitro and each attached to the ring either directly or via a sulfonyloxy and optionally additionally a CI -4-alkylene bridge, particularly methylene.

Other examples of preferred R 3 groups are hydrogen, halogen or methyl, ethyl, propyl, isopropyl. butyl, iso or ter-butyl, 2,2,2-trimethyiethyl or the same substituted by at least one halogen atom such as trifluoromethyl,

trichloromethyl, 2,2,2-trichloroethyl, 1, l-dimethyl-2. 2-dibromoethyl, 1,1- dimethy 1 -2 ,2,2- richl oroethy 1.

R 3 is especially CI -4-alkyl optionally substituted by one or more substituents selected from halogen, thiocyanato or azido; C2-5-alkenyl or- alkynyl, each optionally substituted by one or more halogens; thienyl; or phenyl optionally substituted by one or more substituents selected from halogen, Cl-6-aikyi, CI -6 haloalkyl or azido. Further examples of preferred R ~ groups are CI -6 alkyl and C2-6 haloalkenyl.

Preferably R 5 and R° are independently hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-triniethylethyl, especially hydrogen or methyl.

Especially at least one and most preferably both of R 5 and R 6 are hydrogen.

Preferably R/ is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or tert-butyl, 2,2,2-trimethylethyl, methoxy, ethoxy, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorophenyl.

Preferably R' is hydrogen, methyl or ethyl especially hydrogen.

Preferably R 5 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, iso or ter-butyl, 2,2,2-trimethylethyl, phenyl, benzyl or the same substituted by at least one halogen atom such as trifluoromethyl, chlorobenzyl.

Preferably * is hydrogen or methyl.

Combinations of one or more of these preferred compound groups are especially preferred.

A particular group of compounds of formula I (Compounds 1A) comprises those wherein,

A " is oxygen;

X is~CONR 5 R 6 or-COOR 7 or-CO-R 8 or CN ;

R f is hydrogen or alkyl, aryl, halogen, hydroxy, amino, nitro, cyano;

R "' , R " I 4 , are the same or different and each is independently hydrogen or halogen, hydroxy, amino, nitro, cyano, acyl, acyioxy, a sulfonyl derivative, a sulfinyl derivative, an amino derivative, carboxy, ester, ether, amido, sulfonic acid, sulfonamide,,, alkoxycarbonyl,,, a thio derivative,, alkyl, alkoxy, oxyester, oxyaniido, aryl,, an oxy derivative, lieterocycle, vinyl and R J may additionally represent C2-5 alkenyl, C2-5 alkynyl or azido each optionally substituted by one or more halogen, cyano, thiocyano, azido,, cyelopropyl, acyl and/or phenyl ; or phenyl suifonyloxy whereby any phenyl moiety may be substituted by one or more halogen, alkyl, haloalkyl, alkoxy, nitro, amino, and/or phenyl ; most preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl.

R 2a , R Ja and R 4a are hydrogen;

R ' \ R 6 , R' are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, lieterocycle or oxy derivative; and

R is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, lieterocycle, alkylthio or thio deri ative.

Within these Compounds 1 A, R ! is preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl ; most preferably methyl, ethyl or n-propyl.

R 2 and are preferably independently hydrogen or halogen or methyl, ethyl, propyl, isopropyl, butyl, isobutyl ; and, most preferably, are each hydrogen.

R' is preferably C I -5 alkyl, C2-5 alkenyl, C2-C5 alkynyl, cyelopropyl, azido, each optionally substituted by one or more halogen, cyano, thiocyano, azido, alkylthio, cyelopropyl, acyl and/or phenyl ; phenyl ; phenyl sulfonyl ; phenylsulfonyloxy, tetrazole, thiazoie, thienyl, furyl, pyrrole, pyridine, whereby any phenyl moiety may be substituted by one or more halogen, alkyl, haloalkyl, alkoxy, nitro, amino, and/or phenyl ; most preferably methyl, ethyl, propyl, isopropyl, butyl, or isobutyl.

X is preferably -CGOH or -COOMe or -COOEt or -CONH 2 ; most preferably -CONH .

A further particular group of compounds of formula I (Compounds IB) comprises those wherein, X is-CA 1 NH 2 ,-CA 1 NHCH 3 or-CA ¾ N (CH 3 ) 2 ; 1 is alkyl or phenyl ;

R J is alkyl, alkenyl, alkynyl, cyano, isotliiocyanato, ether, carboxyl, amido, aryl, heterocycle ; or

R J is CH 2 R 1J wherein R 10 is hydrogen, cycloalkyl, oxyester,

oxyalkylsulfonyl, oxyarylsufonyl, aminoalkylsulfonyl, aminoarylsulfonyl, nitrooxy, cyano, isothiocyanato, azido, alkylthio, arylthio, alkylsulfinyl, alkylsulfonyi, heterocycle, aryloxy, alkoxy or trifluoroethyl;

R a is hydrogen, alkyl or aryl (especially with the proviso that when R a is hydrogen, R 3 other than methyl); or R J R 3a form a cycloalkyl ; and z , R 2a , R 4 and R 4a are each hydrogen.

Within the compounds of formula I,

R 1 is preferably alkyl especially Cl-12- more particularly Cl-6-alkyl and is most preferably ethyl;

R 2 , R ~a , R 3a and R 4a are preferably hydrogen;

R J is preferably selected from hydrogen; Cl-12-alkyl, especially Cl-6- alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato or alkoxy and attached to the ring either directly or via a thio, su!finyl, sulfonyi, carbonyi or oxycarbonyl group and optionally additionally a Cl-4-alkylene bridge, particularly methylene; C2-6- alkenyl or-alkynyi, especially C2-3-alkenyi or-aikynyl, each optionally substituted by one or more halogens ; azido ; cyano ; amido ; carboxy ;

triazolyl, tetrazolyi, pyrrolidinyi, pyridyl, l-oxidopyridyl, thiomorpholinyl, benzodioxolvl, furyl, oxazolyl, pyrimidinyl, pyrrolyl, thiadiazolvl, thiazolvl, thienyl or piperazinyl each optionally substituted by one or more substituents selected from halogen, Cl-6-alkyl and phenyl and attached to the ring either directly or via a carbonyl group or a Cl-4-a!ky!ene bridge, particularly methylene ; naphthyl ; or phenyl, phenylalkyl or phenySalkenyl each optionally substituted by one or more substituents selected from halogen, Cl-6-alkyl, Cl- 6 haloalkyS, Cl-6-alkoxy, Cl-6-alkylthio, amino, azido, phenyl and nitro and each attached to the ring either directly or via an oxy, sulfonyl, sulfonyloxy, carbonyl or carbonyloxy group and optionally additionally a CI -4- alkylene bridge, particularly methylene ;

R " ' a is preferably hydrogen or Cl -4-alkyl ;

R 4 and R 4a are preferably, independently hydrogen, CI -4-alkyl, phenyl or benzyl.

A further group of compounds of formula I (Compounds 1C) comprises those in racemic form wherein, when X is-CO R'R 6 and R 1 is hydrogen, methyl, ethyl or propyl, then substitution on the pyrrolidine ring is other than mono-, di-, or tri-methyl or mono-ethyl.

A further group of compound of formula I (Compounds ID) comprises those in racemic form wherein, when X is-CONR 5 R° and R 1 is hydrogen or Cl-6-alkyl, C2-6-alkenyl or- alkynyl or cycloalkyl, each unsubstituted, then substitution in the ring is other than by alkyl, alkenyl or alkynyl, each unsubstituted.

A further particular group of compounds of formula I (Compounds IE) comprises those wherein,

X is-CA ] NH 2 ;

R 1 is 1 ! ;

R J is azidomethyl, iodomethyl, ethyl optionally substituted by 1 to 5 halogen atoms, n- propyl optionally substituted by 1 to 5 halogen atoms, vinyl optionally subsituted by one or two methyl, and/or 1 to 3 halogen atoms, acetylene optionally substituted by CI -4-alkyl, phenyl or halogen ; R ja is hydrogen or halogen, preferably fluorine ; and R , R 2a , R 4 and R 4a are each hydrogen ; as their racemates or in enantiomerically enriched form, preferably the pure enantiomers. A further particular group of compounds of formula I (Compounds IF) comprises those wherein,

X is-CA ! NH 2 ;

R 1 is H ;

R' is Cl-6-alkyl, C2-6-aikenyl or C2-6-aIkynyl optionally substituted by azido, oxynitro, 1 to 6 halogen atoms ;

R 3a is hydrogen or halogen, preferably fluorine ; and R 2 , R 6 *, R 4 and R 4a are each hydrogen : as their racemates or in enantiomerically enriched form, preferably the pure enantiomers.

In all the above mentioned scopes when the carbon atom to which R 1 is attached is asymmetric it is preferably in the "S"-configuration.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of:

(2S)-2-[4-(¾romomethyl)-2-oxo-l-pyrrolidinyl]butanamide;

(2S)-2-[(4R.)-4-(iodomethyl)-2-oxopyrro3.idinyl]butanamid e; (2S)-2-(2-oxo-4-phenyl- 1 -pyrrplidmyl)butanamide;

(2S)-2-[4-(iodomethyl)-2-oxo-l-pyrrolidinyl]butanamide;

(2S)-2-[4-(chloromethyl)-2-oxo-l-pyrrolidinyl]butanarnide ; {l-[(lS)-l-(a.minocarbonyl)propyl]-5-oxo-3-pyrrolidinyl}meth yl 4- methylbenzenesulfonate;

(2S)-2-[(4 )-4-(azidomethyl)-2-oxopyrrolidinyl]butanamide;

2-[4-(2, 2-dibromovinyl)-2-oxo- 1 -pyrrolidinyljbutanamide;

{1 - [(I S) -1 - (aminocarbonyl)propyl]-5-oxo-3-pyTTolidinyl}m.ethyl nitrat

(2S)-2-[2-oxo-4-(lH-tetraazol-l -ylmethyl)-l -pyrrolidinyljbutanamide;

2-(2-oxo-4- vinyl- l-pyrrolidinyl)butanamide;

2- {2-oxo-4-[(phenylsulfonyl) methyl j- 1 -pyrrolidinyljbutanamide;

(2S)-2-[(4R)-4-(2, 2-dibromovinyl)-2-oxopyrrolidinylJbutanamide;

(2S)-2-[(4S)-4-(2, 2-dibromovmyl)-2-oxopyrrolidinyl]butanamide;

(2S)-2-[4-(isothiocyanatomethyl)-2-oxo-l -pyrrolidinyljbutanamide;

2-[2-oxo-4-(l,3-thiazol-2-yl)-l -pyrrolidinyljbutanamide;

(2S)-2-[2-oxo-4-(2-thienyl)-l-pyrrolidinylJbutanamide;

(2S)-2-[4-(2-methoxyphenyl)-2-oxo- 1 -pyrrolidinyljbutanamide;

(2S)-2-[4-(3-methoxyphenyl)-2-oxo-l -pyrrolidinyljbutanamide;

(2S)-2-[4-(4-azidophenyl)-2-oxo- 1 -pyrrolidinyljbutanamide;

(2S)-2-[2-oxo-4-(3-thieny[)-l-pyrrolidinylJbutanamide;

(2S)-2-[4-(3-azidophenyl)-2-oxo-l -pyrrolidinyljbutanamide;

(2S)-2-[2-oxo-4-(3-thieny[)-l-pyrrolidinylJbutanamide;

(2S)-2-[(4S)-2-oxo-4-viny[pyrrolidinylJbutanamide;

(2 S)-2- [(4R)-2-oxo-4- vinylpyrro I idinyl Jbutanamide; 2-[4-(2-bromophenyl)-2-oxo-l-pyrrolidinyl]butanamide:

2-[2-oxo-4-(3-pyridinyl)-l-p}Trolidinyl]butanamide;

(2S)-2-(4-[ 1 , 1 '-biphenyl]-4-yl-2-oxo- 1 -pyrrolidinyl)butanamide;

(2S)-2- |4-[(methylsulfanyl) methyl]-2-oxo- 1 -pyrrolidinyl} butanamide; 2-[4-(iodomethyl)-2-oxo- 1 -pyrrolidinyl] butanamide;

(2S)-2-[(4R)-4-(iodomethyl)-2-oxo-l -pyrro[idinyl]pentanamide;

(2S)-2-[(4R)-4-(iodomethyl)-2-oxopyrrolidinyl]propanamide ;

2-(2-oxo-4-propyl- 1 -pyrrolidiny [)propanamide;

2-(2-oxo-4-propyl- 1 -pyrrolidiny [)butanamide;

2-(2-oxo-4-penty !- 1 -pyrrolidinyl ) butanamide;

(2S)-2-[(4R)-4-(iodomethyl)-2-oxopyrrolidinyl]- -methylbutanamide; (2S)-2-(4-neopentyl-2-oxo-l -pyrrolidiny! )butanamide;

(2S)-2-(4-ethyl-2-oxo-l-pyrrolidinyi)butanamide;

2-[4-(2,2-difluorovinyl)-2-oxo-l -pyrroiidinyl]butanamide;

2-[4-(2,2-difluoroethyl)-2-oxo-l-pyrrolidinyl]butanamide;

(2S)-2-[(4S)-2-oxo-4-propylpyrrolidinyl]butanamide;

(2S)-2-[(4R)-2-oxo-4-propylpyrrolidinyl]butanamide;

2- {4-[(Z)~2-fluoroethenyl]~2-oxo- 1 ~pyrrolidinyl}buiariarmde;

2-[4-(2-methyl-l -propenyl)-2-oxo-l-pyrro3.idinyl]butanamide;

2-(4~butyi~2~oxo-l-pyrrolidmyl)buianamide;

2-[4-(cyclopropylmethyl)-2-oxo- 1 -pyrrolidinyljbutanamide; 2-(4-isobutyl-2-oxo-l-p>Trolidinyl)butana.mide;

2-[4-(4-chlorophenyl)-2-oxo-l-pyrrolidinyl]butanamide;

2-[4-(3-chlorophenyl)-2-oxo-l-pyrrolidinyl]butanamide;

2-{2-oxo-4-[2-(trifluoromethyl)phenyl]-l-pyrrolidinyl}butana mide;

2-[4-(2-fluorophenyl)-2-oxo-l-pyrrolidinyl]butanamide;

2-[4-(3-methylphenyl)-2-oxo-l-pyrrolidinyl]butananiide;

(2S)-2-[2-oxo-4-(2-phenylethyl)-l-pyrrolidinyl]butananiide;

(2S)-2-[4-(3-bromophenyl)-2-oxo-l-pyrrolidinyl]butanamide;

2-{4-[3,5-bis(trifluoromethyl)phenyl]-2-oxo-l-pyrrolidinyl}b utanamid

2-[4-(3,4-dichlorophenyl)-2-oxo-l -pyrroiidinyl]butananiide;

2-[4-(2,4-dichlorophenyl)-2-oxo-l -pyrroiidinyl]butananiide;

2-[4-(2-furyl)-2-oxo-l-pyrrolidinyl]butanai ' nide;

(2S)-2-[2-oxo-4-(3-pheivylpropyl)-l-pyrrolidinyl]butanamide;

(2 S)-2- [4-(3 ,5 -dibromophenyl)-2-oxo- 1 -pyrro 1 idinyljbutanami de;

2-[4-(3,4-dichlorophenyl)-2-oxo-l -pyrroiidinyl]butananiide;

2-(2-oxo-4-propyl-l-pyrrolidinyl)butanamide;

2-[4-(3-chlorophenyl)-2-oxo-l-pyrrolidinyl]buta.namide;

2-(4~ethynyl-2~oxo- 1 -pyrroiidiriyl) butanamide;

2-[4-(2-fluorophenyl)-2-oxo-l-pyrrolidin ljbutanamide;

(2 S)-2- [4-(cycl opropylmetby l)-2-oxo- 1 -pyrrol .idinyl } b utanamide;

(2S)-2-[(4S)-4-(2„ 2-difluorovinyl)-2-oxopyrrolidinyl]bulanamide; (2S)-2- [2-oxo-4-(3 , 3, 3 -trifluoropropyl)- 1 -pyrrolidinyljbutanamide; 2-[4-(3-methylphenyl)-2-oxo-l-pyrrolidinyl]butanamide:

(2S)-2-[4-(cyclopropylmethyl)-2-oxo-l-p>Trolidinyl]but ana.mide;

(2S)-2-[(4R)-4-(2, 2-difluorovinyl)-2-oxopyrrolidinyl]butanamide: (2S)-2-[2-oxo-4-( 1 H-pyrrol- 1 -yl)- 1 -pyrrolidinyljbutananiide;

(2S)-2-(4-al[yl-2-oxo-l -pyrro[idinyl)butanannde;

(2S)-2-[4-(2-iodopropyl)-2-oxo-l-pyrrolidinyl}butanamide;

(2S)-2-(4-al[yl-2-oxo-l -pyrro[idinyl)butanannde;

(2S)-2-[2-oxo-4-(2-oxopropyl)-l -p>Tro[idinyl]butanamide;

(2S)-2-[4-(2-bronio- 1 H-pyrrol- 1 -yi)-2-oxo- 1 -pyrrolidinyljbutanamide; (2 S)-2-(4-methy I -2-oxo-4-propy 1 - 1 -pyrrolidinyl)b utanamide;

(2R)-2-[4-(2, 2-dichlorovinyl)-2-oxo-l -pyrrolidinyljbutanamide;

2-[4-(bronioethynyi)-2-oxo-l -pyrrolidinyljbutanamide;

2-[(4S)-4-(2, 2-difluoropropyl)-2-oxopyrrolidinylJbutanamide;

(2S)-2-[4-(bromoethynyl)-2-oxo-l -pyrrolidinyljbutanamide;

2- (2-oxo-4-propyl-l-pyrrolidinyl)pentanamide;

3- cyclopropyl-2-(2-oxo-4-propyl-l-pyrrolidinyl)propanam.ide;

2-(2-oxo-4-propyl-l-pyrrolidinyl)-3-(l ,3-thiazo3-4-yl)propanamide; 2-(2-oxo-4-propyl-l-pyrrolidinyl)-4-penten amide;

(2S)-2-[(4R)-2-oxo-4-vmylpyrrolidinylJbutanam.ide; including all isomeric forms and mixtures thereof or a pharmaceutically acceptable salt thereof.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of : (2S)-2-[(4S)-4-(2, 2-difiuorovinyl)-2-oxopyrrolidinyl]butanamide;

(2S)-2-[(4S)-2-oxo-4-propy[pyrrolidinyl]butanamide;

(2S)-2-[(4R)-2-oxo-4-propylpyrro[idinyl]butanamide. ii) International Patent Application WO 2002/094787:

Compounds of the formula I

wherein n represents 0 or 1 whereby R 1 is not existent when n~0 and R' is existent when n= 1 ;

A 1 represents an oxygen or a sulfur atom;

X is-CONR 7 R 8 ,-COOR 9 ,-CO-R 10 0 r CN;

R 1 when existent, R 2 , R J , R " and R 3 are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyl, ester, ether, aryl, heterocycle, or an oxy derivative, thio derivative, amino derivative, acy! derivative, sulfonyl derivative or suifmyl derivative, provided that at least one of the substituents R chosen from R 1 when existent, R 2 , R J , R 4 or R 5 is not hydrogen;

R° is hydrogen, alkyl, aryl or-CH?-R iia wherein R 6a is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;

R', R s and R 9 are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative; and

R f 0 is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative; their pharmaceutically acceptable salts, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers).

In the above formula, at least one substituent R 1 to R 5 is different from hydrogen. Some non-substituted compounds are referred to in US Patent No. 5,468,733 and 5,516, 759. US Patent No. 5,468,733 refers to non-ring substituted 2-oxo- 1 -pyrrolidinyl and 2-oxo-J -piperidinyl derivatives as inhibitors of the oncogene Ras protein. In particular, these compounds block the ability of Ras to transform normal cells to cancer cells, and therefore ca be included in se veral chemotherapeutic compositions for treating cancer.

US Patent No. 5,516,759 refers to non-ring substituted 2-oxo- 1- pyrrolidinyl, 2-oxo- 1- piperidinyl and azepanyl derivatives present at the N- terminus of dodecapeptides possessing LHRH (luteinizing hormone-releasing hormone) antagonistic activity. Such LHRH antagonists are useful in the treatment of a variety of conditions in which suppression of sex steroids plays a key role including contraception, delay of puberty, treatment of benign prostatic hyperplasia a, o. In the definitions set forth below, unless otherwise stated, R" and R 12 are the same or different and each is independently amido, alkyl, alkenyl, alkynyl, acyl, ester, ether, aryl, aralkyl. heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyS derivative, or sulfmyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.

The term "oxy derivative", as used herein, is defined as incIuding-O-R 1 ' groups wherein R 11 is as defined above except for "oxy derivative", Non- limiting examples are alkoxy, alkenyl oxy, alkynyloxy, acyloxy, oxyester, oxyamido, aikyisulfonyloxy, alkylsulfinyloxy, arylsuifonyloxy,

arylsuifmyloxy, aryioxy, aralkoxy or heterocyclooxy such as pentyloxy, allyioxy, methoxy, ethoxy, phenoxy, benzyioxy, 2-naphtiiyloxy, 2-pyridyloxy, methylenedioxy, carbonate.

The term "thio derivative", as used herein, is defined as including-S-R 11 groups wherein R 1 1 is as defined above except for "thio derivative". Non- limiting examples are alkylthio, alkenylihio, alkynylthio and arylthio.

The term "amino derivative", as used herein, is defined as including-NHR *1 or-NR f f R 12 groups wherein R 11 and R ] " are as defined above. Non-limiting examples are mono- or di-alkyl-, alkenyl-, alkynyi-and arylaniiiio or mixed amino.

The term "acyl derivative", as used herein, represents a radical derived from carboxyiic acid and thus is defined as including groups of the formula R^-CO-, wherein R *1 is as defined above and may also be hydrogen. Preferred are acyl derivatives of formula -COR 11 wherein R ¾ ¾ is selected from hydrogen, Cl-12 alkyl, C2-12 alkenyl, C2-12 alkenyl, heterocyle and aryl, Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyl,

heptanedioyl, cyclohexanecarbonyl, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxaiyl, ethoxalyl, cysteinyl, oxamoyl. The term "sulfonyl derivative", as used herein, is defined as including a group of the formula -S0 2 - ] ] , wherein R f 1 is as defined above except for "sulfonyl derivative", Non-limiting examples are alkylsulfonyl,

aikenylsulfonyl, aikynylsulfonyl and arylsulfonyi.

The term "suffirryl derivative", as used herein, is defined as including a group of the formul a -SO-R 11 , wherein R 11 is as defined above except for "sulfinyl derivative". Non-limiting examples are alkylsulfinyl, alkenylsulfinyl, alkynylsulfmyl and aryl sulfinyl.

The term "a!ky!", as used herein, is defined as including saturated, monovalent hydrocarbon radicals ha ving straight, branched or cyclic moieties or combinations thereof and generally containing 1 -20 carbon atoms, most often 1 to 12 carbon atoms, preferably 1-7 carbon atoms for non-cyclic alkyl and 3-7 carbon atoms for cycloalkyS (in these two preferred cases, unless otherwise specified,"lower alkyl"), each optionally substituted by, preferably 1 to 5, substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, alkylamino, carboxy, ester, ether, amido, azido, cvcloalkyl, sulfonic acid, sulfonamide, thio derivative, alkylthio, oxyester, oxyamido, heterocycie, vinyl, alkoxy (preferably CI -5), aryloxy (preferably C6-10) and aryl (preferably C6-10).

Preferred are alkyl groups containing 1 to 7 carbon atoms, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl. Most preferred are CI -4 alkyl and C3-7 cycloalkyl, each optionally substituted by one or more hydroxy, halogen, lower alkyl or/and azido.

Most preferred alkyl groups are hydroxymethyl, propyl, butyl, 2, 2,2- trifluoroethyl, 2- bromo-2,2-difluoroethyl, 2-chloro-2,2-difluoroethyl, 3,3,3- triiluoropropyi, cyclopropylmethyi, iodomethyl, azidomethyl, 2,2- difluoropropyS, 2-iodo-2,2-difiuoroethyi . The term "lower alkyl", as used herein, and unless otherwise specified, refers to Ci to C 7 saturated straight, branched or cyclic hydrocarbon. Non limiting examples are methyl, ethyl, propyl, isopropyl, butyl, tertiobutyl, pentyl, cyc!opropyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methypentyi, 2,2-dimethylbutyl, optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably, lower alkyl is methyl.

The term "alkenyl". as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, thiocyanato, azido, alkvlthio, cycioalkyl, acyl, nitro, cyano, aryl and heterocycle.

Prefered alkenyl groups are C2-C12 alkenyls, especially C2-6 alkenyis, such as ethenyl ( :::: vinyl), 1 -methyl- 1 -ethenyl, 2,2-dimethy -l-ethenyl, 1- propenyl, 2-propenyl (= ailyl), 1-butenyl, 2- butenyl, 3-butenyl, 4-pentenyl, 1- methyi~4-pentenyi, 3-methy3-l-pentenyl, 1-hexenyl, 2-hexerryl and the like, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkvlthio, cycioalkyl, phenyl and acyl. Most prefered is vinyl, optionally substituted by one or more halogen or/and lower alkyl, and especially 2,2- difluorovfnyl, 2,2-dibromovinyl and 2,2-dichiorovinyi.

The term "alkynyl" as used herein, is defined as including a monovalent branched or unbranched hydrocarbon radical containing at least one carbon- carbon triple bond, for example ethynyl, 2-propynyl (= propargyl), and the like, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, aryl, heterocycle, thiocyanato, azido, alkvlthio, alkyl and acyl.

Preferred alkynyl groups are C2-12 alkynyl, especially C2-6 alkynyl, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, acyl, aryl such as phenyl and alkyl, preferably cycloalkyl.

Most preferred are ethynyl, propynyl and butynyl, optionally substituted by lower alkyl or/and halogen, and especially l-propynyl, cyclopropylethvnyl, 3- methyl- 1 -butynyl and 3,3,3- trifluoro- l-propynyl.

When present as bridging groups, alkyl, alkenyl and alkynyl represent straight- or branched chains, C l-12, preferably C l-4-alkylene or C2-12-, preferably C2-4-alkenylene or- alkynylene moieties respectively.

Groups where branched derivatives are conventionally qualified by prefixes such as "n", "sec", "iso" and the like (e. g."n~propyl", "sec-butyl'') are in the n- form unless otherwise stated.

The term "aryl", as used herein, is defined as including an organic radical deri ved from an aromatic hydrocarbon consisting of at least one ring, most often 1 to 3 rings and generally containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl, each optionally substituted by one or more substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyl, sulfmyl, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, Cl-6-alkylthio, ox ester, oxyamido, aryl, Cl -6-aLkoxy, C6-10- aryloxy, Cl-6-alkyl, Cl-6-haloalkyl. Aryl radicals are preferably monocyclic or bicyclic containing 6-10 carbon atoms. Preferred aryl groups are phenyl and naphthyl each optionally substituted by one or more substituents independently selected from halogen, nitro, amino, azido, Cl-6-alkoxy, Cl-6-alkyl, Cl-6- haloalkyl, sulfonyl and phenyl. Preferred aryl is phenyl, optionally substituted by one or more halogen, lower alkyl, azido or nitro, such as 3-chlorophenyl and 3-azidophenyL

The term "halogen", as used herein, includes an atom of CI, Br, F, I.

The term "hydroxy", as used herein, represents a group of the formula -OH. The term "thiol", as used herein, represents a group of the formula -SH.

The term "cyano", as used herein, represents a group of the formula -CN.

The term "nitro", as used herein, represents a group of the formula -N0 2 .

The term "nitrooxy", as used herein, represents a group of the formula - ON0 2 .

The term "amino", as used herein, represents a group of the formula -NH 2 .

The term "azido", as used herein, represents a group of the formula -N 3 .

The term "carboxy", as used herein, represents a group of the formula - COOH. The term "sulfonic acid", as used herein, represents a group of the formula -

The term "sulfonamide", as used herein, represents a group of the formula -

The term "ester", as used herein, is defined as including a group of formula -COO-R 11 wherein R' ' is as defined above except oxy derivative, thio derivative or amino derivative, Preferred are esters of formula -COOR 11 wherein R J J is selected from C I -12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aryi. Most preferred are esters where R * 1 is a lower alkyl, especially methyl.

The term "ether" is defined as including a group selected from CI -50- straight or branched alkyl, or C2-50-straight or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.

The term "amido" is defined as including a group of formula -CONH 2 or - CONBR 5 ' or -CONR R 12 wherein R n and R 12 are as defined above.

The term "heterocycle", as used herein, is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl, and optionally being substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. Non- limiting examples of heterocycles are pyridyl, furyl, pyrrolyl, thienyl, isothiazolyl, triazolyl, imidazolyl, benzimidazolyi, tetrazolyl, quinazolinyl, quinoiizinyl, naphthyridinyl, pyridazinyl, pyrimidinyl, pyrazinyi, quinolyl, isoquinolyl, isobenzofuranyi, benzothienyS, pyrazolyi, indoiyl, indolizinyl, purinyl, isoindolyl, carbazoiyl, thiazolyl, 1,2,4-thiadiazolyi, thiomoi * phoiinyl, thieno (2,3-b) furanyl, furopyranyl, benzofuranyl, benzoxepinyl, isooxazolyS, oxazolvl, thianthrenyl, benzothiazolyl, or benzoxazolyl, cimiolinyl, phthalazinyl, quinoxalinyl, l-oxidopyridyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenothiazinyl, furazanyl, benzodioxolyl, isochromanyl, indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5~azacytidinyl,

5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazoiopyrimidinyl, tetrahydro furanyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholino, morpholinyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, sugar moieties (i. e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) optionally substituted by alkyl or as described above for the alkyl groups. The

term"heterocycle"also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryi ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1) heptanyl, 7- oxabicyclo (2.2.1) heptanyl, 8-azabicyclo (3.2.1) octanyl.

The heterocycle is preferably selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1 - oxidopyridyl, thiomorpholinyl, benzodioxolyl, furyl, oxazolvl, pyrimidinyl, pyrrolyl, thiadiazolyL thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alky I, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl.

More preferably the heterocycle is selected from tetrazolyl, pyrrolidmyl, pyridyi, furyl, pyrrolyl, thiazolyl and thienyi, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl, and especially from 2-and 3- thienyl, optionally substituted by one or more halogen, acyl such as formyl, cyano and/or lower alkyl, such as methyl.

In the above definitions it is to be understood that when a substituent such as R 1 , R 2 , R 3 , R 4 , R 5 , R', R*, R 9 , R 10 is attached to the rest of the molecule via a heteroatom or a carbonyl, a straight- or branched chain, CI -12-, preferably C1 ~ 4-alkylene or C2-12, preferably C2-4-alkenyiene or-aikynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule. The term"R substituenfrefers to R 1 , R 2 , R 3 , R^ or R s , independently.

According to a preferred embodiment, a compound of formula I is as defined above wherein n represents 0, The compound is a 6-ring structure (2- thioxo- or 2-oxo-piperidinyl derivative) wherein R 1 is not existent since n=0, and is depicted by the formula (I-A).

According to a following embodiment, the compound of formula I is as defined above wherein n represents 1. The compound is a 7-ring stmcture (2- thioxo- or 2-oxo-azepanyl derivative) wherein R is existent since 11= 1 and depicted by the formula (I-B).

According to a more preferred embodiment, said compound is as defined above wherein n=0, R 3 and/or R 4 are different from hydrogen and R " and R 3 represent hydrogen,

According to another more preferred embodiment, said compound is as defined above wherein n=l, R "' , R " ' and/or R 4 are different from hydrogen and wherein R 1 and R 3 represent hydrogen.

According to a yet more preferred embodiment, said compound is as defined above wherein only one R substituent chosen from R 3 or R 4 when n :::: 0 or from R z , R 3 or R 4 when 11= 1 , is different from hydrogen and the remaining R substituent(s) is/are hydrogen, We hereby refer to a mono-substituted 2-thioxo- or 2-oxo-piperidinyl or 2-thioxo- or 2-oxo-azepanyl derivatives.

According to another preferred embodiment, compounds of formula I are as defined above wherein A 1 represents an oxygen atom. We hereby refer to 2- oxo-piperidinyl or 2-oxo-azepanyl derivatives.

According to another preferred embodiment, compounds of formula I are as defined above wherein X is CONR ' R , especially CON3¾. We hereby refer to amido derivatives of 2-oxo (or thioxo)~piperidinyl or 2-oxo (or thioxo) - azepanyl. According to another preferred embodiment, compounds of formula I are as defined above wherein R 6 represents hydrogen, CI -4 alkyl, or a CH 2 -R 6 group wherein R 5a represents a heterocycle. Most preferably R 6 is a Cl-4 alkyl, especially ethyl. When R° is ethyl we refer to 2- (2-oxo (or thioxo)-!- piperidinyl) butanamide or 2- (2-oxo (or thioxoXl -azepanyl) butanamide derivatives.

According to another preferred embodiment, compounds of formula I are as defined above wherein the carbon atom to which R° is attached is of the S configuration. In case where R 6 is ethyl, A is oxygen and X is CONR'R 8 we refer then to (2S)-2-(2-oxo-l-piperidinyl) butanamide or (2S)-2- (2-oxo- 1- azepanyl) butanamide derivatives.

According to a prefered embodiment, the compound is as defined above wherein R' when n= , R 3 and R 4 are the same or different and each is independently hydrogen, halogen, itro, nitrooxy, cya o, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkerryl, alkynyl, ester, ether, aryl, heterocycle, acyl derivative, sulfonyl derivative or sulfmyl derivative;

R 1 when existent, R 2 when n=0 and R 5 are hydrogen;

R 6 is hydrogen, alkyl, aryl or-CH 2 -R 'Jil wherein R oa is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;

According to this preferred embodiment, the compound is generally such that when R 6 is benzyl, X is-COOCH 3 and n=l, R "' is different from methyl when R 3 and R 4 are both hydrogen and R 4 is different from methyl when R" and R 3 are both hydrogen.

According to another preferred embodiment, the compound is as defined above wherein R 2 when n= 1 , R J and R 4 are the same or different and each is independently hydrogen; cyano; carboxy; amido Cl-12 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cycloalkyl, acyl, aryl and heterocycle;

C2-12 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl;

C2-12 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl ; acyl derivative of formula -C()-R ! f , wherein R f 1 is selected from Cl -12 alkyl, C2-12 alkenyl, C2-12 alkynyl, heterocycle and aryl; ester of formula -CO-O-R 11 wherein R n is selected from Cl-12 alkyl, C2- 12 alkenyl, C2-12 alkynyl and aryl; heterocycle selected from triazo!y!, tetrazolyl, pyrrol idinyl, pyridyl, 1- oxidopyridyl, thiomorpholinyl, benzodioxolyl, fui l, oxazolyl, pyrimidinyl, pyrroiyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl; aryl, each optionally substitued by one or more substituents selected from CI -6 alkyl, C I -6 haloalkyl, CI -6 alkoxy, CI -6 alkylthio, amino, azido, sulfonyi, aryl and nitro.

According to another preferred embodiment, the compound is as defined above, wherein R" when n :=: 1 , R 3 and R 4 are the same or different and each is independently hydrogen;

CI -7 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cyclopropyl, acyl and phenyl; C2-6 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycioalkyl, phenyl and acyi ;

C2-6 alkynyl. each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycioalkyl, phenyl and acyi; heterocycle selected from tetrazolyl, pyrroiidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyi, aikoxy, nitro, amino and phenyl; phenyl, each optionally substitued by one or more substituents selected from CI -6 alkyl, halogen substituted alkyl, halogen, aikoxy, amino, azido, sulfonyi, phenyl and nitro,

According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R 2 , R 3 and R 4 when n=l or from the group R 3 and R 4 when n=0, represents independently Cl-4-alkyl or C3-7-cycloalkyl, optionally substituted by one or more halogen, hydroxy, lower alkyl and/or azido.

According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R 2 , R 3 and R 4 when n=l or from the group " and R 4 when n=0, represents independently vinyl, optionally substituted by one or more halogen or/and lower alkyl.

According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R", R' and R ' ' when n-l or from the group R J and R 4 when n :::: 0, represents independently ethynyl, propynyl or butynyl, optionally substituted by one or more halogen and/or lower alkyl.

/ J According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R , R 3 and R 4 when 11= 1 or from the group R 3 and R 4 when n=0, represents independently phenyl, optionally substituted by one or more halogen, lower alkyl, azido and/or nitro.

According to another preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R 2 , R 3 and R 4 when n=l or from the group R J and R 4 when n=0, represents independently 2-or 3-thieiiyl, optionally substituted by one or more halogen, acyl, cyano or/and lower alkyl.

According to a particular preferred embodiment, the compound is as defined above wherein at least one of the R substituents chosen from the group R 3 , R 4 and " when n :=: 1 or from the group R' and R 4 when n :=: 0, is hydroxymethyi, propyl, butyl, 3,3,3-trifluoropropyl, 2,2,2-trifluoroethyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2- thienyi, 3-thienyl, phenyl, 3- chlorophenyl, 3-azidophenyl, 2,2-difluorovinyl, 2,2-dibromovinyl, 2, 2- dichiorovinyl, 2-ethynyl, 5-methyl~2-thienyl, 5~ibrmyl~2-eihynyl, 5~eyano~2- thienyl, 3-bromo- 2-thienyl, 4-methyl-2 -thienyi, 3,3,3-trifluoro-l-propynyl, 1- propynyl, cyciopropylethynyl, 3- methyl- 1-butynyl, 1-butynyl, 2,2- difluoropropyl, 2-chloro-2,2-difluoroethyl, 2-bronio-2,2- difluoroethyl and 2- iodo-2,2-difluoroethyl,

According to yet another preferred embodiment, the compound is as defined above wherein R ! , R " , R and ' f are hydrogen.

According to even another preferred embodiment, the compound is as defined above wherein R', R 2 , R 3 and R 3 are hydrogen.

According to even another preferred embodiment, the compound is as defined above wherein n :::: l and R 1 , R J , R' and R 5 are hydrogen,

In all the above-mentioned scopes when the carbon atom to which R° is attached is asymmetric it is preferably in the"S"-configuration. Representative compounds useful in the methods and compositions of this invention as defined abo ve are selected from the group consisting of

2-[5-(hydroxyniethyl)-2-oxo-l -piperidinyl]butanarnide,

2-(2-oxo-5-propyl- 1 -piperidinyi)butanamide,

2-[2-oxo~5~(33,3-trifluoroprop>4)~l-piperidinyl]butanarni de,

2-[5-(cyclopropylrn.ethyl)-2-oxo-l-piperidinyl]butanamide,

2-[5-(iodomethyl)-2-oxo- 1 -piperidmyl] butanamide,

2-[5~(azidomethy{)~2-oxo-l ~piperidiny]]butana.niide,

2-(2-oxo-5 -phenyl- 1 -piperidmyl)butanamide,

2-[2-oxo-5-(2-thienyl)- 1 -piperidinyljbutanamide,

2-[2-oxo-5 -(3 -thienyl)- 1 -piperidinyljbutanamide,

2-[5-(3-chlorophenyl)-2-oxo-l -piperidinyljbutanamide,

2-[5-(3-azidophenyl)-2-oxo-l -piperidinyljbutanamide,

2-[5-(2, 2-difluorovinyl)-2-oxo-l -piperidinyljbutanamide,

2-[5-(2, 2-dibromovinyl)-2-oxo-l -piperidinyljbutanamide,

2-[5-(2, 2- dichlorovinyl)-2-oxo-l -piperidinyljbutanamide,

2-(5-ethynyl-2-oxo- 1 -piperidinyljbutanamide,

2[5-(5-methyl-2-thieny[)-2-oxo-l -piperidinyljbutanamide,

2-[5-(5-formyl-2-thienyl)-2-oxo-l -piperidinyljbutanamide,

2-[5-(5-cyano-2-thienyl)-2-oxo- 1 -piperidinyljbutanamide,

2-[5-(3-bromo-2-thienyl)-2-oxo-l -piperidinyljbutanamide, -[5-(4-methyl-2-thienyl)-2-oxo-l -piperidmyljbutanamide,-[2-oxo-5 -(3 ,3,3 -trifluoro- 1 -propynyl)- 1 -piperidmyljbutanamide,-[2-oxo-5-(l -propynyl)- 1 -piperidmyljbutanamide,

-[5-(cyclopropylethynyl)-2-oxo- 1 -piperidmyljbutanamide,-[5-(3-methyl-l -butynyl)-2-oxo- 1 -piperidmyljbutanamide,-[5-( 1 -butynyl)-2-oxo- 1 -piperidmyljbutanamide,

-[5-(2,2-difluoropropyl)-2-oxo-l -piperidmyljbutanamide,-[5-(2-chloro-2,2-difluoroethyl)-2-ox o-l -piperidmyljbutanamide,-[5-(2-bromo-2,2-difluoroethyl)-2-oxo -l -piperidmyljbutanamide,-[4-(hydroxymethyl)-2-oxo-l -piperidmyljbutanamide,

-(2-oxo-4-propyl- 1 -piperidinyl)butanamide,

-[2-oxo-4-(3,3,3trifluoropropyl)-l -piperidinyijbutan amide,-[4-(cyclopropylrnethyl)-2-oxo-l-piperidinyiJbutanamid e,-[4-(iodomethyl)-2-oxo-l -piperidmyljbutanamide,

-[4-(azidomethyl)-2-oxo-l -piperidmyljbutanamide,

-(2-oxo-4-phenyl- 1 -piperidmyl.)butanamide,

-[2-oxo-4-(2-thienyl)-l -piperidinyljbutanami.de,

-[2-oxo-4-(3-thienyl)-l -piperidinyljbutanami.de,

-[4-(3-chlorophenyl)-2-oxo-l -piperidmyljbutanamide,

-[4-(3-azidophenyl)-2-oxo-l -piperidinyljbutanami.de,

-[4-(2,2-difluorovinyl)-2-oxo-l-piperidinylJbutanamide, -[4-(2,2-dibromovinyl)-2-oxo-l-piperidinyl]butanamide,-[4-(2 ,2-dichlorovinyl)-2-oxo-l-piperidinyl]butanamide,

-(4-ethynyl-2-oxo- 1 -piperidinyl)butanamide,

-[4-(5-methyl-2-thienyl)-2-oxo-l-piperidinyl]butanamide,- [4-(5-fom yl-2-thienyl)-2-oxo-l-piperidinyl]butanamide,-[4-(5-cyano-2- thienyl)-2-oxo-l -piperidinyl]butanamide,-[4-(3-bromo-2-thienyl)-2-oxo-l-pipe ridinyl]butanamide,-[4-(4-methyl-2-thienyl)-2-oxo- 1 -pipendmyljbutanamide,-[2-oxo-4-(3 ,3 ,3-trifluoro- 1 -propynyl)- 1 -pipendmyljbutanamide,-[2-oxo-4-(l -propynyl)-! -piperidiny!jbutanamide,

-[4-(cyclopropylethynyl)-2-oxo-l -piperidinyi]butanamide,-[4-(3-methyl-l -butynyl)-2-oxo-l-piperidinyl]butanamide,-[4-(l -butynyl)-2-oxo- 1 -piperidinyl jbutanamide,

-[4-(2, 2-difluoropropyl)-2-oxo- 1 -piperidinyl] butanamide,-[4-(2-chloro-2,2-difluoroethyl)-2-oxo-l -piperidinyl jbutanamide,-[4-(2-bromo-2 5 2-di.fluoroethyl)-2-oxo-l-piperidinyl]butanamide,[4-(2 ,2 5 2-trifluoroethyl)-2-oxo-l -piperidiny3.jbutanamide,-[5-(hydroxymethyl)-2-oxo-l-azepany l jbutanamide,

-(2-oxo-5-propyl-l-azepanyl)butanamide,

-[2-oxo-5-(3 ,3,3-trifluoropropy 3.)- 1 -azepanyl jbutanamide,-[5-(cyclopropylmethyl)-2-oxo- 1 -azepanyljbutanamide, -[5-(iodomethyl)-2-oxo- 1 -azepanyljbutanamide,

-[5-(azidomethyl)-2-oxo- 1 -azepanyljbutanamide,

-(2-oxo-5 -phenyl- 1 -azepanyljbutanamide,

-[2-oxo-5-(2-thienyl)- 1 -azepanyljbutanamide,

-[2-oxo-5-(3-thienyl)- 1 -azepanyljbutanamide,

-[5-(3-chlorophenyl)-2-oxo-l -azepanyljbutanamide,-[5-(3-azidophenyl)-2-oxo- 1 -azepanyljbutanamide,

-[5-(2,2-difluorovinyl)-2-oxo- 1 -azepanyljbutanamide,-[5-(2,2-dibromovinyl)-2-oxo-l -azepanyljbutanamide,-[5-(2,2-dichlorovinyl)-2-oxo-l -azepanyljbutanamide,-(5-ethynyl-2-oxo- 1 -azepanyl)butanamide,

-[5-(5-methyl-2-thienyl)-2-oxo-l -azepanyljbutanamide,-[5 ~(5 -formy ! -2-thi enyl)-2-oxo- 1 -azepany I jbutan amide,-[5-(5-cyano-2-thienyl)-2-oxo-l -azepanyljbutanamide,-[5 ~(3 -bromo-2 -thieny l)-2-oxo- 1 -azepanyljbutanamide,-[5-(4-methyl-2-thienyl)-2-oxo-l -azepanyljbutanamide,-[2-oxo-5-(3 ,3 ,3-trifluoro- 1 -propynyl)- 1 -azepanyljbutanamid-[2-oxo-5-(l -propynyl)-l -azepanyljbutanamide,

-[5-(cyclopropylethynyl)-2-oxo-l -azepanyljbutanamide,-[5-(3-methyl-l -butynyl)-2-oxo-l -azepanyljbutanamide,-[5-(l-butynyl)-2-oxo-l-azepanyljbutana mide, 2-[5-(2,2-difluoropropyl)-2-oxo-l-azepanyl]butananiide.

2-[5-(2-cMoro-2 > 2 lifluoroethyl)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(2-bromo-2.2-difluoroethyl)-2-oxo-l-azepanyl]butanamide , 2-[5-(2,2,2-trifluoroethyl)-2-oxo-l-azepanyl]butanamide, 2-[6-(hydroxymethyl)-2-oxo- 1 -azepanyl]butananiide,

2-(2-oxo-6-propyl- 1 -azepanyl)butananiide,

2-[2-oxo-6-(3,3,3-trifluoropropyl)-l -azepanyl]butanamide, 2-[6-(cyclopropylmethyl)-2-oxo-l-azepanyl]butanamide, 2-[6-(iodomethyl)-2-oxo- 1 -azepanyljbutanamide,

2-[6-(azidomethyl)-2-oxo- 1 -azepanyljbutanamide,

2-(2-oxo-6-phenyl- 1 -azepany i)butanamide,

2-[2-oxo-6-(2-thieny !)- 1 -azepanyljbutanamide,

2-[2-oxo-6-(3-thieny !)- 1 -azepanyljbutanamide,

2-[6-(3-chlorophenyl)-2-oxo-l -azepanyljbutanamide,

2-[6-(3-azidophenyl)-2-oxo- 1 -azepanyljbutanamide,

2-[6-(2,2-difluorovmyl)-2-oxo-l -azepanyljbutanamide,

2-[6-(2,2-dibromovinyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(2, 2-dicblorovinyl)-2-oxo- 1 -azepanyljbutanamide, 2-(6-ethynyl-2-oxo-l-azepanyl)butanamide,

2-[6-(5-methyl-2-thienyl)-2-oxo- 1 -azepanyljbutanamide,

2-[6-(5-formyl-2-thienyl)-2-oxo- 1 -azepanyllbutanamide, -[6-(5-cyano-2 hienyl)-2-oxo-l-azepanyl]butanamide >

-[6-(3-bromo-2-thienyl)-2-oxo-l-azepanyl]butanamide,-[6-( 4-methyl-2-thienyl)-2-oxo-l-azepanyl]butanamide,-[2-oxo-6-(3 , 3, 3 -trifluoro- 1 -propynyl)- 1 -azepanyljbutanamide.-[2-oxo-6-( 1 -propynyl)- 1 -azepanyl]butanamide,

-[6-(cyclopropylethynyl)-2-oxo-l-azepanyl]butanamide,-[6- (3-methyl- 1 -butynyl)-2-oxo-l -azepanyl] butanamide,-[6-( 1 -butynyl)-2-oxo- 1 -azepanyljbutanamide,

-[6-(2, 2-difluoropropyl)-2-oxo- 1 -azepanyljbutanamide,-[6-(2-chloro-2,2-difluoroethyl)-2-oxo- l -azepanyljbutanamide,-[6-(2-bromo-2 ,2-dif!uoroethy i)-2-oxo- 1 -azepanyljbutanamide,-[6-(2,2,2-trifluoroethyl)-2-oxo-l -azepanyljbutanamide,-[4-(hydroxymethyl)-2-oxo-l -azepanyljbutanamide,

-(2-oxo-4-propyl- 1 -azepanyljbutanamide,

-[2-oxo-4-(3,3,3-trifluoropropyl)-l -azepanyljbutanamide,-[4-(cyclopropylmethyl)-2-oxo- 1 -azepanyljbutanamide,-[4-(iodomethyl)-2-oxo-l -azepanyljbutanamide,

-[4-(azidomethyl)-2-oxo-l -azepanyljbutanamide,

-(2-oxo-4-phenyl- 1 -azepanyl)butanamide,

-[2-oxo-4-(2-thienyl)-l -azepanyljbutanamide,

-[2-oxo-4-(3-thienyl)- 1 -azepanyljbutanamide, 2-[4-(3-chlorophenyl)-2-oxo-l-azepanyl]butanamide,

2-[4-(3-azidophenyl)-2-oxo-I-azepanyl]butanamide,

2-[4-(2, 2-difluorovinyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(2, 2-dibromovinyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(2,2-dichlorovinyl)-2-oxo- 1 -azepanyljbutanamide,

2-(4-ethynyl-2-oxo- 1 -azepanyl)butanamide,

2-[4-(5-methyl-2-thienyl)-2-oxo-l-azepanyl]butananiide, 2-[4-(5-fom yl-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(5-cyano-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[4-(3-bromo-2-thienyl)-2-oxo-l -azepanyljbutanamide,

2-[4-(4-methyl-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[2-oxo-4-(3 ,3 ,3-trifluoro- 1 -propynyl)- 1 -azepanyljbutanamide, 2-[2-oxo-4-( 1 -propynyl)- 1 -azepanyljbutanamide,

2-[4-(cyclopropylethynyl)-2-oxo-l -azepanyljbutanamide, 2-[4-(3-methyl-l ~butynyS)~2-oxo~l -azepanyljbutanamide,

2-[4-( 1 -butyny l)-2-oxo- 1 -azepanyljbutanamide,

2-[4~(2,2~difluoropropyl)-2-oxo~l -azepanyljbutanamide, 2-[4~(2-ehioro-2,2-diiiuoroethyi)~2~oxo-l -azepanyljbutanamide, 2-[4-(2-bromo-2,2-difluoroetbyl)-2-oxo-l -azepanyljbutanamide, 2-[4-(2,2,2-tritluoroethyl)-2~oxo- 1 -azepanyljbutanamide, In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of:

(2S)-2-[5-(iodomethyl)-2-oxo-l -piperidinyl]butanarnide,

(2S)-2-[5-(azidomethyl)-2-oxo-l-piperidinyl]butananiide, 2-(2-oxo~5~phenyi- 1 -piperidmyljbutanamide,

(2S)-2-[4-(iodora.ethyl)-2-oxo-l -piperi.dinyl]butanamide,

2-[5 -(i odomethyl)-2-oxo- 1 -azepanyijbutan amide . iii) International Patent Application WO 2004/087658:

A compound having the formula I or a pharmaceutically acceptable salt thereof or stereoisomer! c forms thereof,

wherein

R 1 is hydrogen,

R 2 is hydrogen or Cl-20-aikyi, R 3 is hydrogen, C l-20-alkyl, C4-8-cycioalkyi, C5-8-cycloalkenyl, aryi, aromatic or non aromatic heterocycie, Cl-20-alkoxy, or a group of formula - W-R , R. " ' is hydrogen, Cl-20-alkyl or a group of formula :

or NR 3 R 3a is a group of formula

R is hydrogen, R ' is hydrogen; nitro; halogen; azido; cyano; -S-Cl-4-alkyl; -SO-C1-4- alkyl; -S0 2 -Cl-4-alkyl; -SONH 2 ; Cl-20-aikyl unsubstituted or substituted by halogen; or Cl-20-alkoxy unsubstituted or substituted by halogen,

R 6 is hydrogen, CI -20-alkyl or halogen,

R 7 is hydrogen, CI -20-alkyl or halogen, W is Cl-12-alkylene, -NH- or -NHC(=0)-,

X is O, S or NH,

Y is O, S, -CR 12 R 13 -, -NR. 14 - or -U OK R 8 is aryl or heterocycle,

R 9 , R 10 , R 10a and R l ! are independently selected from hydrogen, Cl-4-alkyL, halogen, hydroxy or methoxycarbonyl, or R 50 and R 10a together form a C3-6-alkylene,

R L: is hydrogen, C l-4-alkyl, halogen or hydroxy, R 13 is hydrogen, or CR 12 R is dioxolanyl,

R 14 is aryl, heterocycle or a group of formula -V-R 15 ,

V is Ci-1 2-aikyiene,

R 15 is aryl or heterocycle, m is 1 to 4, n is 0 or 1 , and at least one of R 5 , R 6 or R ? is different from hydrogen when z is hydrogen, R 3 is H or 2, 6-diisopropylphenyl, and R 3a is H,

In another aspect, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,

wherein

R f is hydrogen, R 2 is hydrogen or C 1 -20-aikyi,

R' is hydrogen, Cl-20-alkyL C4-8-cycioalkyl, C5-8-cycloalkenyl, aryl, aromatic or non aromatic heterocycle, Cl-20-alkoxy. or a group of formula - W~R\

R a is hydrogen, C 1 -20-alkyl or a group of formula: or NR¾ j is a group of formula

R 4 is hydrogen,

R 5 is hydrogen; nitro; halogen; Cl-20-alkyl unsubstituted or substituted by halogen; or Cl-20-aikoxy unsubstituted or substituted by halogen,

R 6 is hydrogen, C l-20-alkyl or halogen,

R ? is hydrogen, C l-20-alkyl or halogen,

W is C l-12-alkylene, -NH- or -NHC(=0)-,

X is O, S or H,

Y is ). S, -CR i : ; ; -. -N 54 - or -C( ( }··.

R 8 is aryl or heterocyele,

R 9 , R Uj , R ]0a and R' 1 are independently selected from hydrogen, C1 -4-alkyl, halogen, hydroxy or methoxycarbonyl, or R 30 and R iJa together form a C3-6-alkylene,

R 1 " is hydrogen, C I -4-alkyl, halogen or hydroxy,

R is hydrogen, or CR 12 R B is dioxolanyi, 14 is aryl, heterocycle or a group of formula -V-R 15 ,

V is Cl-12-alkylene,

R 1 ^ is aryl or heterocycle, m is 1 to 4, n is 0 or 1 , and at least one of R 5 , R 6 or R ? is different from hydrogen when z is hydrogen, R 3 is H or 2,6-diisopropylphenyi, and R ja is H.

The term "alkyl", as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and containing 1-20 carbon atoms, preferably 1-6 carbon atoms and more preferably 1-4 carbon atoms for non-cyclic alkyl and 3- 8 carbon atoms for cycloalkyl. Alliyl moieties may optionally be substituted by 1 to 5 substituents independently selected from halogen, hydroxy, aikoxy, alkoxycarbonyl, ester or alkylamino. Preferred alkyl groups are methyl, ethyl, n-propyl, isopropyl, triiiuoromethyl, n-butyl, 2~ fluoroethyl, 3-hydroxypropyl, 3-hydroxy-2, 2-dimethylpropyi, l-(hydroxymethyl) propyl, 3,3, 3-trifiuoro-2- hydroxypropyl, 3-ethoxypropyl, 2-ethoxy-2~oxoethyl and 3- (dimethylamino) propyl.

The term "cycloalkyl", as used herein, refers to a monovalent group of 3 to 18 carbon atoms, preferably 4-8 carbon atoms, derived from a saturated cyclic or polycyclic hydrocarbon which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group is cycloheptyl.

The term "alkylene", as used herein, represents a divalent alkyl group, having straight or branched moieties, containing 1-12 carbon atoms, preferably 1-6 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred alkylene groups are methylene, ethylene, hydroxyethylene, trimethylene or propylene.

The term "cycioalkenyl", as used herein, is defined as a cyclic unsaturated hydrocarbon radical having at least one double bond, containing 4-20 carbon atoms, preferably 5-8 carbon atoms, and being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycioalkenyl group is 6- (hydroxymethyl) cyclohex-3-en-l-yi.

The term "aryl", as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of 1-3 rings and containing 6- 30 carbon atoms by removal of one hydrogen, such as phenyi and naphthyl each optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, nitro, Cl -6-alkyl, CI -6-aikoxy, Cl-6-alkylsuifonyl, trifluoromethyithio or pyridinyialkyl. Aryl radicals are preferably phenyi radicals. Preferred aryl groups are phenyl, 3-hydroxyphenyl, 3 -fluorophenyl, 3- methylphenyl, 4-methy [phenyi, 4- hydroxyphenyl, 4-hydroxy-3- methoxyphenyl, 3-(2-pyridin-2-ylethyl) phenyl, 3,4- dimethyiphenyi, 4-tert- butyiphenyl, 4-methyisulfonylphenyl, 2-nitrophenyl, 2-chloro- 6-fluorophenyi, 2-[(trifluoromethyl) thio] phenyl, 2-chlorophenyl or 4-bromophenyl.

The term "halogen", as used herein, includes an atom of CI, Br, F, I.

The term "nitro", as used herein, represents a group of the formula -N0 2 .

The term "hydroxy", as used herein, represents a group of the formula -QH.

The term "alkoxy", as used herein, represents a group of formula -OR b wherein R b is a alkyl group, as defined above.

The term "ester", as used herein, represents a group of formula -COQR wherein R c is an aikyl group or an aryl group, as defined above.

The term "alkoxycarbonyi", as used herein, represents a group of formula - COOR d wherein R d is an alkyl group, as defined above. The term "amino", as used herein, represents a group of the formula -NH 2 .

The term "alkylamino", as used herein, represents a group of formula - NHR e or -NR e R f wherein R e and R' are alkyl group as defined above.

The term alkylsulfonyl, as used herein is defined as representing a group of formula -SO 2 -R 8 , wherein R s is Cl-4-alkyl.

The term "heterocycle", as used herein is defined as including an aromatic or non aromatic cycloalkyl or cycloalkenyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring structure and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyi.

Non-limiting examples of aromatic heterocycles are pyrazo!yl, furyi imidazolyl, triazolyl, oxazolyl, pyridinyl, pyrrolyl, thienyl, isothiazolyl, benzimidazolyl, tetrazolyl, isooxazolyl, oxazolyl, thiazolyl, 1,2, 4-thiadiazolyi, oxadiazole, pyridazinyl, pyrimidinyl, pyrazinyl, isoindolyl, triazolopyridinyl, irmdazolopyridirryl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, quinazolinyl, quinolizinyl, naphthyridinyl, quinolyl, isoquinoiyl, isobenzofuranyl, henzothierryl, indolyl, indoiizirryl, purinyl, carbazolyl, thieno (2,3- b) furanyl, thianthrenyl, benzothiazolyl, benzoxazolyl, cinnolinyi, quinoxaiinyl, phenothiazinyl, isochromanyl and xanihenyl, optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cvano, azido, Cl-6-alkoxy, Cl-6-alkylthio, Cl-6-alkyl, Cl-6-haloalkyl, formyl or ester. More preferred aromatic heterocycles are pyrazolyl, furyi, imidazolyl, triazolyl, oxazolyl and pyridinyl.

Non- limiting examples of non aromatic heterocycles are tetrahydro furanyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyl, morpholinyl,

thiomorpholinvi, pyrrolidinvi, thiazolidmyl, indolinyl, tetrahydrobenzazocinyl, dihydroisochromenyl, tetrahydropyranyl, oxooctahydroquinolinyl, dioxolanyl, 1-oxaspiro (4.5) dec-2-yl, pyrrolidinyl, 2-oxo-pyrrolidinyl, 8-thiabicyclo [3.2. 1] cyclooctanyi, 1,4-dithiepanyl, tetrahydro-2H-thiopyraiiyl, azepanyl and azocanyl, optionally substituted by 1 to 5 substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, azido, Cl-6-aikoxy, Cl-6- alkylthio, Ci-6-alkyl, Cl-6-haloalkyl, forn yl or ester. More preferred no aromatic heterocycles are tetrahydrofurany!, piperidinyl, piperidyl, piperazinyi, imidazolidinyl, morpholinyl, thiomorpholinyl, pyrrolidinyl, thiazolidinyl, indolinyl, tetrahydro-l-benzazocin-l (2H)~yl, 3, 4-dihydro H-isochromen-l- yl, tetrahydropyranyl, oxooctahydroquinolinyl and dioxolanyl. The

term"heterocycle"also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cycloaikyi ring, a cycloalkenyi ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an alkylene group, such as quinuclidinyl, 7-azabicyclo (2.2.1 )heptanyl, 7-oxabicyclo (2.2.1)heptanyl and 8- azabicycio (3.2.1 )octanyl.

The term "pyridinylalkyl", as used herein, represents a group of formula - R G - pyridinyl in which R f> is Cl-4-alkylene.

The term "azido" as used herein, represents a group of the formula -N 3 .

The term "cyano" as used herein, represents a group of the formula -CN.

Generally, R 2 is hydrogen or C l-4-alkyl.

Preferably, R" is hydrogen, methyl or ethyl. More preferably, z is hydrogen or methyl

Generally, R J is hydrogen; Cl-6-aikyi unsubstituted or substituted by 1 to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarbonyl or alkylamino; C5-7-cycloalkyl: (hydroxymethyl) cyclohexenyl; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, C1-4- alkyl, hydroxy, methoxy, nitro, methylsulfonyl, trifluoromethylthio or pyridinylalkyl ; pyridinyl unsubstituted or substituted by methoxy; triazolyl ; Cl-4-alkoxy ; or a group of formula -W-R ' wherein: Generally, W is Cl-4-alkylene unsubstituted or substituted by halogen, hydroxy, Cl -4-alkyl or alkoxy ;-NH- ; or- HC { ())- ; and

R s is phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, Cl-4-alkyl, hydroxy, methoxy, nitro, methyisulfoiiyl or trifiuoromethylthio; fur 3. unsubstituted or substituted by methyl; pyrazolyl; pyridinyl; morpholinyi ; tetrahydrobenzazocinyl; piperidinyl unsubstituted or substituted by methyl; dihydroisochrornenyi or dihydroimidazolyl.

Preferably, R J is hydrogen, n-butyl, cyeloheptyl, 2~iiuoroethyl, 3~ hydroxypropyl, 3-hydroxy-2, 2-dimethyipropyl, l-(hydroxymethyl) propyl, 3,3, 3- trifluoro-2-hydroxypropyl, 3-ethoxypropyl, 2~ethoxy-2-oxoethyL 3- (dimethylamino) propyl, 6-(hydroxymethyl) cyciohex-3-en-l-yl, 3- hydroxyphenyl, 3- fluorophenyl, 3- (2-pyridin-2-ylethyl) phenyl, 3, 4- dimethylphenyl, 4-tert-butylphenyl, benzyl, 4-hydroxy-3-methoxybenzyi, 4- methylsulfonylbenzyl, 2-nitrobenzyl, 2-chloro- 6-iluorobenzyl, 2- [(trifluoromethyl) thio] benzyl, 2-hydroxy-2-phenylethyl, 2- (3,4- dimethoxyphenyl) ethyl, 2- (2-chlorophenyi) ethyl, 2- (4-methylphenyl) ethyl, (4- bromophenyl) amino, pyridin-3-yl, 6-methoxypyridin-3-yl, 4H-1 , 2, 4- triazol-3-yl, pyridin-4-yimethyl, (5-methy[-2-furyl) methyl, 3-(lH-pyrazol-l- yl)propyl, 2-morpholin- 4-ylethyl, 2~ ( (3, 4,5, 6-tetrahydro-l-benzazocin-l (2H)-yl) propyl, 2- (2-methylpiperidin-l- yi) ethyl, 3, 4-dihydro-lH- isochromeii- 1 -ylmethy 1, methoxy, (4-pyridinylcarbonyl) amino or 4, 5-dihydro- l H-imidazoi-2-ylamino. More preferably, R J is hydrogen.

Generally, R 3i! is hydrogen, Cl-4-alkyl or a group of formula

wherein m is 1 to 4. Preferably, Rr a is hydrogen, methyl or tetrahydrofuraii-2-ylmethyl. More preferably, R ia is hydrogen.

In another embodiment, NR'R' 3 is piperidinyS unsubstituted or substituted by hydroxy; thiomoqiholi yl; tiiiazolidinyl unsubstituted or substituted by CI- 4- alkoxycarbonyl ; 2, 5 ~dihydro-l H -pyrrol - 1 -yl ; 1 , 4~dioxa~8-azaspiro [4.5] dec-8-yl; 4- oxooetahydro-l(2H)-quinolinyi; or a group of formula wherein R is pyridinyl ; phenyl unsubstituted or substituted by halogen, hydroxy, Cl-4-aikyi ; or a group of formula - V-R l j wherein V is unsubstituted CI -4- aikyiene and R 1" is phenyl or morpholinyl. in a preferred embodiment, NR R ia is 4-pyridin-2-ylpiperazin-l-yl, 4-(3- methyiphenyi) piperazin-l-yl, 4- (4-hydroxyphenyl) piperazin-l-yl, 4- (2- phenylethyi) piperazin-l-yi, 4- (2-morpholin-4-ylethyl) piperazin-l-yl, 3- hydroxypiperidin- 1 -yl, thiornorpholin-4-yl, 4-methoxycarbonyl- 1 ,3-thiazolidin- 3-yl, 2, 5-dihydro-lH-pyrrol-l-yl, 1, 4-dioxa-8-azaspiro [4.5] dee-8-yl or 4- oxooctahydro- 1 (2H)-quinolinyl.

Generally, R 5 is hydrogen, nitro, halogen, Cl-4-alkyl, unsubstituted or substituted by halogen, or Cl-4-alkoxy unsubstituted or substituted by halogen.

Preferably, R J is hydrogen, methyl, ethyl, trifluoromethyi,

trifluoromethoxy, n- propyl, isopropyi, nitro, or halogen. More preferably, R 5 is halogen or trifluoromethyi.

Generally, R 6 is hydrogen, Cl -6-alkyl or halogen.

Preferably, R° is hydrogen, methyl or CI. More preferably, R 6 is hydrogen. Generally, R' is hydrogen, methyl or halogen. Preferably, R '' is hydrogen, methyl, Br, F or CI. More preferably, R 7 is hydrogen, Br or F.

Combinations of one or more of these preferred compound groups are especially preferred.

In a preferred embodiment, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,

wherein R 5 is hydrogen,

R " is hydrogen or Cl-4-alkyS, R 3 is hydrogen; Cl-6-alkyl unsubstituted or substituted by I to 5 substituents selected from halogen, hydroxy, alkoxy, alkoxycarboiiyl or alkylamino ; C5-7-cycloa!ky! ; (hydroxymethy!) cyclohexeny!; phenyl unsubstituted or substituted by 1 to 5 substituents selected from halogen, Cl-4- alkyl, hydroxy, methoxy, nit.ro, methylsulionyi, trirluorom.ethylth.io or pyridinylalkyl ; pyridinyl unsubstituted or substituted by methoxy; triazoiyl;

Cl -4-alkoxy ; or a group of form.ula-W-R s ,

R ja is hydrogen, Cl-4-alkyl or a group of formula

or NR J R a is piperidiny] unsubstituted or substituted by hydroxy;

thiomorpholinyl ; thiazoiidinyl unsubstituted or substituted by C 1 -4- alkoxycarbonyl ; 2,5-dihydro- H-pyrrol- 1 -yl; l,4-dioxa-8-azaspiro [4.5] dec-8- yl; 4-oxooctahydro-l(2H)-quinolinyi ; or a group of formula

is hydrogen, R 3 is hydrogen; nitro; halogen; Cl-4-alkyl, unsubstituted or substituted by halogen; or Cl-4-alkox.y unsubstituted or substituted by halogen,

R6 is hydrogen, Cl-6-allyl or halogen,

R7 is hydrogen, methyl or halogen,

W is C 1 ~4-alkyiene unsubstituted or substituted by halogen, hydroxy, Cl-4- aikyi or alkoxy ;-NH- ; or-NHC { () ;·-.

R8 is phenyl unsubstituted or substituted by 1 to 5 substiluents selected from halogen, Cl-4-aikyi, hydroxy, methoxy, nitro, methylsulfonyl or trifiuoromethylthio ; fur l unsubstituted or substituted by methyl; pyrazolyl; pyridinyl ; morpholinyl; tetrahydrobenzazocinyl ; piperidmyl unsubstituted or substituted by methyl ; dihydroisochromenyi or dihydroimidazolyl,

R 14 is pyridinyl; phenyl unsubstituted or substituted by halogen, hydroxy, Cl-4-alkyl ; or a group of formula- V-R 15 ,

V is unsubstituted Cl-4-alkylene,

R f 5 is phenyl or morpholinyl, m is 1 to 4, and at least one of R 5 , R h or R 7 is different from hydrogen when R is hydrogen, R J is H or 2,6-diisopropylphenyi, and R Ja is H. In a more preferred embodiment, the compound has the formula I or a pharmaceutically acceptable salt thereof or stereoisomeric forms thereof,

wherein 1 is hydrogen,

R 2 is hydrogen, methyl or ethyl,

R J is hydrogen, n-butyi, cycloheptyl, 2-fluoroethyl, 3-hydroxypropyl, 3- hydroxy-2,2- dimethyipropyl, 1 -(hydroxymethyS) propyl, 3,3, 3~trifruoro~2- hydroxypropyl, 3- ethoxypropyl, 2-ethoxy-2-oxoethyl, 3- (dimethylamino) propyl, 6- (hydroxymethyl) cyclohex-3-en- 1 -yl, 3-hydroxyphenyl, 3- fluorophenyl, 3- (2-pyridin-2-ylethyl) phenyl, 3, 4-dimethy [phenyl, 4-tert- butylpherryl, benzyl, 4-hydroxy-3- methoxyhenzyl, 4-methylsulfonylbenzyl, 2- nitrobenzyi, 2-chloro-6-fluorobenzyl, 2- [(trifluoromethyl)thio] benzyl, 2- hydroxy-2-phenylethyl, 2~ (3, 4-dimethoxyphenyl) ethyl, 2- (2-chlorophenyl) ethyl, 2- (4-methylphenyl) ethyl, (4-bromophenyS) amino, pyridin-3-yl, 6- methoxypyridin-3-yl, 4H- 1 ,2,4-triazol-3-yi, pyridin-4-ylmethyl, (5-methyl-2- furyl) methyl, 3- (IH-pyrazol-l -yl) propyl, 2-morpholin-4-ylethyl, 2- ( (3, 4,5, 6-tetrahydro- 1-benzazocin-l (2H) -yl) propyl, 2- (2-methylpiperidin-l-yl) ethyl, 3, 4-dihydro-lH- isochromen-l -ylmethyl, methoxy, (4-pyridmylcarbon l) amino or 4, 5-dihydro-lH- imidazol-2-ylamino,

R a is hydrogen, methyl or tetrahydrofuran-2-ylmethyl, or R'R Ja 4- pyridm-2-ylpiperazm- 1 -yl, 4-(3-methylphenyi) piperazin-1 -yl, 4-(4- hydroxyphenyl) piperazin-l-yl, 4-(2-phenylethyl) piperazin-1 -yl, 4-(2- morpholin-4- ylethyl) piperazin-l-yl, 3-hydroxypiperidin-l-yL thiomorpholin- 4-yl, 4- methoxycarbonyl-1, 3-thiazolidin-3-yl. 2, 5 -dihydro- lH-pyrrol-l-yl, 1 ,4-dioxa-8- azaspiro [4.5]dec-8-yl or 4-oxooctahydro-l(2H)-quinolinyl,

R 4 is hydrogen,

R5 is hydrogen, methyl, ethyl, trifluoromethyl, trifSuoromethoxy, n-propyl, isopropyl, nitro or halogen,

R° is hydrogen, methyl or CL

R' is hydrogen, methyl, Br, F or CL and at least one of R 3 , R 6 or R' is different from hydrogen when R ~ is hydrogen, R J is H or 2,6-diisopropylphenyl, and R 3tl is H. More preferably, R 2 is hydrogen or methyl, R J is hydrogen, R 3a is hydrogen, R 5 is halogen or trifluoromethyl, R° is hydrogen and R is hydrogen, Br or F.

In all the above-mentioned scopes, when R z is Cl-20-alkyl, the carbon atom to which R 2 is attached is preferably in the"S"-configuration. In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5-iodo-2-oxo- 2,3-dihydro-l H-indol-l- yl) acetamide ; 2- (5-chloro-2-oxo-2, 3-dihydro-lH- indol-l-yl) acetamide ; 2- (5, 7-dibromo- 2-oxo-2, 3-dthydro-lH-indol- l-yl) acetamide ; 2-(5-mtro-2-oxo-2,3-dihydro-lH-indol-l- yl) acetamide ; 2-(5- metiiyl-2-oxo-2 ,3 -dihydro- 1 H-indol- 1 -yl) acetamide; 2- (5-chloro-2- oxo-2, 3- dihydro-lH-indol-l-yl) propanamide ; (2R)-2- (5-chloro-2-oxo-2, 3-dihydro- 1H- indol-l-yi) propanamide ; (2S)-2-(5-chloro-2-oxo-2,3-dihydro-l Fi-mdol-l - yl) propanamide; 2-[2-oxo-5-(trifluoromethoxy)-2, 3-dihydro-lH-indol- 1 -yl] acetamide ; 2~ (5-isopropyl-2-oxo-2, 3 -dihydro- 1 H-indol- 1 -yl)acetamide ; 2- (5-ethyl-2-oxo-2, 3-dihydro- 1 H-indol- 1 -yl) acetamide ; 2-(5-fluoro-2-oxo-2,3- dihydro-l H-indol-l-yl) acetamide; 2- (5,7-dimethyl-2-oxo-2, 3-dihydro-lH- indol-l-yl) acetamide; 2- (5-bromo-2-oxo-2, 3- dihydro-lH-indol-l-yl) acetamide ; 2-(2-oxo-5-propyl-2, 3-dihydro-lH-indol-l - yl) acetamide ; 2-[2- Qxo-5-(trifluoromethyl)-2, 3-dihydro-lH-indol-l-yl] acetamide ; 2- (5, 6- dimethyl-2-oxo-2, 3-dihydro-lH-indol-l -yl) acetamide; 2- (7-chloro-2-oxo-2, 3-dihydro- IH-indol-l-yl) acetamide; 2- (6-chloro-2-oxo-2, 3-dihydro-IH- indol-l-yi) acetamide; 2- (5- chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl) butanamide ; ( ) - 2- (5-chloro-2-oxo-2, 3- dihydro-lH-indol-l-yl) butanamide; (-)~2~ (5-chloro-2-oxo-2, 3-dihydro-IH-indol-l- yl) butanamide; 2-(5-methyl-2- oxo-2,3-dihydro-l H-indo[-l-yl)propanamide ; (+)-2- (5- methyl-2-oxo-2, 3- dihydro-l H-indol-l- l) propanamide; (~)-2- (5-methyl-2-oxo-2, 3- dihydro-lH- indol-l-yi) propanamide ; 2-(5-brorno-2-oxo-2,3-dihydro-lH-indol-l - yl) propanamide ; (-)-2- (5-bromo-2-oxo-2, 3-dihydro-lH-indol-l-yl) propanamide ; (+)-2- (5-bromo-2-oxo-2, 3-dihydro-l H-indol-l -yl) propanamide; 2- (5- chloro-7-fluoro-2-oxo- 2, 3 -dihy dro- 1 H-indol- 1 -yl) acetamide; 2-(5-chloro-2- oxo-2,3-dihydro-lH-indol-l -yl)-N- (3-hydroxyphenyi) acetamide ; 2- (5- chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl)-N- (3- fluorophenyl) acetamide ; 2- (5-chloro-2-oxo-2, 3~dihydro-lH-mdol-l ~yI)~N- [3- (2-pyridin- 2-ylethyl) phenyllacetarnide ; 2-(5-chloro-2-oxo-2,3-dihydro-l H-indol- l-yi)-N-[6- (b y dro ymeth 3 ) cyclohex-3 -en- 1 -yl] acetan aide ; 5 -cbloro- 1 - [2-oxo-2~(4- pyridin-2- ylpiperazin-l-yl) ethyl3-l , 3-dihydro-2H-indoi-2-one ; 5-chioro-l - {2- [4- (3- methylphenyl) piperazin~l-yl]-2-oxoetiiyl}-l , 3-dihydro-2H-indol- 2 -one ; 2- (5-ch3oro-2- oxo-2, 3 -dihy dro- 1 H-indol- 1 -yl)-N-(4-hy droxy-3- methoxybeiizyl)acetamide ; 2- (5-chloro- 2-oxo-2, 3-dihydro-lH-indol-l-yl)-N- (pyridin-4-ylniethyl)-N- (tetrahydrofuran-2- yimethyl) acetamide ; 5-chloro- 1- [2-(3-hydroxypiperidin- 1 -yl)-2-oxoethyl]~ 1 ,3-dihydro- 2H-indol-2-one; 2-(5- ch3oro-2-oxo-2, 3-dihydro- 1 H-indol- l-yl)-N'- isonicotinoylacetohydrazide ; 5- chloro-l -(2-oxo-2-thiomorpholin-4-ylethyl)-l , 3-dihydro- 2H-indol-2-one; 2-(5 chloro-2-oxo-2 , 3-dih dro- 1 H-indol- 1 -yl)-N-(4H - 1 , 2, 4~triazoi-3 - yl) acetamide; 2- (5-chloro-2-oxo-2, 3-dihydro-IH-indol-l -yl)-N- [4- (methylsulfonyl) benzyl] acetamide ; l-[(5-chloro-2-oxo-2,3-dihydro-lH-indol 1 - yl) acetyl] octahydroquinolin-4 (lH)-one ; N ! - (4-bromophenyl)-2- (5- chloro-2-oxo-2, 3- dihydro-lH-indol- 1 -yl) acetohydrazide; 2-(5-chloro-2-oxo- 2,3-dihydro-l H-indol-l-yl)-N- (6-methoxypyridin-3-yl) acetamide; N-butyl-2- (5 -chloro-2-oxo-2, 3 -dihy dro- 1 H-indol- 1- yl) acetamide ; 2-(5-chioro-2-oxo- 2,3-dihydro-lH-mdol-l-yl)-N-(3- hydroxypropyl) acetamide : 2-(5-chloro-2- oxo-2,3-dihydro-l H-indoi-l-yl)-N- [3- (dimethylaniino) propyl] acetamide ; 5- chloro- 1 - {2-oxo-2[4-(2-phenylethyl)pperazin- 1 - yl] ethyl} -1, 3-dihydro-2H~ indol-2-one; ethyl {[(5-chloro-2-oxo-2, 3-dihydro-lH-indol-l- yl)

acetyl]amino } acetate ; 2-(5-chloro-2-oxo-2,3-dihydro- 1 H-indol- 1 -yi)-N-(3- ethoxypropyl) acetamide ; 2~ (5-chloro-2-oxo-2, 3-dihydro-lH-mdol-l -yl)-N- (2- tluoroethyl) acetamide ; 2- (5-chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl)-N- methoxy-N- methylacetamide ; 2-(5-chloro-2-oxo-2,3-dihydro-lH-indol-l-yl)- N-(3, 4- dimethylphenyl) acetamide ; N- (4-tert-butylphenyl)-2- (5-chloro-2~ oxo-2, 3-dihydro-lH- iiidol-l-yl) acetamide; 2- (5-chloro-2-oxo-2, 3-dihydro- 1.H-indol- l -yl)-N- (3-hydroxy-2, 2- dimethyipropyl) acetamide ; 2-(5-chloro-2- oxo-2,3-dihydro-lH-mdol-l-yl)-N-[l- (hydroxymethyl) propyl] acetamide : 2- (5-chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl)-N- (3,3, 3-trif!uoro-2- hydroxypropyl) acetamide; 2- (5-chloro-2-oxo-2, 3-dihydro-lH-indol-l- yl)-N- (2-hydroxy-2-phenylethyl) acetamide ; 5-chioro-l- {2- [4- (4- hydroxyph nyl) piperazin-l-yl]-2-oxoethyl} -l, 3-dihydro-2H-indol-2-one; 2- (5-chioro-2- oxo- 2, 3-dihydro-lH-indol-l -yl)-N-(pyridin-4-ylmethyl)acetamide ; 2- (5-ch.io.ro-2- oxo- 2, 3-dihydro-lH-indol-l-yl)-N-[(5-methyl-2-fur}4)methyl]acetami de ; 2- (5-chloro-2-oxo- 2, 3-dihydro-lH-indol-l-yl)-N- [3- (lH-pyrazol-l-yl) propyl] acetamide ; methyl 3- [ (5- chioro-2-oxo-2, 3-dihydro-l H-indoi-l-yl] acetyl]- 1, 3-thiazolidine-4-carboxylate ; 5- chloro-l-[2-(2, 5-dihydro-lH-pyrrol-l-yl)-2- oxoethyS]-!, 3-dihydro-2H-indol-2-one; 2- (5- chloro-2-oxo-2, 3-dihydro-!H- indol-l-yl)-N'- (4, 5-dihydro-lH-imidazol-2- yl) acetohydrazide ; 2- (5-chloro- 2-0X0-2, 3-dihydro-lH-indo3.-l-yl)-N- [2- (3, 4- dimethoxyphenyl.) ethyl] acetamide ; 2-(5-chloro-2-oxo-2,3-dihydro- 1 H-indol- 1 -yI)-N-[2- (2- chiorophenyi) etl-lyllacetaniide ; 2-(5~chioro~2-oxo-2,3-dihydro-lH-indoi-l~ yl)-N-[2-(4- methylphenyl } ethyl] acetamide ; 2-(5-chloro-2-oxo-2,3-dihydro- 1 H-indol- l-yl)-N-(2- morpholin-4-ylethyl) acetamide : 2- (5-chloro-2-oxo-2. 3- dihydro-lH-indol-l-yl)- - [2- (3,4, 5, 6-tetrahydro-l -benzazocin-l (2H) -yl) propyl] acetamide ; 2- (5-chloro-2-oxo-2, 3- dihydro-lH-indol-l-yl)-N-[2-(2- methylpiperidin-l-yi) ethyl] acetamide ; 2- (5-chloro-2- oxo-2, 3-dihydro-iH- indol-l-yl)-N-(2-mtrobenzyl) acetamide ; 2- (5-chloro-2-oxo-2, 3- dihydro-lH- indol-l-yl)-N- (3, 4-dihydro-lH-isochromen-l-ylinethyl) acetamide ; N- (2- chloro-6~fiuorohenzyl)~2~ (5-chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl) acetamide : N- benzyi-2 (5-chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl)-N- methylacetamide ; 2- (5-chloro- 2-oxo~2, 3-dthydro-lH-vndol-l-yl)-N-{2- [(trifluoromethyl) thio] benzyl} acetamide ; 5- chloro-1 - [2- ( 1, 4-dioxa-8- azaspiro [4,5] dec-8-yl)-2-oxoethyl]-l , 3-dihydro-2H-indol-2- one; 2-(5-chloro- 2-0X0-2, 3-dihydro-lH-indol-l-yl)-N-cycloheptylacetamide ; 5-chloro-l- {2- [4- (2-morpholin-4-ylethyl) pipera.zm-1 -yl]~2-oxoetbyl}-l, 3-dihydro-2H- indol-2-one ; and 2-(5-chloro-2-oxo-2,3-dihydro-l H-mdol-l-yl)-N-pyridin-3- ylacetamide.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: 2-(5~iodo~2-oxo- 2.3-dihydro-lH-indol-l- yl) acetamide ; 2- (5-chloro-2-oxo-2, 3-dihydro-lH- indol-l-yl) acetamide ; 2- (5, 7-dibromo- 2-oxo-2, 3-dihydro-l H-indoi-l- yl)acetamide ; (2S)-2-(5-chioro-2-oxo-2,3-dihydro-lH- indol- l-yl)

propanamide ; 2-[2-oxo~5-(triiluoromethy])-2, 3~dihydro-lH~indoi-l ~ yl] acetamide and 2-(5-chloiO-7-fluoro-2-oxo-2,3-dihydro-lH-indol-l-yl) acetamide.

In another embodiment, compounds useful in the methods and

compositions of this invention are selected from the group consisting of: 2- (5- chloro-2-oxo-2, 3-dihydro-lH-indol-l-yl) acetamide and (2S) -2- (5-chioro-2- oxo-2, 3 -dihydro-lH -indo 1 - 1 -yl ) propanamide . iv) US Patent No. 7,244,747:

A compound having the formula I or a pharmaceutically acceptable salt thereof.

wherein R is hydrogen, C 1-20 alkyl, C 3- s cycloa!ky!, halogen, hydroxy, aikoxy, aryloxy, ester, amido, cyano, nitro, amino, guanidine, amino derivative, alkylthio, aryithio, alkylsulibnyl, arylsulfonyl, alkylsulfmyl, arylsulfmyl, aryl or heteroeycle;

R 2 is hydrogen, CV 2 0 alkyl, aikoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;

R J is hydrogen, Cj- 20 alkyl, aikoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl; or R _ and R J can form together with the imidazole ring the following 1 H benzimidazole cvcle

R 4 is hydrogen, C 1 . 2 0 alkyl, C2-12 alkenyl,€ 2 - 12 alkynyl, aryl, azido, aikoxycarbonylamino, arylsulfonyloxy or heierocycie;

R 4 is hydrogen or Cj -20 alkyl; or R 4 and R 4d can form together a C 3-8 cycloalkyl; R 5 is hydrogen: or R 4 , R 4d and R 5 can form together with the 2-oxo-i -pyrrolidine ring the following l ,3-dihydro-2H-indol-2-one cycle

R° is hydrogen or C 1-20 alkyl;

R is hydrogen; or R° and R/ are linked together to form a C 3-6 cycloalkyl;

R 8 is hydrogen, halogen, nitro, cyano, C1-20 alkyl or alkoxy;

R 9 is hydrogen, Cj.2o alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonvl, arylsulfonyl, alkylsuiflnyl or arylsulfinyl;

R 10 is hydrogen, Ci-20 alkyl, halogen, hydroxy, alkoxy, aiyloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsulfonvl, arylsulfonyl, alkylsuiflnyl or arylsulfinyl;

R 11 is hydrogen, halogen, nitro, cyano, C 1-2 o alkyl or alkoxy;

R L: is hydrogen or halogen;

R° is hydrogen, nitro, halogen, heterocycle, amino, aryl, C 1-20 alkyl unsubstituted or substituted by halogen, or alkoxy unsubstituted or substituted by halogen;

R 14 is hydrogen, C 1-2 o alkyl or halogen;

R lj is hydrogen, C 1-20 alkyl or halogen; with the proviso that R 4 is different from hydrogen when represents a group of formula

The asterisk * indicates the point of attachment of the substituents.

In a preferred embodiment, the compounds have the formula I, their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,

wherein R 5 is hydrogen, C 1-2 o alkyl, C 3-8 cycloalkyl, halogen, hydroxy, ester, amido, cyano, nitro, amino, guanidine, alkylthio, alkylsulfonyl, aikyisulfinyl, aryS or heterocycle;

R 2 is hydrogen, C 1 -2 o alkyl, halogen, cyano, ester, carbamate or amido;

R J is hydrogen, cyano, C 1-2 o alkyl, halogen or ester; or R 2 and R 3 can form together with the imidazole ring the following 1H- benziniidazo!e cycle

R is hydrogen, C O alkyl, C 2-12 aikenyl or aiyl; R 4a is hydrogen; R 3 is hydrogen; or R 4 , R 4a and R* can form together with the 2-oxo-i -pyrrolidine ring the ibllowing l,3-dihydro-2H-indol-2-one cycle

R ' is hydrogen or C 1-20 alkyl;

R 7 is hydrogen: or R 6 and R' are linked together to form a C3-0 cycloalkyl; R l is hydrogen;

R 9 is hydrogen, d- o alkyl, halogen or alkoxy; R 10 is hydrogen, Ci -20 alkyl, halogen or cyano; R is hydrogen;

R L: is hydrogen or halogen;

R 13 is hydrogen, halogen, heterocycle or C 2 alkyl; ' is hydrogen:

R 1 ^ is hydrogen: with the proviso that R 4 is different from hydrogen when

represents a group of formula

The term "alkyl", as used herein, represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more pre preferred alkyl groups have 1 -3 carbon atoms. Alky] moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkythio, alkanoylamino, arylcarbonylamino, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyi or aryl. Usually alkyl groups, in the present case, are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1 -ethylpropyl, n-heptyl, 2,4,4-trimethylpentyl, n-decyl, chloromethyl, trifl oromethyl, 2-bromo~2,2-difi.uoroethyl, 2,2,2-trifluoroethyl, 3,3,3- trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl,

(acetylamino)methyl, (propionylammo)methyl, (benzoyl amino)methyl, (4- chlorophenoxyjmethyl, benzyl, 2-phenylethyl or 2-(methyithio)ethyl. Preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1- ethylpropyl, 2,4,4-trimethylpentyl, chloromethyl, trifluoromethyl, 2,2,2- trifluoroethyl, hydroxymethyi, cyanomethyl, azidomethyl,

(acetylaniino)methyl, (propionyiaiTimo)methyl, (benzoyl amino)methy! or 2- (methylthio)ethyl. More preferred alkyl groups are methyl, ethyl, n-propyl, i- propyl, n-butyi, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.

The term "cycloalkyl", as used herein, represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl groups are cyclopropyl and cyclohexyl.

The term "alkenyi" as used herein, represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof ha ving at least one carbon-carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms. Alkenyi groups are being optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Usually an alkenyi group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens. Preferred alkenyi group, in the present case, is 2, 2-difluorovinyl.

The term a"alkynyl" as used herein, represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon- carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably an alkynyl group is a halogenoalkynyl group

(h aloalkynyl group) .

Groups qualified by prefixes such as "s", "i", "t" and the like (e.g. "i- propyl", "s-butyl") are branched derivatives.

The term "aryl" as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cya.no, alkoxy, aikyithio, C1.3 alkyl or azido, preferably halogen or azido. Usually ary groups, in the present case are phenyl, 3-chlorophenyl, 3-fluorophenyl, 4-chlorophenyl, 4 -fluorophenyl, 3 ,4-difluorophenyl, 3 ,5 -difluorophenyl, 3 -chloro-4- fluorophenyl, 2,3,4-trifiuorophenyl, 2,4,5-trifluorophenyl, 2,3,5- trifluorophenyl, 3,4,5-trifluorophenyl, 3-azido-2,4-difluorophenyl or 3-azido- 2,4,6-trifl orophenyl. Preferably, aryl groups are phenyl, 3-chlorophenyl, 3- fluorophenyl, 4-chlorophenyl, 4-fluorophenyL 3 ,4-difluorophenyl, 3,5- difluoropheny 1 , 3 -chl oro-4-fluoropheny ί , 2,3 ,4-trifl uorophenyl, 2 ,4,5 - trifluorophenyS, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2,4- difluorophenyl. Most preferred aryl groups are phenyl, 3-chlorophenyl, 3- fluorophenyl, 3,5-difluorophenyl, 2,3,4-trifluorophenyl, 2,4,5-trifluorophenyl, 2,3,5-trifluorophenyl, 3,4,5-trifluorophenyl or 3-azido-2.4-difluorophenyl.

The term "heterocycie", as used herein, is defined as including an aromatic or non aromatic cycioalkyl moiety as defined above, having at least one O, S and/or N atom interrupting the carbocyclic ring stmcture. Heterocyclic ring moieties can be optionally substituted by alkyl groups or halogens and optionally, one of the carbon of the carbocyclic ring stmcture may be replaced by a carbonyl, Usually heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyi, 3-furyl, 2-thienyl, 3-thienyl, 2-tetrahydrofuranyl, lH-pyrrol-2-yl, 1 -methyl- 1.H- pyiTol-2-yl, 1 H-pyrazol-2-yl, 1 H-pyrazol-3 -yl, 4-chloro- 1 -methyl- 1 H-pyrazol- 3-yi, 5-chioro-l,3-dimethyl-lH-pyrazoi-4-yl, l,2,3-thiadiazol-4-yl, 3,5- dimethyl-4-isothiazyl, lH-imidazol-2-yl, 1 -methyl- 1 H-imidazol-2-y 1, 4- methyl-1 H-imidazol-5-yl, or 2-methyi-l ,3-thiazol-4-yl. Preferred heterocycles are 1 H-imidazol-2-yI, l,2,3-thiadiazol-4-yl, 1 H-pyrazol-3 -yl, 2-furyl, 3-fur l, 2-thienyl, 1 -methyl- 1 H-pyrrol-2-yl, 1 H-pyrrol-2-yl ,

The term "halogen", as used herein, includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine. Preferred halogens are fluorine, bromine and chlorine.

The term "hydroxy", as used herein, represents a group of formula—OH.

The term "alkoxy", as used herein, represents a group of formula -OR a wherein R a is an alkyl group, as defined above, Preferred alkoxy group is methoxy.

The term "aryloxy", as used herein, represents a group of formula—OR 0 wherein R b is an aryl group, as defmed above. Preferred aryloxy group is phenoxy.

The term "ester", as used herein, represents a group of formula— COOR wherein R c is an aikyi group or aryl group, as defined above. Preferred ester group is methoxycarbonyl.

The term "amido", as used herein, represents a group of formula --CONH 2 .

The term "amino", as used herein, represents a group of formula— NH 2 .

The term "aminoderivative", as used herein, represents an alkylamino or an arylamino group, wherein the terms "alkyl" and "aryl" are defmed as above.

The term "cyano", as used herein, represents a group of formula— CN.

The term "nitro", as used herein, represents a group of formula -N0 2 .

The term "azido", as used herein, represents a group of formula ~N 3 .

The term "guanidine", as used herein, represents a group of formula— NHC(=NH) H 2 .

The term "alkylthio", as used herein, represents a group of formula— SR d wherein R d is an alkyl group, as defined above. Preferred alkylthio group is methylthio.

The term "alkylsulfonyl", as used herein, represents a group of formula— wherein R e is an alkyl group, as defined above. Preferred alkylsulfonyl group is methylsuifonyl. The term "alkyisulfinyl", as used herein, represents a group of formula - S(=0) t wherein R 1 is an alkyl group, as defined above. Preferred alkyisulfinyl group is methylsulfinyl.

The term "arylthio", as used herein, represents a group of formula— SR g wherein R s is an aryl group, as defined above.

The term "arylsulfony l", as used herein, represents a group of the formula— wherein R h is an aryi group, as defined above.

The term "arylsulfinyi", as used herein, represents a group of the formula ~ S(=0)R ! wherein R 1 is an aryl group, as defined above.

The term "carbamate" as used herein, represents a group of formula— (H)C(0)()R j , wherein R J is an alkyl or an aryl, as defined above. Usually carbamate groups are (propoxycarbonyl)amino or (benzyloaxycarbonyl)amino. Preferred carbamate group is (benzyloaxycarbonyl)amino.

The term "alkanoylamino" as used herein, represents a group of the formula ~NHC(=0)R K: wherein R K is an alkyl group, as defined above.

The term "( ai icarbonyl)amino" as used herein, represents a group of the formula ~NHC(=0)R. m wherein R m is an aryl group, as defined

above. Preferred (arylcarbonyl)amino is benzoylamino.

Usually, R 1 is hydrogen; C 1-10 alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chloropheiiQxy; hydroxy; C 3-6 cycloaikyi; halogen; ester; ami do; nitro; cyano; amino; phenyl; alkylthio;

aikyisulfonyl; alkyisulfinyl; heterocycle unsubstituted or substituted by alkyl groups; or guanidine. Preferably, R 1 is hydrogen; methyl; ethyl; i-propyi; n- propyl; cyclopropyi; n-butyl; i-butyl; t-butyl; 1 -ethylpropyl; 2,4,4- trimethylpentyl; hydroxymethyi; chloromethyl; triiluoromethyl; 2,2,2- trifluoroethyl; cyanomethvl; 2-(methylthio)ethyl; chloro; bromo; nitro; cyano; amino; aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl;

methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1 H-pyrrol-2-yl; 1 -methyl- IH-pyrrol- 2-yl; 2-thienyl; lH-pyrazol-3-yl; l,2,3-thiadiazol-4-yl or lH-imidazol-2-yl, More preferably, R f is hydrogen; methyl; ethyl; i -propyl; n-propyl; n-butyl; niethylthio; nitro; cyano; amino; chloro or lH-pyrrol-2-yl. Most preferably, R l is hydrogen; methyl; niethylthio; nitro; cyano; amino or chloro.

Usually, R 2 is hydrogen; C alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate; [(N- methoxy-N~methyl)amino]carbonyl. Preferably, R ~ is hydrogen; methyl;

hydroxymethyl; (acetylamino)methyl; (propionylamino)methyl;

(benzoylamino)methyl; [(benzyloxy)carbonyl]a.miiio; chloro or cyano. More preferably, R 2 is hydrogen; chloro or cyano.

Usually, R' is hydrogen; C M alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano. Preferably, R J is hydrogen; hydroxymethyl; chloro; cyano. More preferably, R 3 is hydrogen or cyano. Most preferred R' is hydrogen.

Usually, R 4 is hydrogen; CM alkyl unsubstituted or substituted by halogens; C 2- alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens. Preferably, R 4 is hydrogen; n-propyl; 2,2- difluorovinyl; phenyl; 3-ehloropherryl; 3 -fluorophenyl; 4-chlorophenyl; 4~ fluorophenyl; 3,5-difluorophenyl; 3, 4-di fluorophenyl; 3-chloro-4-fiuorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluoropheny ; 3,4,5- trifluorophenyl; 3-azido-2,4-difluorophenyl or 3-azido-2,4,6 rifluorophenyl. More preferably, R 4 is hydrogen; n-propyl; 2,2-difluorovinyl; phenyl; 3- chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4-fiuorophenyl; 3,5- difluorophenyl; 3,4-difluorophenyi; 3-chioro-4-fluorophenyl; 2,3,4- trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-triiiuorophenyl; 3,4,5- trifluorophenyl or 3-azido-2,4-difluorophenyi. Most preferably, R 4 is n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3-fluorophenyl; 3,5-difluorophenyl; 2,3,4-trifluorophenyl; 2,4,5-trifluorophenyl; 2,3,5-trifluorophenyl; 3,4,5- trifluorophenyl or 3-azido-2,4-difluorophenyl.

Usually, R 4a is hydrogen. Usually, R 3 is hydrogen.

Usually, R° is hydrogen or C O alkyl unsubstituted or substituted by hydroxy or azido. Preferably, R 6 is hydrogen or azidomethyl. More preferably R 6 is hydrogen.

Usually R' is hydrogen.

In other preferred embodiments, R 6 and R' are linked to form a cyclopropyL

In other preferred embodiments, R 2 and R J can form together with the imidazole ring the following lH-benzimidazole cycle

Usually, R x is hydrogen.

Usually, R 9 is hydrogen; halogen; C 1-3 alkyl or alkoxy. Preferably, R is hydrogen; methyl; chloro or methoxy. More preferred R ' ' is hydrogen.

Usually, R lU is hydrogen; halogen; cyano; C 1-3 alkyl unsubstituted or substituted by halogens; or alkoxy. Preferably, R ! ° is methyl; hydrogen; trilluoromethyl; fluoro; cyano or methoxy. More preferred R 10 is hydrogen; trifluoromethyl; fluoro or cyano.

Usually, R 1 1 is hydrogen.

In other preferred embodiments, R 4 , R ¾! and R 3 can form together with the 2-oxo-l -pyrrolidine ring the following l,3-dihydro-2H-indol-2-one cycle

Usually, R is hydrogen or halogen. Preferably R l _ is hydrogen; chloro or fluoro. More preferred R" : is hydrogen,

Usually, R i3 is hydrogen; Ci- 3 alkyl; halogen or thiazolyi unsubstituted or substituted by alkyl groups, such as methylthiazolyl. Preferably R 13 is hydrogen; chloro; bromo or methyl. Most preferred R ¾ J is chloro; bromo or methyl.

Usually R 14 is hydrogen,

Usually, R 13 is hydrogen.

Combinations of one or more of these preferred compound groups are especially preferred.

Generally , among the embodiments, the compounds of formula I, or pharmaceutically acceptable salts thereof, are those wherein

R 1 is selected from hydrogen; C J - IO alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy; C 3 . 6 cycloaikyi; halogen; ester; amido; nitro; cyano; amino; phenyl; alkylthio; alkylsulfonyl; alkylsulfinyl; heterocycle unsubstituted or substituted by alkyl group; or guanidine;

R 2 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [ -methoxy-N-niethyl)amino]carbonyl. R J is selected from hydrogen; C alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano;

R 4 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by halogens; C 2-4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or /and halogens;

R is hydrogen;

R " is hydrogen;

R 6 is selected from hydrogen or Cj -10 alkyl unsubstituted or substituted by hydroxy or azido;

R 7 is hydrogen; or R 6 and R' can be linked to form a cyclopropyi; or R " and R 3 can form together with the imidazole ring the following 1 H- benzimidazoie cycle

R l is hydrogen:

R 9 is selected from hydrogen; halogen; C1-3 alkyl; alkoxy;

R llJ is selected from hydrogen; halogen; cyano or Cj.3 alkyl unsubstituted or substituted by halogens; or alkoxy;

R ! l is hydrogen; or R 4 , R 4d and R 5 can form togetlier vvith the 2-oxo-i -pyrrolidine ring the following l ,3-dihydro~2H-mdol-2-one cycle

R l is selected from hydrogen or halogen:

R 1J is selected from hydrogen; C 1-3 alkyl; halogen; thiazoiyl unsubstituted or substituted by alkyl groups, such as methylthiaz'

R 14 is hydrogen;

R 15 is hydrogen; ith the proviso that R 4 is different i om hydrogen when

represents a group of formula

In a preferred embodiment, the compounds of formula 1, or

pharmaceutically acceptable salt thereof, are those wherein R 1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyi;

cyclopropyi; n-butyl; i-butyl; t-butyl; 1-ethylpropyl; 2,4,4-trimethylpentyl; trifluoromethyl; 2,2,2-trifluoroethyl; hydroxymethyl; chloromethyl;

cyanomethyl; 2-(methylthio)ethyi; chloro; bromo; nitro; cyano; amino;

aminocarbonyl; methoxycarbonyl; methylthio; methylsulfinyl; methylsulfonyl; phenyl; 2-furyl; 3-furyl; 1 H-pyrrol-2-yl; 1 -methyl- l H-pyrrol-2-yl; 2-thienyl; lH-pyrazol-3-yl; l,2,3-thiadiazol-4-yl; or lH-imidazol-2-yi;

R 2 is selected from hydrogen; methyl; hydroxymethyl;

(acetyiamino)methyi; (propiony3.amino)methyl; (benzoyiamino)methyi;

(benzyloxycarbonyl)amino; chioro; or cyano;

R' is selected from hydrogen; hydroxymethyl; chloro; cyano; or R _ and R J can form together with the imidazole ring the following benzimidazole cycle

R is hydrogen;

R 9 is selected from hydrogen; methyl; choro; methoxy;

R 10 is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy;

R 1 * is hydrogen;

R 4 is selected from hydrogen; n-propyl; 2,2-diiluorovinyl; phenyl; 3- chiorophenyi; 3-fluorophenyl; 4-chlorophenyl; 4-fiuorophenyi; 3,5- difluorophenyl; 3,4-difiuorophenyl: 3-chloro-4-fluorophenyl; 2,3,4- tri fluorophenyl; 2,4,5-trifiuorophenyl; 2,3,5-trifluorophenyl; 3,4,5- trifluorophenyl; 3-azido-2,4-difluorophenyl; or 3-azido-2,4,6-trifluoropheiiyl.

R ',a is hyd.rogen;R 5 is hydrogen; or R\ R 4a and R 5 can form together with the 2-oxo-i -pyrrolidine ring the following l ,3-dihydro-2H-indol-2-one cycle

R 12 is selected from hydrogen; chloro; f!uoro;

R 13 is selected from hydrogen; chloro; bromo; methyl;

R 14 is hydrogen;

R 1" ^ hydrogen;

R 6 is selected from hydrogen; azidomethyl;

R 7 is hydrogen;

or R (1 and R ? are linked to form a cyclopropyi;

with the proviso that R 4 is different from hydrogen when represents a group of formula

In a more preferred embodiment, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein

R 1 is selected from hydrogen; methyl; ethyl; i-propyl; n-propyi; n- butyl;methylthio; nitro; cyano; amino; chloro; or lH-pyrrol-2-yl;

R 2 is selected from hydrogen; chloro; cyano;

R' is selected from hydrogen; cyano; or R ~ " and R J can form together with the imidazole ring the followin henzimi dazole cycle

R° is hydrogen; R 9 is hydrogen;

R lu is selected from hydrogen; trifluoromethyl; fluoro; cyano; R 11 is hydrogen;

R 4 is selected from hydrogen; n-propyl; 2,2-difiuorovinyl; phenyl; ch!orophenyi; 3-fluorophenyl; 4-chlorophenyl; 4-fiuorophenyi; 3,5- difluorophenyl; 3,4-difluorophenyl; 3-chloiO-4-fluorophenyl; 2,3,4- trifluorophenyS; 2,4,5-trifluorophenyi; 2,3,5-trifluorophenyl; 3,4,5- trifluorophenyl; or 3-azido-2,4-difluorophenyl;

R 4a is hydrogen;

R 5 is hydrogen; or R 4 , R 4a and R 5 can form together with the 2-oxo-l -pyrrolidine ring the following 1 ,3-dihydro-2H-indol-2-one cycle

wherein

R 1" is hydrogen;

R 1" is selected from methyl; chloro; bromo;

R 14 is hydrogen;

R 13 hydrogen;

R fc is hydrogen:

R' is hydrogen;

with the proviso that R 4 is different from hydrogen when

represents a group of formula

In a most preferred embodiment, the compounds of formula I, or pharmaceutically acceptable salt thereof, are those wherein

R l is selected from hydrogen; methyl; methylthio; nitro; cyano; amino; chloro:

R ' is selected from hydrogen: chloro; cyano; R' is hydrogen;

R 4 is selected from n-propyl; 2,2-difluorovinyl; phenyl; 3-chlorophenyl; 3 fluorophenyl ; 3,5-difluorophenyl; 2,3,4-t.riiluorophenyl ; 2,4,5-trifluorophenyi 2,3,5-trifluorophenyl; 3,4,5-trifiuorophenyi; 3 -azido~2,4~d fluorophenyl;

R 4il is hydrogen;

R 5 is hydrogen; or R 4 , R 4a and R 5 can form together with the 2-oxo-l-pyrrolidine ring the following l ,3-dihydro-2H-indol-2-one cycle

R " is hydrogen;

R lj is selected from chloro; brorno; methyl;

R i4 is hydrogen; 1^ hydrogen;

R 6 is hydrogen; R 7 is hydrogen, In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: l -(1 H-imidazol-l- ylmethyl)pyrrolidin-2-one; 1 -( 1 H-imidazol- 1 -ylmethy1)-4-plienylpyrrolidin-2~ one; 4-(3-azido-2,4,6-trifluorophenyl)-l-(l H-imidazol-l-yimethyl)pyrrolidin- 2— one; 1 -(1 H-imidazol- 1 -ylmethyl)-4-propylpyrrolidin-2-one; (-)-4-(3-azido- 2,4-difluorophenyl)-l ~(1 H-imidazol- 1 -ylmetbyl)pyrrolidin-2- -one; (-t-)-4-(3- azido-2,4-difluorophenyl)- 1 -( 1 H-imidazol- 1 -ylmethyl)pyrralidin-2-one; 1 - [(2- ethyl-l H-imidazol-l-y3.)m.ethyl]-4-propylpyrroHdin-2-one; l-[(2-isopropyl-l H- imidazol-l-yi)methyl]-4-propylpyrrolidin-2-one; 1 -[(2-niethyl-lH-imidazol-l - yl)methyl]-4-propylpyiTolidin-2-one; 1 -[(2 -phenyl- 1 H-imidazol- 1 -yl)methyl]~ 4-propylpyrrolidin-2-one; 4-propyl-l-[(2-propyl-l H-imidazol-l- yl)methyl]pyiTolidin-2-oiie; (+)- 1 -( 1 H-imidazol- 1 -ylmethyl)-4- propylpyrrolidm-2-one; (-)-l-(l H-imidazol- l-ylmethyl)-4-propylpyrrolidin-2- one; 4-(2,2-diiliwrovinyi)-l-(lH-imidazoi-l-y1met1iyl)pyiTolidin- 2-one; 4-(3- chlorophenyl)-l ~(1 H-imidazol- 1 -ylmethyl)pyrro3.idin-2-one; 1 ~{[2- (methylthio)- 1 H-imidazol- 1 -yljmethyl} -4-propylpyrrolidin-2-one; 1 - { [2- (methylsulfinyl)-ni-imidazo3-l-yl]m.ethyl}-4-propylpyrrolidi n-2-one; l-[(2- tert-butyl- 1 H-imidazol- 1 -y [)methyl]-4-propylpyrrolidin-2-one; 1 -[ 1 -( 1 H- imidazol- 1 -yl)cyciopropyl]pyrrolidin-2-one; 1 - [(2 -methyl- 1 H-imidazol- 1 - yl)methyl] -4-pheny Ipyrroii din-2-one; 3 - { [2-( methylsul fony 3 )- 1 H-imidazol - 1 - yljmethyl} -propylpyrrolidin-2-one; 1 -[(2-oxo-4-propylpyrrolidin- 1 -yl)methyl]- lH-imidazole-2-carboxamide, 4-(4-fluorophenyl)-l-(l H-imidazol- 1- ylmethyl)pyrrolidin-2-one: 1 -( 1 H-imidazol- 1 -ylmethyl)-4-(3 ,4,5- trifluorophenyl)pyTrolidin-2-one; 4-(3-fluorophenyl)-l-(l H-imidazol-l- ylmethyl)pyrrolidin-2-one; 4-(3,5-difluorophenyl)-l-(lH-imidazol- l- ylmethyl)pyrrolidin-2-one; 4-(3 ,4-diiluorophen 3)- 1 -( 1 H-imidazol- 1 - ylmethy l)pyrrolidin-2-one; 4-(3 -chioro-4-fluorophenyl)- 1 -( 1 H-imidazol- 1 - ylmethyl)pyrroiidin-2-one; 4-(4-chlorophenyl)~ 1 -( 1 H-imidazol- 1 - ylmethyl)pyrroiidin-2-one; 1 -(1 H-imidazol- l-ylmethyl)-4-(2,3 ,4- trifluorophenyl)pyrrolidin-2-one; l-(lH-imidazol-l-ylmethyl)-4-(2,3.5- trifluorophenyS)pyrro3idin-2-one; l -(3 H-imidaz.ol-l -yimethyS)-4-(2,4,5- trifluorophenyl)pyrrolidm-2-one; 1 - { [2-(hydroxymethyl)~ 1 H-imidazol- 1 - yl]methyl}-4-propylpyrrolidin-2-one; methyl l~[(2-oxo-4-propylpyirolidin-l- yI)methyl]-lH-imidazole-2-carboxyla- te; 1 -[(2-nitro-l H-imidazol-1- yl)methyl]-4-(3,,4,5-trifluorophenyl)pyrrolidin-2-one; l-{[2-oxo-4-(3.4,5- trifiuorophenyl)pyrrolidin~ 1 -yljmethyl} - 1 H-imidazole-2-carbonitrile; 1 -[(2- amino- 1 H-iniidazol- 1 -yl)methyl] -4-propylpyrrolidin-2-one; 1 - [(2,4-dichloro- l H-imidazol-l-yl)m€thyl]-4-(3,4,5-trifluorophenyl)pyrrolidi n-2-one; l-[(5- chloro-lH-imidazol-l-yl)methyl]-4-(3,4,5-1rifluorophenyl)pyr rolidin- -2-one; 1 - { [2-oxo-4-(3 ,4,5-trifluorophenyl)pyrrolidin- 1 -yljmeth l} - 1 H~imidazole-4~ carbonitriie; 1 - {[2-oxo-4-(3,4,5-trifluorophenyl)pyrroiidin- 1 -yljmethyl } - 1 H- imidazole-5 -carbonitrile; (+)- 1 -( 1 H-imidazol- 1 -y lmethyl)-4-phenylpyrrolidin- 2 -one; (-)-l-(l H-imidazol- l-ylniethyl)-4-phenyipyrrolidin-2-one; l -{[2-oxo-4- (2.3 ,5-trifluoroplieiiyl)pyiTolidin- 1 -yljmethyl } -1 H-imidazole-5-carbonitrile; (- )-l- {[2-oxo-4-(2,3,4-trifluorophenyl)pyrrolidin-l-yiJmethyl}-l H-imidazole-5- carbonitrile; (+)- 1 - { [2-oxo-4-(2,3 ,4-trifluorophenyl)pyrrolidin- 1 -yljmethyl} - l H-imidazole-5-carbonitrile; (-)-l-{[2-oxo-4-(2,3,4-trifluorophenyl)pyrroiidin- 1 -yljmethyl } - 1 H-imidazole-4-carbonitrile; (+)- 1 - { [2-oxo-4-(2,3 ,4- trifluorophenyl)- 1 - pyrroiidinyljmethy ί } - 1 H-imidazole-4-carbonitrile; (-)- 1 - { [2-oxo-4-(3 ,4,5-trifluorophenyl)pyrrolidin- 1 -yljmethyl } - 1 H-imidazole-4- carbonitrile; (+)- 1 - { [2-oxo-4-(3 ,4,5-trifluorophenyl)pyrrolidin- 1 -yljmethyl} - l H-imidazoie-4-carbonitrile; (+)-l- {[2-oxo-4-(2,4,5-trifluorophenyl)pyrroiidin- 1 -yljmethyl} - 1 H-imidazole- -carbonitrile; (-)- 1 - { [2-oxo-4-(2,4,5- trifluorophenyS)pyrrolidin- 1 -yljmethyl} - 1 H-imidazole-4-carbonitrile; (-)- 1 - {[2- oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-l -yljmethyl} -lH-imidazole-4- carbonitrile; (-)- 1 - { [2-oxo-4-(3 ,4,5-trifluorophenyl)pyrrolidin- 1 -yljmethyl } - lH-imidazoie-5-carbonitrile; l-{[2-oxo-4-(2,3,5-trifluorophenyl)pyrrolidin-l- yljmethyl}-! H-imidazole-5~ -carbonitrile; l-{[2-oxo-4-(2,3,5- trifli!orophenyl)pyrrolidm- 1 -yljmethyl} -lH-imidazole-5- -carbonitrile; l-[(5- methyl-2-phenyl- 1 H-imidazol- 1 -yl)methylJ-4-propylpyrrolidin-2-oiie; 1 -[(5- methyl- 1 H-imidazol- 1 -yl)methy 1 J -4-propylpyrro 1 idi -2-one; 1 - [(5 -phenyl- 1 H- imidazol- 1 -yl)methyl] -4-propy lpyrrolidin-2-one; 1 - [(2-ethyl-5 -methyl- 1 H- imidazol-l-yi)methyl]-4-propylpyrrolidin-2-one; 1 -[(2,5-dimethyl-l H- imidazol- 1 -yl)methyl] -4 -propylpyrrolidin-2-one; 1 - [(2-chloro- 1 H-imidazol- 1 - yl)methyl]-4-(3,4,5-triiluorophenyl)pyrrolidin- -2-one; l-[2-azido-I-(lH- imidazol- 1 -yl)ethy I ] -4-propylpyrro i idin -2-one; 1 -[(4-chloro- 1 H-imi dazol- 1 - yl)methyl]-4-(3,,4,5-trifluorophenyl)pyrrolidin- -2-one; l-[(2-bromo-4,5- dichloro-lH-imidazol-l -yl)methyi]-4-propylpyrroiidin-2-one; l-[(2-chloro-lH imidazol-l-y[)methyl]-4-propy[pyrrolidin-2-one; (+)-l-{[2-oxo-4-(3,4,5- trifluorophenyl)pyrrolidin-l -yljmethyl} -lH-imidazole-5-carbonitrile; 1 ~{[5~ (hydroxymethyl)- 1 H-imi dazol- 1 -yljmethyl } -4-propylp>irolidin-2-one; 1 - { [4- (hydroxymethyl)-l Bl-imidazol-l -yljmethyl} -4-propylpyrrolidin-2-one; benzyl

1- [(2-oxo-4-propylpyrrolidin-l -yl)methyl j-1 H-imidazol-5-yicarbamat- e; N- [( 1 - { [2-oxo-4-(3 ,4 ,5-frifluorophenyljpyrrolidm- 1 -yljmethyl} - 1 H-imidazol-5- yl)methyljacetamide; -[(l-{[2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidin-l- yljmethy 1} - 1 H-imidazol-5 -yl)methyl jbenzamide; N- [( 1 - { [2-oxo-4-(3 ,4,5- trifluorophenyljpyrrolidin- 1 -yljmethyl} - 1 H-imidazoi-5- yl jmethyl jpropanamide; 1 -( 1 H-benzimidazol- 1 -ylmethyl)-4 -propylpyrrolidin-

2 - one; l-[(2-memyl-lH-benzimidazol-l -yljmethyl j-4-propylpyrrolidin-2-one; 4-propy i- 1 -[(2-propy i- 1 H-benzimidazol- 1 -yl)methy [ jpyrrolidin-2-one; 1 - [(2- isopropyl- 1 H-benzimidazol- 1 -yl)m.ethyl j-4-propy 3pyrrolidin-2-one; 4-propy - l- {[2-(trifluoromethyl)-lH-benzimidazol-l-yljmethyl}pyrrolidin -2- -one; 1-

{ [2-(methy lthio)- 1 H-benzimidazol- 1 -yljmethyl} - -propylpyrrolidin-2- -one; 1 [(2-amino-l H-benzimidazol-l-yi)methylj-4-propylpyrrolidin-2-one; 1.-{[2- (chloromethyl)- 1 H-benzimidazol- 1 -yljmethyl} -4-propyipyrrolidin-2-on- e; { 1 - [(2-oxo-4-propylpyrrolidin-l -yljmethyl j-lH-benzimidazol-2-yl}acetoni- triie; 1 -[(5-methoxy- 1 H-benzimidazol- 1 -yljmethylj-4-propylpyrrolidin-2-one- ; 1 - [(5-methyl-l H-benzimidazol- 1 -yljmethyl j-4-propylpyrrolidm-2-one; 1 -[(5,6- dimethyl- lH-benzimidazol-l-yljmethylj-4-propylpyrrolidin-2-one; 1- {[2- isopropyi-5~(trifl.uoromethyl)-l H-benzimidazol- 1. -yljmethyl }-4-propyl- pyrrolidin-2-one; l-[(6-chloro-l H-benzimidazol- 1 -yljmethyl j-4- propylpyrrolidin-2-οηε; 1 -[(2-oxo-4-propylpyrrolidin- 1 -yl)methyl]-2-propyl- 1 H-benzimidazole-5-car- bonitriie; 1 -{[2-ethyl-5-(trifluoromethyl)-l H- benzimidazol- 1 -yljmethyl} -4— propylpyrrolidiii-2-one; -propyl- 1-{ [2-( 1 H- pyrrol-2-ylj-l H-benzimidazol- 1-yl jmethyl}pyrrolidin-2— one; 1 -[(5-fluoro-2- propyl- 1 H-benzimidazol- 1 -yljmethyl j -4-propylpyiTolidin- -2-one; 1 - { [6- methyl-2-( lH-pyrrol-2-yl)- 1 H-benzimidazol- 1 -yljmethyl} -4-pro- pylpyrrolidin-2-one; 1 -[(6-methoxy-2-propy 1 - 1 H-benzimi dazol- 1 -yl)methy l]-4- propylpyrrolidin-2— one ; 2-butyl- 1 -[(2-oxo-4-propylpyrrolidin- 1 -yl)niethylj- lH-benzimidazole-5- -carbonitriie; 1- {[2-[2-(methylthio)ethyl]-5- (trifluoromethyl)- 1 H-benzimidazol- 1 -yljmethyl} -4-propylpyrrolidm-2-one; 1 - [(5-fluoro-2-isobutyl-ni-benzimidazol-l-yl)rn.ethyl]-4-propy lpyrrolidin-2- - one; 1 - { [5-fluoro-2-(2,4,4-triniethylpenty 1)- 1 H-benzimidazol- 1 -yljmethyl }— 4- propylpyrrolidin-2-one; 2-cyclopropyl- 1 -[(2-oxo~4-propylpyrrolidm- 1 ~ yS)methyl]-l H-benzimidazole— 5 -carbonitriie; 1 -[(2-oxo-4-propylpyrrolidin-l- yljmethyl] -2-(l H-pyrazol-3-yl)- 1 H-benzimidazole-5 -carbonitriie; l-[(2- cyclopropyl-5-fluoro-lH-benzimidazol-l -yl)methyl]-4-propylpyrrolidin-2-one;

1- [(5-fluoro-2-isopropyl-lH-beiizimidazol-l-yl)methyl]-4-propy lpyrrolidin-2- one; l-{[2-(3-furyl)-6-methoxy-l H-benzimidazol- 1 -yljmethyl} -4- propylpyrrolidin- -2-one; 1 - [(2-cyclopropyl-6-methoxy- 1 H-benzimidazol- 1 - yljmethyl] -4-propy p- yrrolidin-2-one; l -[(2-isopropyl-6-methoxy-lH- benzimidazol-l-yl)methyl]-4-propylpyrrolidin- -2-one; l-[(2-oxo-4- propylpyrrolidin-l-yl)methyl]-2-(l,2,3-thiadiazol-4-yl- )- 1 H-benzimidazole-5- carbonitrile; l-{[2-(lH-imidazol-2-yl)-5-(trifluoromethy[)-l H-benzimi dazol- 1 - yljmethyl}- -4-propyipyrrolidin-2-one; l-{[5-fluoro-2-(2,2,2-trifluoroethyl)-

1 H-benzimidazol-l-yiJmethyl}-4-propyipyrrolidin-2-one; 1 -{[2-(l- ethylpropyl)-6-methoxy- 1 H-benzimidazol- 1 -yljmethyl} -4-propylpyrr- olidin-

2- one; l -{[6-methoxy-2-(l -methyl- lH-pyrrol-2-yi)-l H-benzimidazol- 1 - yljmethyl} -4— propyipyrrolidin-2-one; 1 - { [2-(2-fuiyl)-5-(trifluoromethyi)- 1 H- benzimidazol- 1 - ljmethyl ' } -4-prop 3 - pyrro3.idm-2-one; 4-propyi- 1 - { [2-thien-2- yl-5-(trifluoromethyl)-l H-benzimidazol- 1 -yljmethyl- }pyrrolidin-2-one; l-{[2- (3-fuTyl)-5-(tri " fluoromethyl)- 1 H-benzimi dazol- 1 -yljmethyl }-4-propyl~ pyrrolidin-2-one; l-{[2-cyclopropy[-5-(trifluoromethyl)-lH-benzimidazo[-l- yljmethyl} -4-propylpyrrolidin-2-oiie; 4-propyl- 1 - { [2-( 1 H-pyrrol-2-yl)-5- (trifiuoromethyl)-lH-benzimidazol-l-ylJ- methyl}pyrrolidin-2-one; 1-fl H- imidazol- 1 -yimethy 1)- 1 ,3 -dihy dro-2H-indol-2-one; 5 -bromo- 1 -( 1 H-imidazol- 1 - ylmethyl)-l ,3-dihydro-2H-indol-2-one; 5-chloro-l-(l H-imidazol-l-ylmethyl)-

1 ,3-dihydro-2H-indol-2-one; 4-fluoro- 1 -(1 H-imidazol- 1 -yimethy 1)- 1 ,3- dihydro-2H-indol-2-one; 4-chloro- 1 -( 1 H-imidazol- 1 -ylmethyl)- 1 ,3 -dihydro- 2 H -indo I -2-one; 1 -( 1 H-imi dazol- 1 -ylmethy l)-5 -methyl- 1 ,3 -dihydro-2 H-indol- 2-one; 1 -[(2-oxo-2,3-dihydro- IH-indol- 1 -yl)methyl]- 1H- imidazole-5- carbonitrile; and 1 -[(5-chloro-2-oxo-2,3-dihydro-l H-indol- 1 -yl)niethyljj- 1 H- imidazole-5-c- arbonitrile. in some embodiments, compounds useful in the methods and compositions of this invention are selected irorn the group consisting of: l-(l H-imidazol-l- ylmethyl)pyrro[idin-2-one, l-(l H-imidazo[-l-ylmethyl)-4-phenylpyrrolidm-2- one: 1 -( 1 H-imidazol- 1 -y imethyl)-4-propy lpyrrolidiii-2-one; (-)-4-(3-azido-2,4- difluoropheny!)-l-(l H-iniidazo!-l-ylmethyl)pyrrolidin-2- -one; (+)-4-(3-azido- 2 ,4-difluorophenyl)- 1 -( 1 H-imidazol- 1 -ylmethy l)pyrrolidin-2-one; 1 - [(2-ethyl- l H-imidazoi-l-yl)methyl]-4-propylpyrrolidin-2-one; l-[(2-isopropyl-l H- imidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one; 1 -[(2 -methyl- 1 H-imidazol- 1 - yl)methyl]-4-propylpyrrolidin-2-one; 4-propyl- 1 -[(2 -propyl- 1 H-imidazol- 1 - yl)methyl]pyrrolidin-2-one; (+)- 1 -( 1 H-imidazol- 1 -ylmethy [)-4- propylpyrrolidin-2-one; (-)-l-(lH-imidazol-l-ylmethyl)-4-propylpyrrolidin-2- one; 4-(2,2-difluorovinyI)-l-(lH-imiciazoI-l-ymiethyl)pyrrolidin- 2-one; 4-(3- chlorophenyl)- 1 -(1 H-imidazol- 1 -ylmethyl)pyrrolidin-2~one; l-{[2- (niethylthio)-l H-imidazol-l-yl]niethyl}-4-propylpyrrolidin-2-one; l -[(2- methyl-lH-imidazol-i-yl)methyi]-4-phenylpyiTolidin-2-one; 4-(4- fluorophenyl)- 1 -( 1 H-imidazol- 1 -ylmethy! )pyrrolidm-2-one; 1 -( 1 H-imidazol- 1 - ylmethyi)-4-(3 l,5-trifluoiOphenyi)pyrrolidin-2-one; 4-(3 -fluorophenyl)- 1-(1 H- imidazol-l-ylniethyl)pyn lidm-2-one; 4-(3 ,5-difluorophenyi)- 1 -( 1 H-imidazoi- 1 -y lmethyl)pyrrolidin-2-one; 4-(3 ,4-difluorophenyl)- 1 -( 1 H-imidazol- 1 - ylrnethyi)pyrrolidin~2-one; 4-(3-chloro-4-fluorophenyl)-l -(1 H-imidazol- 1 - ylmethyl)pyrrolidin-2-one; 4-(4-chlorophenyl)-l-(l H-imidazol- 1- ylmethyl)pyrrolidin-2-one; 1-(1 H-imidazol- 1 -ylmethy l)-4-(2, 3,4- trifluorophenyl)pyrrolidvn-2-one; 1 -(11 H-imidazol- l-ylmethyl)-4-(2,3,5- trifluorophenyl)pyiTolidin-2-one; 1 -( 1 H-imidazol- 1 -yimethyl)-4-(2,4, 5- trifluorophenyl)pyrrolidin-2-one; l-[(2-nitro-l H-imidazol-l-yl)methyl]-4- (3,4,5-trifluorophenyl)pyiTolidin-- 2-one; l- {[2-oxo-4-(3.4,5- trifluorophenyl)pyrrolidin- 1 -yljmethyi] - 1 H-imidazole-2-carbonitrile; 1 -[(2- amino- 1 H-imidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one; 1 - [(5-chloro- 1 H- imidazol-1 -yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; 1- {[2-oxo- 4 -(3 ,4 ,5-trifluoroplienyl)pyrro1idin- 1 -yijniethyl} - 1 H-imidazole-4-carbonitrile; l- ^P-oxo^-CS^jS-trifluoropheny^pyrroiidm-l-ylJmethylj-l H-iiTiidazole-S- - carbonitriie; (+)-l-(lH-imidazol-l-ylmethyr)-4-phenylpyrrolidin-2-one; (··;··· I · (l H-iniidazol-l-y]rneihyi)-4~phenyipyrro]idin~2-one; (+); l -{[2-oxo-4-(3,4,5- trifluorophenyl)pyrrolidin- 1 -yljmethv i } - 1 H-imidazole-4- -carbonitriie; 1 - [(2- chloro-lH-imidazol-l -yl)methyl]-4-(3,4,5-lTifluorophenyl)pyrrolidin- -2-one; 1 -[2-azido- 1 -f 1 H-imidazol- 1 -yi)ethyl]-4-propylpyrrolidin-2-one; 1 -[(2-chloro- 1 H-imidazol- 1 -y1)methyl]-4-propylpyrrolidin-2-one; (+)- 1 - { [2-oxo-4-(3 ,4,5- trifluorophenyl)pyrrolidin-l -yl]methyi}-lH-imidazole-5-carbonitrile; l-[(2- oxo-4-propylpyrrolidin- 1 -y l)methyl] -2 -propyl- 1 H-benzimidazole-5 -carbonitriie; 1 - {[2~ethyi-5~(trifiuoromethyl)~ 1 H-benzimidazol- 1 -yijmethyl } -4— propylpyrrolidin-2-οηε; 4-propyi- 1 - { [2-( 1 H-pyrrol-2-yl)- 1 H-benzimidazol- 1 - yijmethyl } pyrrol idin-2-- one; 1 - [(5 -fluoro-2-propyl- 1 H -benzimidazol- 1 - yl)methyl] -4-propylpyrrolidin- -2-one; 2-butyl- 1 - [(2-oxo-4-propylpyrrolidin- 1 - yl)methyl]-l H-benzimidazole- -5-carbonitrile; 1 -[(5-fluoro-2-isopropyl-l H- benzimidazol- 1 -yl)methyl]-4-propylpyrrolidin~ 2-one; 1 -(1 H-imidazoi- 1 - ylmethvl)- 1 ,3-dihydro-2H-indol-2-one; 5-bromo- 1 -( 1 H-imidazol- 1 -ylmethyl)- 1 ,3-dihydro-2H-indol-2-one; 5-chloro- 1 -(1 H-imidazol- 1 -ylmethvl)- 1,3- dihydro-2H-indol-2-one; 1 -( 1 H-imidazol- 1 -ylmethyl)- 5 -methyl- 1 , 3 -dihydro- 2H-indoi-2-one; l-[(5-chloro-2-oxo-2,3-dihydro-l.H-indol-l -yl)methyl]-lH- imidazole-5-carbo- nitrile. in some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: l-(l H-imidazol-l- ylmethyl)-4-phenylpyrrolidin-2-one; 1 -( 1 H-imidazol- 1 -ylmethyl }-4- propylpyrrolidin-2-one; (-)-4-(3-azido-2,4-difluorophenyl)-l -(lH-im.idazol-l - ySmethyl)pyrroiidin-2- -one; (+)-4-(3-azido-2,4-difluorophenyl)-l -(lH- imidazol- 1 -ylmethyl)pyrrolidin-2-one; 4-(2,2-difluorovinyl)- 1 -( 1 H-imidazol- 1 - ylmethyl)pyrroiidin-2-on- e; 4-(3-chlorophenyl)-l -(lH-imidazol-1 - ylmethyl)pyrrolidin-2-one; 1 - { [2-(methylthio)- 1 H-imidazol- 1 -yijmethyl} -4- propylpyrrolidm-2-one; l-[(2-methyl-lH-imidazol-l -yl)methyl]-4- pheiiylpyiTolidhi-2-Qne; 1 -( 1 H-iniidazol- 1 -y lmethyl)-4-(3 ,4,5- trifluorophenyl)pyrrolidin-2-one; 4-(3-fluorophenyl)-l-(l H-imidazol-l- ylmethyl)pyrrolidin-2-one; 4-(3 ,5-difluorophenyl)- 1 -( 1 H-imidazol- 1 - ySmethy!)pyrrolidin-2-one; 1-(1 H-imidazol- 1 -ySmethy l)-4-(2,3,4- trifluorophenyl)pyrrolidin-2-one; l -(lH-imidazol-l -ylmethyl)-4-(2,3,5- trifiuorophenyl)pyTrolidin-2-one; l-(l H-imidazol-l-ylmethyl)-4-(2,4,5- trifluorophenyl)pyrrolidin-2-one; 1 -[(2-nitro- 1 H-imidazoi- 1 -yl)methyl]-4- (3,4,5-trifluorophenyl)pyrrolidin-- 2-one; l~ {[2-oxo-4~(3,4,5- trifiuorophenyl)pyrro!idin~ 1 -yi jmethyl} - 1 H-imidazole-2-carbonitrile; 1 -[(2- amino- 1 H-imidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one; 1 - [(5-chloro- 1 H- imidazol-1 -yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin- -2-one; (+)-!-( 1 H- imidazol- 1 -ylmethyl)-4-phenylpyrrolidin-2-one: (-)- 1 -( 1 H-imidazol- 1 - ylmethyl)-4-phenylpyrrolidin-2-one; l-[(2-chloro-lH-imidazol-l -yl)methyi]-4- (3 ,4,5 -trifluorophenyi)pyrrolidin- -2-one: 1 - [(2-cliloro- 1 H-imidazol- 1 - yl)rnethyl]~4-propylpyn , olidin-2-one; (+)-!- {[2-oxo-4-(3 ,4,5- trifluorophenyl)pyrrolidin-l -yljmethyi} -lH-imidazole-5-carbonitrile; 5-bromo- l-(l H-imidazol-l-yimethyl)-l ,3-dihydro-2H-indol-2-one; 5-chioro-l -(l H- imidazol- 1 -yimethyl)-! ,3-dihydro-2H-indol-2-one; 1 -(1 H-imidazol- 1 - ylmethyl)-5-methyl-l ,3-dihydro-2H-indol-2-one; l-[(5-chloro-2-oxo-2,3- dihydro-lH-indol-1 -yl)methyl j-1 H-imidazole-5-carbo- nitrile.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: (~)-4-(3-azido-2,4- difluorophenyl)-l -(l H-imidazol- l -ylmethyl)pyrroiidin-2- -one; (+)-4-(3-azido- 2,4-difluorophenyi)-l -(1 H-imidazoi- l-ylmethyl)pyrrolidin-2-one; 4-(3-azido- 2,4-difiuorophenyl)-l -(lH-im.idazol-l -ylmethyi)pyrro3.idin-2-one. v) International Patent Application WO 2007/065595:

Compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof, wherein

R 1 is hydrogen or C 1-6 alkyl; R 2 is hydrogen or C 1-4 alkyl;

R J is a group of formula -CHR R 6 or a benzyl group;

R 4 is Cj-s alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle;

R 5 is C2-4 alkyl;

R 6 is C2-4 alkyl, amido or -COO 7 ; R 7 is CI -4 alkyl;

Usually when R J is a benzyl group, then R 4 is C s alkyl optionally substituted by alkoxycarbonyl.

Usually when R J is a group of formula -CHR^R 6 then R 4 is Ci-s alkyl optionally substituted by C 3-6 cycloalkyl, aryl or heterocycle.

The term "alkyl", as used herein, is a group which represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched moieties, or combinations thereof, and containing 1-8 carbon atoms, preferably 1 -6 carbon atoms; more preferably alkyl groups have 1-4 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl, acyl, aryl or heterocycle. Alkyl moieties may be optionally substituted by a cycloarkyl as defined hereafter, Preferred alkyl groups are methyl, cyanomethyi, ethyl, 2-ethoxy-2-oxoethyl, 2- methoxyethyi, n-propyl,

2- oxopropyl, 3-hydroxypropyl, 2-propynyl, n-butyl, i-butyl. n-pentyl, 3-pentyl, n-hexy!, cyelohexylmethyi, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4- bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, 4- (aminosulfonyl)benzyl, 1 - phenylethyl, 2-phenyl ethyl, (3,5-dimethylisoxazol- 4-yl)methyl or (5-nitro-2-furyi)methyl. More preferred alkyl groups are methyl, ethyl, cyanomethyi, 2-methoxyethyl, n-propyl, 3- hydroxypropyl, 2-propynyl, n-butyl, 3-pentyl, n-hexyl, benzyl, 3-bromobenzyl, 3- methoxybenzyl, 3- nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro- 2- furyl)methyl. Most preferred alkyl groups are methyl, ethyl, 3-methoxybenzyl,

3- iiitrobenzyl or (5-nitro-2-furyl)methyl.

The term "cycloalkyl", as used herein, represents a monovalent group of 3 to 8, preferably 3 to 6 carbon atoms derived from a saturated cyclic

hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group is cyclohexyl.

The term "ar i" as used herein, is defined as a phenyl group optionally substituted by 1 to 4 substituents independently selected from halogen, amino, nitro, aikoxy or aminosulfonyl. Preferred aryi groups are phenyl, 2- bromophenyl, 3-bromophenyl, 4- bromophenyl, 3-methoxyphenyl, 3- nitrophenyl, 3-aminophenyl or 4-(aminosulfonyl)phenyi.

The term "phenyl", as used herein, represents an aromatic hydrocarbon group of formula -C0H5, The term "benzyl group", as used herein, represents a group of formula -

CH 2 -aryl. Preferred benzyl groups are benzyl, 2-bromobenzyl, 3-bromobenzyl,

4- bromobenzyl, 3- methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyi or 4- (aminosulfonyl)benzyl. More preferred benzyl groups are benzyl, 3- bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl or 3- aminobenzyl. Most preferred alkyl groups are 3-methoxybenzyl or 3-nitrobenzyl, The term "halogen", as used herein, represents an atom of fluorine, chlorine, bromine, or iodine. Preferred halogen is bromine.

The term "hydroxy", as used herein, represents a group of formula -OH.

The term "cyano", as used herein, represents a group of formula -C .

The term "amino", as used herein, represents a group of formula ~N¾.

The term "ethynyl", as used herein, represents a group of formula -C=≡CH.

The term "alkoxy", as used herein, represents a group of formula -OR a wherein R a is an alkyl group, as defined above, Preferred alkoxy group is methoxy.

The term "nitro", as used herein, represents a group of formula -N0 2 .

The term "amido", as used herein, represents a group of formula - C(=0)NH2.

b

The term "acyl", as used herein, represents a group of formula -C(=G)R wherein R b is an alkyl group, as defined here above. Preferred acyl group is acetyl (-C(=0)Me).

The term "alkoxycarbonyl (or ester)", as used herein, represents a group of formula -COOR 0 wherein R c is an alkyl group; with the proviso that R c does not represent an alkyl alpha-substituted by hydroxy. Preferred alkoxycarbonyl group is ethoxycarbonyl.

The term "heterocycle", as used herein, represents a 5-membered ring containing one or two lieteroatoms selected from O or N. The heterocycle may be substituted by one or two C 1-4 alkyl or nitro. Preferred heterocycies are (3, 5-dm ethylisoxazol-4-yl) or (5-nitro- 2-furyl). Most preferred heterocycle is (5- nitro-2-furyl).

Generally R 1 is hydrogen or C 1-6 alkyl. Usually R 1 is hydrogen or C 1-6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl or acyl. Preferably R is hydrogen, methyl, cyanometlivl, 2-ethoxy-2-oxoethyi, 2- methoxyethyl, n- propyl, 2-oxopropyi, 3-hydroxypropyl, 2-propynyl, n-pentyl or ii-hexyl. More preferably R L is hydrogen, methyl, cyaiiomethyl, 2- methoxyethyl, n-propyi, 3-hydroxypropyl or 2- propynyl. Most preferably R 1 is hydrogen,

Generally R ~ " is hydrogen or CM alkyl. Usually R 2 is hydrogen or unsubstituted C 1-4 alkyl. Preferably " is hydrogen, methyl or n-but l. More preferably, R "' is methyl.

Generally R 1 is a group of formula -CHR J R 6 or a benzyl group. Preferably R 3 is 3-pentyl, l-(aminocarbonyl)propyl, l-(ethoxycarbonyl)propyl or 3- bromobenzyS. Most preferably R J is l -(ethoxycarbonyl)propyl.

Generally R " is C 1-8 alkyl optionally substituted by alkoxycarbonyl, C 3-6 cycloalkyl, aryl or heterocycle. Usually R 4 is Cj-g alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyi, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyi. Preferably R 4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2- bromobenzyl, 3-bromobenzyl, 4-bromobenzyl, 3- methoxybenzyl, 3-nitrobenzyl, 3- aminobenzyl, 4-(aminosu]fonyl)benzy], 1 - phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2- furyi)methyl or l-(ethoxycarbonyi)propyl. More preferably R 4 is n- butyl, n- hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3- aminobenzyl, (3,5-dimethyiisoxazol-4-yi)methyl, (5-nitro-2-furyl)methyl or 1 - (ethoxycarbonyi)propyl. Most preferably R " is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2-furyl)methyl.

Generally R 3 is C 2 -4 alkyl. Usually R 5 is unsubstituted Cz^4 alkyl.

Preferably R 5 is ethyl.

Generally R° is C 2 -4 alkyl, amido or -COOR' . Usually R° is unsubstituted C 2 -4 alkyl, amido or -COOR'. Preferably R 6 is ethyl, amido or ethoxycarbonyi. Most preferably R 6 is ethoxycarbonyi. Generally R' is CM alkyl. Usually R' is uiisubstituted CM alkyl.

Preferably, R' is ethyl.

In some embodiments, the compounds are those having formula and their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,

wherein

R l is hydrogen, C 1-6 alkyl optionally substituted by hydroxy, alkoxy, cya.no, ethynyl, alkoxycarbonyl or acyl;

R " is hydrogen or uiisubstituted C 1-4 alkyl;

R 3 is a group of formula -CHR J R 6 or a benzyl group;

R 4 is Ci..g alkyl optionally substituted by cyclohexyl, phenyl, bromophenyl, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5- di.methylisoxazol-4-yl, 5-mtro-2-furyl or ethoxycarbonyl;

R 5 is unsubstituted C 2-4 alkyl;

R is uiisubstituted C 2-4 alkyl, amido or -COOR';

R 7 is uiisubstituted C 1-4 alkyl; with the proviso that when R 1 is hydrogen, R 2 is methyl, R 3 is -CHR 5 R°, R 6 is ethoxycarbonyl and R J is ethyl, then R 4 is different from n-propyl, i- propyl, n-peiityl, n- heptyl, 3 -bromobenzyl, 4-chiorobenzyi, 4-methylbenzyl or 2-phenylethyl. In the above embodiment, preferably, when R" is a benzyl group, then R C 1-8 alkyl optionally substituted by alkoxycarbonyi.

In the above embodiment, preferably, when R 3 is a group of formula - CHR 5 R 6 , then R 4 is C 1-8 alkyl optionally substituted by C 3-6 cycloaikyi, aryl heterocycle.

In a preferred embodiment,

R 1 is hydrogen, methyl, cyanomethyl, 2-ethoxy-2~oxoethyl, 2- methoxyethyl, n- propyl, 2-oxopropyl, 3-hydroxypropyi, 2-propynyI, n-pentyl or n-hexyl; R 2 is hydrogen, methyl or n-butyl;

R J is 3-pentyl, l-(aminocarbonyi)propyl, 1 -(ethoxycarbonyl )propyl or 3- bromobenzyi;

R 4 is n-butyl, i-butyi n-pentyl, n-hexyl, cyelohexylmethyi benzyl, 2- bromobenzyl, 3- bromobenzyl, 4-bromobenzyi, 3-methoxybenzyi, 3- niirobenzyl, 3-aminobenzyl, 4- (aminosulibnyl)benzyl, 1 -phenyl ethyl, 2~ phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5- nitro-2-furyl)methyl or 1 (ethoxycarbonyl with the proviso that when R. 1 is hydrogen, " is methyl and R 3 is l~ (ethoxycarbonyl)propyl, then R 4 is different from n-pentyl, 3-bromobenzyl or 2- phenylethyl.

In the above embodiment, preferably, when R 3 is 3-bromobenzyl, then R' is Ci-8 alkyl optionally substituted by alkoxycarbonyi.

In the above embodiment, preferably, when R:' is 3-pentyl, 1 - (aminocarbonyl)propyl or l-(ethoxycarbonyl)propyl, then R 4 is different from 1 - (ethoxycarbonyl)propyi.

In a more preferred embodiment. R 1 is hydrogen, methyl, cyanomethyl , 2-methoxyethyl, n-propyl, 3- hydroxypropyl or 2-propynyl;

R 2 is methyl;

R' is 3-pentyi l -(aminocarbony!)propyI, l ~(ethoxycarbonyl)propy! or 3- bromobenzyl;

R 4 is n-butyl, n-hexyl, benzyl, 3-bromobenzyl, 3-niethoxybenzyl, 3- nitrobenzyl, 3- aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2- fui l)methyl or 1- (ethoxycarbonyl)propyi; with the proviso that when R 1 is hydrogen, R 2 is methyl and R' is 1 - (ethoxycarbonyljpropyl, then R' is different from 3-bromobenzyl.

In the above embodiment, preferably, when R 3 is 3-bromobenzyl, then R 4 is 1 - (ethoxyearbonyl)propyl;

In the above embodiment, preferably, when R 3 is 3-pentyS, 1- (ammocarbonyl)piOpyl or l-(ethoxycarbonyl)propyl, then R' is different from 1 - (ethoxycarbonyi)propy 1 ;

In a most preferred embodiment, R 1 is hydrogen; R" is methyl; R J is 1- (ethoxycarbonyi)propyl; and R 4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro- 2- furyl)methyl.

A further embodiment consists in compounds wherein R." is methyi, R J is a group of formula -CHR 5 R 6 with R 5 being C 2-4 alkyl, R 6 being amido or - COOR 7 and R' being methyi or ethyl,

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzy 1!-1 ,3-dimethyl-2,6-dioxo-2,3,6,7- tetrahydro- lH-purin-8-yl)thio]butanoate; ethy 2-{[7~(3-bromobenzyl)-l-(2~ethoxy~2- oxoethyl)~3~meth l-2,6-dioxo-2,3,6,7- tetrahydro-1 H-purin-8-yl]thio}butanoate; ethyl 2-{[7- (3-bromobenzyl)- 1 -(2- niethoxyethyl)-3-methyi-2,6~dioxo-2,3,6,7-tetrahydro-l H-purin-8- yl]thio}butanoate; ethyl 2-{[7-(3-liiOniobeiizyl)-2,6-dioxo-2,3,6,7-tetrahydiO-l H-purin-8- yi]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-l ,3-dimethyi-2,6- dioxo-2,3.6,7-telTahydro- 1 H purin-8-yl]thio}butanoate; ethyl 2-{[7-(2- bromobenzyS)-! ,3-dimethyl-2,6-dioxo-2,3,6,7- tetrahydro-1 H-purin-8- yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-l-(cyanomethyl)-3- methyi-

2,6-dioxo-2,3,6,7~tetrahydro-l H-purin-8- ljthio}butanoate; ethyl 2-{[7-(3- bromobenzyl)-3-methyl-2,6-dioxo-l-propyl-2,3,6,7-tetrahydro- l H-purin-8- yl]ihio}buiarioate; ethyl 2-{[7-(3-bromobenzyt)-3-methyl-2,6-dioxo~l-(2- oxopropyl)-2,3 5 6,7-tetrahydro-lH- purin-8-yl]thio}butanoate; ethyl 2- {[7-(3- bromobenzyl)-l-(3-hydroxypropyl)-3-methyl-2.6- dioxo-2,3,6,7-tetrahydro-l

H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3- methyl-2,6- dioxo- 1 -(2-propynyl)-2,3 ,6,7-tetrahydro- 1 H-purin-8-yl]thio}butanoate; ethyl 2- {[7-(3-methoxybenzyS)-3-niethyl-2,6-dioxo-2,3,6 7-tetrahydro-l H-purin-8- yljthio } butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6-dioxo-2,3,6,7- tetrahydro-1 H-purin-8- yljthio} butanoate; ethyl 2-{[7-(3-aminobenzyl)~3- methyl-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8-yl]thio} butanoate; ethyl 2- ({7-[4-(arn.inosulfonyl)benzyl]-3-methyl-2,6-dioxo-2, 3,6,7- tetrahydro-1 H- purin-8-yl}thio)butanoate; ethyl 2-{[7-(4-bromobenzyl)-l ,3-dimethyl-2,6- dioxo-2,3,6,7-tetrahydro-l H-purin-8-yl]thio}butanoate; ethyl 2- {[7- (cyclohexy Imethyi)- 1 ,3- dimethyl-2,6-dioxo-23,6,7-tetrahydro-lH--purin-8- yl]thio}butanoate; ethyl 2-{[l ,3-dimethyl- 2,6-dioxo-7-(l -phenylethyl)- 2,3,6,7-tetrahydro-l H-purin-8-yl]thio} butanoate; ethyl 2- {[1 ,3- diniethyl-2,6- dioxo-7-(2-phenylethyl)-2,3 i 6,7-tetrahydro-lH-purin-8-yl]thio}butanoate; ethyl 2-({7-[(3,5-dimethyjisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo -2,3,6,7- tetrahydro-1 H-purin-8- yl}thio)butanoate; ethyl 2-( {3-methyl-7-[(5-nitro-2- luryl)rnethyl]-2,6-dioxo~2,3,6,7-tetrahydro~ 1 H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-l H- purin-8- yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-2,6-dioxo-2,3 > 6,7-tetrahydro- 1 H- purin-8-yl]thio}butanoate; ethyl 2-[(l ,7-dihexyl-3-methyl-2,6-dioxo- 2,3 ,6,7-tetrahydro- 1 H- purin-8-yl)thio]butanoate; ethyl 2-[(7-hexyl-3-methyl-

2,6-dioxo-2,3,6,7-tetrahydro-l H-purin- 8-yl)thio]butanoate; ethyl 2-[(3- methyl-2,6-dioxo-l ,7-dipentyl-2,3,6j7-tetrahydro-l H-purin-8- yl)thio]butanoate; 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7- tetrahydro- 1 H-purin-8- yljthio} butanamide; 2-[(7-butyl-3-methy 1-2,6-dioxo- 2.3 ,6,7-tetrahydro- 1 H-purin-8- yl)thio]butanamide; 7-(3-bromobenzyi)-8-[( 1 - ethylpropyl)thio]-3-methyl-3,7-dihydro-l H- purine-2,6-dione; ethyl 2~{8-[{3- bromobenzyl)thio]-l ,3-dimethyl-2,6-dioxo-l ,2,3,6- tetrahydro-7H-purin-7- yljbutanoate; and ethyl 2-[(7-isobutyl-3-methyl-2,6-dioxo-2,3,6,7- tetrabydro- lH-purin-8-yl)thio]butanoate.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-[(7-benzyI-l ,3-dimethyl-2,6-dioxo-2,3,6,7- tetrahydro-1 H-purin-8-yl)thio]butanoate; ethyl 2-{[7-(3-bromobenzyl)-l-(2-methoxyethyl)-3- methyl-2,6-dioxo-2, 3,6,7- tetrahydro-1 H-purm~8~yl]thio} butanoate; ethyl 2-{[7-(3- bromobenzyl)-]. ,3- dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8-yl]tbio}butanoate; ethyl 2- {[7-(3-bromobenzyl)-l.-(cyanomethyl)-3-methyl-2,6-dioxo-2,3, 6,7-tetrahydro-l H-purin-8- yljthio} butanoate; ethyl 2- {[7-(3-bromobenzyl)-3-methyl-2,6- dioxo-l-propyl-2,3,6,7- tetrahydro-1 H-purin-8-yl]thio}butanoate; ethyl 2-{[7- (3-broniobenzyl)- 1 -(3-hydroxypropyl)-3- methyl-2,6-dioxo-2,3,6,7-tetrahydro- 1 H-purin-8-yi]thio}butanoate; ethyl 2-{[7-(3- bromobenzyl)-3-methyl-2,6- dioxo-l-(2-propynyI)-2,3,6,7-tetrahydro-l H-purin-8- yljthio} butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-niethyl-2,6-dioxo-2,3,6,7-tetrahyd ro-l H- purin-8- yljthio} butanoate; ethyl 2-{[3-niethyl-7-(3-nitrobenz}4)-2,6-dioxo-2,3,6,7- tetrahydro- 1 H-purin-8-yl]thio}butanoate: ethyl 2-{[7-(3-aminobenzyl)-3- methyl-2,6-dioxo-2,3,6,7- tetrahydro-1 H-purin-8-yl]thio} butanoate; ethyl 2- ({7-[(3,5-dimethylisoxazol-4-yl)methyl]-3- methyl-2,6-dioxo-2,3 ,6,7- tetrahydro- 1 H-purin-8-yl} thio)butanoate; ethyi 2-({3-methyi-7-[(5- nitro-2- furj )methyi]-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8-y[}thio)butanoate; ethyl 2-[(7- butyl-3-m.ethyl-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8- yl)thio]butanoate; ethyl 2-[(7-hexyS-3-methyi-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8-yl)thio]butanoate; 2- {[7-(3-broniobenzyl)~ 3-methyl-2.6-dioxo- 2,3,6,7-tetrahydro-l H-purin-8-yl]thio}butanamide; 7-(3-bromobenzyl)-8- [(1- ethylpix>pyl)thio]-3-niethyl-3,7-dihydro-l H-purine-2.6-dione; and ethyl 2-{8- [(3- bromobenzyl)thio]-l ,3-dimethyl-2.6-dioxo-l ,2,3,6-tetrahydro-7H-puriri- 7~yI}butanoate.

In some embodiments, compounds useful in the methods and compositions of this invention are selected from the group consisting of: ethyl 2-{[7-(3- methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8- yl]thio}butanoate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)-2,6- dioxo-2,3,6,7- tetrahydro-1 H-purm-8~yl]thio}butanoate; and ethyl 2-({3-rnethyl-7~[(5-nitro- 2- furyl)methyl]-2,6-dioxo-2,3,6,7-tetrahydro-l H-purin-8-yl}thio)butanoate.

In some embodiments, the compounds are those having formula II, their enaniiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts:

wherein R.sup.1 is hydrogen or C.sub, 1 -6 alkyl;

R.sup.2 is hydrogen or C.sub.1-4 alkyl;

R.sup.3 is a group of formula—CHR.sup.5R. sup.6 or a benzyl group; R.sup, 4 is C.sub.1 -8 alkyl optionally substituted by alkoxycarbonyL C.sub.3-6 cycioa!ky!, aryl or heterocycle;

R.sup.5 is hydrogen or C.sub.1-4 alkyl; R.sup.6 is C.sub.1-4 alkyl, amido or— COOR.sup.7;

R.sup.7 is C.sub.1 -4 alkyl;

In the above embodiment, in some cases, when R.sup.3 is a benzyl group, then R.sup.4 is C.sub.1-8 alkyl optionally substituted by alkoxycarbonyL

In the above embodiment, in some cases, when R.sup.3 is a group of formula— CHR.sup.5R.sup.6, then R.sup.4 is C, sub, 1-8 alkyl optionally substituted by C. sub.3 -6 cycioalkyl, aryl or heterocycle. In some embodiments, the compounds are those compounds of formula Π, their enaniiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts

wherein

R.sup. l is hydrogen or C. sub.1-6 alkyl; R.sup.2 is hydrogen or C. sub.1-4 alkyl;

R.sup.3 is a group of formula ~CHR..sup.5R.sup.6 or a benzyl group;

R.sup.4 is C. sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C. sub.3-6 cycioalkyl, aryl or heterocycle;

R.sup.5 is hydrogen or C.sub.1-4 alkyl;

R.sup.6 is C.sub.1-4 alkyl, amido or— COOR.sup.7;

R.sup.7 is C, sub.1-4 alkyl.

In some embodiments, the compounds are compounds of formula II selected from ethyl 2-[(7-heptyl-3-methyl-2,6-dioxo-2,3,6,7-tetTahydro-l H-purin-8-yl)thio]but- anoate; 7-(3-bromobenzyl)-3-methyl-8-(propylthio)-3,7-dihydro-l H-purine-2,- 6- dione; ethyl 2-[(3-methyl-2,6-dioxo-7-pentyl-2,3,6,7-tetrahydro-lH-purin- 8- yl)thio]but- anoate; ethyl 2- {[7-(3-biOmobenzyl)-3-methyi-2,6-dioxo-2,3,6,7- tetrahydro- lH-purin-8-yl- ]thio}butanoate; ethyl 2-[(3-methyl-2,6-dioxo-7-propyl- 2,3,6,7-teirahydro-lH~purin-8-yl)thio]but- anoate; 7-(3~bromobenzyl)~8-[(3- chloro-2-hydroxypropyl)thio]-3-methyl-3,7— diiiydro-1 H-purine-2, 6-dione; and ethyl 2-{[7-(3-bromobenzyl)-3-methyI-2,6-dioxo-2,3,6,7 etrahydro-l H-purin-8- yl- jthiojpropanoate. In some embodiments, the compounds are compounds of formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts

wherein

R.sup. l is hydrogen or C.sub.1 -6 alkyl; R.sup.2 is hydrogen or C. sub.1-4 alkyl;

R.sup.3 is a group of formula—CHR.sup.5R.sup.6 or a benzyl group;

R.sup.4 is C.sub.1 -8 alkyl optionally substituted by alkoxycarbonyl, C. sub, 3-6 cycloaikyi, aryi or heterocycle;

R.sup.5 is C, sub.2-4 alkyl;

R.sup.6 is C. sub.2-4 alkyl, amido or— COOR.sup.7; R.sup.7 is C. sub.1-4 alkyl;

In another embodiment, the compounds are compounds having formula I I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,

R.sup. l is hydrogen or C. sub.1-6 alkyl; R.sup.2 is hydrogen or C. sub.1-4 alkyl;

R.sup.3 is a group of formula— CHR.sup.5R,sup.6 or a benzyl group;

R.sup.4 is C. sub.1-8 alkyl optionally substituted by alkoxycarbonyl, C. sub.3-6 cycloalkyl, aryl or heterocycle;

R.sup.5 is hydrogen or C.sub.1-4 alkyl; R.sup.6 is C.sub.1-4 alkyl, ami do or— COOR.sup.7; R.sup.7 is C.sub.1 -4 alkyl;

vi) International Patent Application Publication No. WO2010/144712

In one embodiment, a chemical composition that includes a LEV derivative of Formula 1 or Formula 2 is disclosed.

Formula I Formula 2 n of Formula 2 and L, X, and Y of Formulas 1 and 2 are defined as follows: a) n is an integer with a value of 0 to 8; b) L is one of the group consisting of CH2, CO, NHCO, NHCOO, CONH, NH, O, or S, and combinations thereof; c) X is an end group, an aromatic group, an aryl group, or a saturated, unsaturated, substituted, unsubstituted, straight chain, or branched chain aliphatic group having from 1 to 10 carbon and/or hetero chain atoms, the hetero chain atoms being selected from the group consisting of oxygen, nitrogen, sulfur, or phosphorus, and combinations thereof; and d) Y is optional and if present is one of a functional group selected from group consisting of alcohol amine, amide, carboxylic acid, aldehyde, ester, iminoester, isocvanate, isothiocyanate, anhydride, thiol, thiolacetone, diazonium, NHS, CO-NHS, O-NHS, maleimido: or e) Y is a Yi-Z where Yi is selected from the group consisting of COO, CO, O, CONH, NHCO, or NH and Z is an operative group,

In one embodiment of the method, the operative group of Z is selected from the group consisting of detectable labels, antigenic carriers, coupling agents, end groups, proteins, lipoproteins, glycoproteins, polypeptides, polysaccharides, nucleic acids, polynucleotides, teichoic acids, radioactive isotopes, enzymes, enzyme fragments, enzyme donor fragments, enzyme acceptor fragments, enzyme substrates, enzyme inhibitors, coenzymes, fluorescent moieties, phosphorescent moieties, anti-stokes up-regulating moieties, chemiluminescent moieties, luminescent moieties, dyes, sensitizers, particles, micropartieles, magnetic particles, solid supports, liposomes, ligands, receptors, hapten radioactive isotopes, and combinations thereof,

vii) International Patent Application Publication No. VVO2010/002869

The present invention provides a compound of Formula Ϊ:

or a pharmaceutically acceptable salt thereof, wherein: each Z is independently selected from hydrogen and deuterium: Rl is an n-propyl group having zero to seven deuterium atoms; R2 is an ethyl group having zero to five deuterium atoms, and when each R has zero deuterium atoms, at least one Z is deuterium.

One embodiment of this invention provides compounds of Formula I wherein Rl is selected from CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2-, CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-. In a more specific embodiment, Rl is CD3CD2CD2- or

CD3CD2CH2-. In one aspect of these embodiments, Zl and Z2 are both hydrogen. In another aspect of these embodiments, Zl and Z2 are both deuterium. In another enibodinient, R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In a more specific embodiment, R2 is selected from CH3CH2- or CD3CD2-. In one aspect of these embodiments, Zl and Z2 are both hydrogen. In another aspect of these embodiments, Zl and Z2 are both deuterium.

The R and Z variables as described above may be selected and taken together to provide more specific embodiments of this invention. For example, in one embodiment, Rl is CD3CH2CH2-, CD3CD2CH2-, CD3CH2CD2-, CH3CH2CD2- , CH3CD2CD2-, CD3CD2CD2- or CH3CH2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In one aspect of this embodiment. R2 is CH3CH2- or CD3CD2-. [0039] In another embodiment, Rl is

CD3CD2CD2- or CD3CD2CH2-; and R2 is selected from CH3CH2-, CD3CH2-, CH3CD2-, or CD3CD2-. In one aspect of this embodiment, R2 is CH3CH2- or CD3CD2-.

Examples of specific compounds of this invention include the following:

viii) 20090312333

The compounds of the present invention are those covered by formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.

Rl is hydrogen, substituted or unsubstituted Cl-12 alkyl, substituted or unsubstituted aryl or substituted or unsubstituted 3-8 membered heterocycle.

R2 is hydrogen. Alternatively, Rl and R2 may be linked together in such a way to form a C3-6 cycloalkyL R3 is either

( a) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its C atoms, said heterocycle is selected from the group consisting of: lH-benzirnidazol-6-yl;

lH-benzirnidazol-7-yl;

imidazo[l ,2-a]pyridin-3-yl;

imidazo [ 1 ,2-a]pyriniidin-3 -yl;

imidazof 1 ,2-b] [ 1 ,2,4]triazhi-7-yl;

irnidazo[l ,2-b]pyridazin-3-yi;

5,6,7,8-tetrahydroiniidazo[l.,2-b]pyridazin-3-yl;

imidazo[2,l -b][l,3,4]thiadiazol-5-yi;

imidazo[2,l -b][l,3]thiazol-5-y[;

3H-imidazo[4.5-b]pyridin-7-yl;

1 H-imi dazol-4-yl;

1 H-imidazol-5-yl;

1 H-indol-2-yl; lH-indol-3-yl;

lH-indol-4-yl;

l H-indol-7-yl;

isoxazol-4-yl;

l H-pyrazoi-4-yl;

lH-pyrazol-5-yl; 1 H-pyrazolo [1,5 -a]pyrirnidin-3 -y 1;

1 H-pyrazolo [3 ,4-b] pyridm~3 -y 1 ;

pyridazin-4-yl;

pyridin-2-yl; pyridin-3-yl;

pyridin-4-yl;

1 H-pyrrolo [2,3 -b] pyridin-3 -y ί ;

l H-pyrrolo[2,3-b]pyridin-4-yi;

l H-pyrrolo[2,3-b]pyridin-5-yi;

1 H-pyrrolo [2,3 -c]pyridin-2-yl;

lH-pyrrolo[2,3-c]pyridin-3-yl;

1 H -pyrrol o [3 ,2-b jpyridin-3 -yl;

1 H-pyrro 1 o [3 ,2-c] pyridin-2-y 1 ;

1 H-pyrrolo [3 ,2-c]pyridin-3 -yl;

l,3,4-thiadiazol-2-yl;

l,3-thiazol-5-yl;

[ 1 ,2,4] iriazolo [4,3 ~b] pyri dazin-7-yl;

[1 ,2,4]triazolo[4,3-b]pyridazin-8-yl;

indolizin-3-yi;

or R3 is

(b) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms, said heterocycle is selected from the group consisting of:

1 H- 1 ,2,3-benzotriazol- 1 -yl;

1 H-imidazo [4,5 -bjpyridin- 1 -yl;

3 H-imidazo [4,5 -b]pyridin-3 -yl; 7H-imidazo[4.5-c]pyridazin-7-yl;

lH-indol-l-yl;

2.3- dihydro- 1 H-indol- 1 -yl;

9H-purin-9-yl;

1 H-pyrazolo [3 ,4-b]pyridin- 1 -y 1;

2H-pyrazolo [3 ,4 -b]pyridin-2-yl;

1 H-pyrrolo[2,3-b]pyridm- 1 -yl;

1 H-pyrrolo[3 ,2-b]pyridin- 1 -yl;

3.4- dihydroquinolin- 1 (2H)-yl;

8H-isothiazolo[5,4-b]indol-8-yl;

1 H- 1 ,2 ,4-triazol- 1 -yl;

lH-pyrrol-l-yl;

2-ehi oro- 1 H -benzimidazol- 1 -yl .

R4 in formula (I) is selected from the group comprising or consisting of hydrogen; Cl-12 alkyl optionally substituted by halogen, Cl-4 alkoxy, Cl-4 alkyltliio, azido, nitrooxy or an aryl; C2-12 alkenyl optionally substituted by halogen; C2-12 alkynyl optionally substituted by halogen; azido; alkoxycarbonylamino;

arylsu!fonyloxy; a substituted or ^substituted aryl; or a 3-8 membered substituted or uiisubstituted heterocycle:

In a specific embodiment R4 is hydrogen; or R4 is Cl-12 alkyl or a CI -6 alkyl, optionally substituted by halogen, Cl-4 alkoxy, Cl-4 alkylthio, azido or nitrooxy; or R4 is C2-12 alkenyl or a Cl-6 alkenyl optionally substituted by halogen; or R4 is C2-12 alkynyl or a CI -6 alkynyl optionally substituted by halogen; or R4 is alkoxycarbonylamino,

R5 is hydrogen;

Alternatively R.4 may form together with R5 and the 2-oxo-l -pyrrolidine ring a l,3-dihydro-2H-indol-2-one ring of the following structure:

The asterisk * indicates the point of attachment of the substituents;

R6 is hydrogen or halogen.

R7 in formula (!) is selected from the group comprising or consisting of hydrogen; nitro; halogen; heterocvcle; amino; aryl; Cl-12 alkyl optionally substituted by at least one halogen; or Cl-12 aikoxy optionally substituted by at least one halogen,

R8 in formula (I) is sel ected from the group comprising or consisting of hydrogen, Cl-12 alkyl optionally substituted by halogen, or halogen.

R9 in formula (I) is selected from the group comprising or consisting of hydrogen, Cl-12 alkyl optionally substituted by halogen, or halogen.

A further aspect of the present invention consists in compounds of formula (I) wherein

Rl and R2 are both hydrogen. R3 is:

(a) a substituted or unsubstituted heterocvcle linked to the rest of the molecule via one of its C atoms selected from the group consisting of:

1 H-benzimidazoi-6-yl ;

1 H-benzimidazol-7-yl;

imidazo[ 1 ,2-a]pyridin-3-yl;

imidazo [ 1 ,2-a]pyriniidin-3 -yl;

imidazo[ 1 ,2-b] [ 1 ,2,4]triazin-7-yl ;

imidazo [ 1 ,2-b]pyridazin-3 -y 1 ;

5,6,7 5 8-tetrahydroimidazo[l ,2-b]pyridazin-3-yl; imidazo[2, 1 -b] [ 1 ,3 ,4] thiadiazol- imidazo[2, 1 -b] [ 1 ,3 jthiazol-5-yl;

3 H-imidazo[4,5-b]pyridin-7-yl ; lH-imidazol-4-yl;

lH-imidazol-5-yl;

lH-indol-2-yl;

l H-indol-3-yl ;

l H-indol-4-yl;

l H-indo!-7-yl;

isoxazol-4-yl;

lH-pyrazol-4-yl;

lH-pyrazol-5-yl;

1 H-pyrazolo[l ,5-a]pyrimidin-3- lH-pyrazolo[3,4-b]pyridin-3-yl; pyridazin-4-yl;

pyridin-2-yl;

pyridin-3-yl;

pyridin-4-yl;

1 H-pyrrolo [2,3 -b]pyridin-3 -y [ ; lH-pyrrolo[2,3-b]pyridin-4-y[; lH-pyrrolo[2,3-b]pyridin-5-yl; 1 H~ pyrrol o [2 ,3 -c] pyri din -2 -y 3 ; 1 H-pyrro ! o [2 ,3 -c] pyridin-3 -y 1; 1 H-pyrrolo [3 ,2-b]pyridin-3 -yl; 1 H-pyrro 1 o [3 ,2-c]pyridin-2-yl; lH-pyrrolo[3,2-c]pyridin-3-yl;

1 ,3 ,4-thiadiazol-2-yl;

1.3- thiazoi-5-yl;

[ 1 ,2,4]triazolo[4,3-b]pyridazin-7-yl;

[ 1 ,2,4]triazolo[4,3-b]pyridazin-8-yl;

hidoliziii-3-yl,

Alternatively R3 is:

(b) a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its atoms selected from the group consisting of:

1 H- 1 ,2 ,3 -benzotriazol- 1 -yl;

1 H-imi dazo [4,5 ~b] pyri din- 1 -y 1 ;

3 H-imi dazo [4,5 -bjpyri din-3 -y 1;

7H-imidazo[4,5-c]pyridazin-7-yl;

lH-indol-l-yl;

2,3-dihydro-lH-indol-l-yl;

9B-purm-9~yl;

1 H-pyrazolo[3,4-b]pyridin-l -yl;

2H-pyrazolo[3,4-b]pyridin-2-yl;

lH-pyrrolo[2,3-b]pyridin- 1 -yl;

1 H-pyrroio [3 ,2-b]pyridin- 1 -yl;

3.4- dihydroquino in- 1 (2H)-y 1 ;

8H-isothiazolo[5,4-b]indol-8-yl;

lH-l,2,4-triazol-l-yl;

lH-pyrrol-1-yl;

2-cliloro- 1 H-benzimidazol- 1 -yl. R4 in formula (I) is selected from the group comprising or consisting of hydrogen; CI-12 alkyl optionally substituted by halogen or Cl-4 alkoxy; C2-12 aSkenyl optionally substituted by halogen; C2-12 alkynyl optionally substituted by halogen.

In a further specific embodiment R4 is n-propyi, 2,2,2-trifluoroethyl, 2-chloro-2,2- difluoroethyi 2 bromo-2,2-difiuoroethyl, 2,2-difluorovinyl.

In another specific embodiment R4 is phenyl, 2,3,5-trifluorophenyl or 3-chloro-4- fiuorophenyl.

R5 is hydrogen;

A further embodiment of the present invention consists in compounds of formula (I) wherein R4 forms together with R5a l,3-dihydro-2H-indol-2-one ring

The asterisk * indicates the point of attachment of the heteroaryl alkylene substituent, and wherein

R6 is hydrogen;

R7 is chlorine;

R8 is hydrogen;

R9 is hydrogen.

A further embodiment of the present invention consists in compounds of formula CD wherein R3 is a substituted or unsubstituted heterocvcle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of:

irnidazo[l,2-a]pyrimidin-3-yl;

imidazofl ,2-b] [ 1 ,2,4]triazin-7-yl;

imidazof 1 ,2-b]pyridazm-3-yl; 5,6,7,8-tetrahydroimidazo[l,2-b]pyridazin-3-yl:

imidazo[2, 1 -b] [ 1 ,3,4]thiadiazol-5~yl;

imidazo[2, 1 -b] [ 1 ,3]th azol-5~yi;

3H-imidazo[4,5-b]pyridin-7-yl;

1 H-imidazol-4-yl;

lH-imidazol-5-yl;

isoxazol-4-yl;

l H-pyrazoi-4-yl; l H-pyrazoi-5-yl;

1 H-pyrazolo [1,5 -a]pyrimidin-3 -yl;

lH-pyrazolo[3,4-b]pyridin-3-yl:

pyridin-3-yl;

1 H-pyrro 1 o [2,3 -b]pyridin-3 -yl;

1 H-pyrrol o [2,3 -b]pyridin-4-yl; 1 H-pyrrol o [2,3 -b]pyridin-5 -yl;

lH-pyrrolo[2,3-c]p>Tidin-2-yl;

l H-pyrrolo[2,3-c]pyridm-3-yl;

1 ,3-thiazol-5-yl;

[l,2,4]triazolo[4,3-b]pyridazin-8-yl;

indolizin-3-yi.

In a further specific embodiment R3 is a heterocvcle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: imidazo [ 1 ,2-b]pyridazin-3 -y 1 ;

imidazo[2, 1 -b] [ 1 ,3„4]thiadiazol-5-yl;

imidazo[2, 1 -b] [ 1 ,3]thiazol-5-yl; 3H-imidazo[4.5-b]pyridin-7-yl;

1 H-imi dazol-4-yl;

1 H-imidazol-5-yl;

lH-pyrazol-4-yl; lH-pyrazolo[ 1 ,5-a]pyrimidin-3-yl;

pyridin-3-yl;

1 H-pyrrolo [2,3 -b]pyridm~3 -y ί ;

l H-pyrrolo[2,3-b]pyridin-4-yi; 1 ,3-thiazol-5-yl;

Said heterocycles are optionally substituted by e.g. a methyl, n-propyl,

trifiuoromethyl, cyclopropyi, bromine, chlorine, fluorine, iodine, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyi oxy, cyciopropylmethoxy, cyclobutylmethoxy, amino, rn.ethylarn.ino, cyclopropyi amino, cyclobutylamino, l-pyrrolidmyl, cyano, phenyl, benzyl or 3-thienyl.

In a further specific embodiment R3 is a heterocycie linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: 6- chloro-2-cyclopropylimidazo[l ,2-b]pyridazin-3-yl, 6-(cyciopropyloxy)-2- (trifluoromethyl)imidazo[ 1 ,2~b]pyridazin~3-yi, 6-propoxy-2- (trifluoromethyl)imidazo[ 1 ,2-b]pyridazin-3-y[, 6-chloroimidazo[2, 1 -b] [ 1 ,3]thiazol- 5-yl. 2,6-dichloroimidazo[2,l-b][l,3]thiazol-5-yl, 5-chloro-lH-imidazol-4-yl, 5- bromo-1 H-imidazoi-4-yl, 4-bromo-lH-imidazoi-5-yl, 4-chloro-lH-imidazoi-5-yl, 1 H-imidazol-5 -yl, 1 -methyl- 1 H-imidazol-5 -yl, -chloro- 1 -methyl- 1 H-imidazol-5 - yi, 1 H-pyrazoi-4-yl, .1 H-pyrrolo[2,3-b]pyridin-3-yl.

A further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycie linked to the rest of the molecule via one of its C atoms and is a substituted or unsubstituted imidazo[1 ,2-a]pyridin-3-yl.

Said imidazo[l,2-a]pyridin-3-y[ is optionally substituted by e.g. a methyl, cyclopropyi, bromine, chlorine, fluorine, iodine. In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of: imidazofl ,2-a]pyridin-3-yl, 6-methylimidazo[i ,2-a]pyridin-3-yl, 2- chloroimidazo[ 1 ,2-a]pyridin-3-yl.

A further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a substituted or unsubstituted heterocycle linked to the rest of the molecule via one of its N atoms and is selected from the group consisting of:

3H-imidazo [4,5 -b]pyridin-3 -yl; lH-indol- l-yl;

lH-pyrrolo[2,3-b]pyridin- 1 -yl:

l H-pyrrolo[3,2-b]pyridm-l-yl;

l H-pyrrol-l -yl;

2-chloro- 1 H-benzimidazol- 1 -yl.

A specific further embodiment of the present invention consists in compounds of formula (I) wherein R3 is a heterocycle linked to the rest of the molecule via one of its atoms and is selected from the group consisting of:

3H-in idazo [4,5 -b]pyridin-3 -yl;

l H-pyrrolo[3,2-b]pyridm-l-yl;

l H-pyrrol-l -yl;

2-chloro- 1 H-benzimidazol- 1 -yl;

Said heterocycles may optionally be substituted by trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, methoxy or cyano.

In a further specific embodiment R3 is a heterocycle linked to the rest of the molecule via one of its C atoms and is selected from the group consisting of 6- bromo-2-chloro-3H-imidazo[4,5-b]pyri.din-3-yl, 6-bromo-2-cyclopropyl-3H- imidazo[4,5-b]pyridin-3-y[, lH-pyrrolo[3,2-b]pyridin-l-yl, 2,5-dichloro-lH- pyrro -1 -yl, 2-chloro-5-methoxy-lH-benzirnidazol-l -yl, 5-brom.o-2-chloro-l H- benzimidazol- 1 -yl or 2,5-dichloro- 1 H-benzimidazol- 1 -yl. A further embodiment of the present invention consists in compounds of formula CD wherein Rl, R2 and R5 are hydrogen.

R4 is a CI -6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen,

R3 is selected from the group consisting of;

imidazo [ 1 ,2 -b]pyri dazin-3 -yl;

imidazo[2, 1 -b] [ 1 ,3 ,4]thiadiazol-5-yl;

imidazo[2, 1 -b] [ 1 ,3]thiazol-5-yl;

3H~imidazo[4,5-b]pyridin-7-yl;

1 H-imi dazol-4-yl;

1 H-imidazol-5-yl;

lH-pyrazol-4-yl;

1 H-pyrazolo [ 1 ,5 -a]pyrimidin-3 -yl;

pyridin-3-yl;

1 H-pyrrolo [2,3 -b] pyridin-3 -y 1 ;

l H-pyrrolo[2,3-b]pyridin-4-yi;

l,3-thiazol-5-yl;

and optionally substituted by methyl, n-propyl, trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, iodine, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cvclopropylmethoxv, cyclobutylmethoxy, amino, methylamino, cyclopropylamino, cyclobutylamino, l-pyrrolidinyl, cyano, phenyl, benzyl or 3- thienvl.

A further embodiment of the present invention consists in compounds of formula (D wherein Rl, R2 and R5 are hydrogen,

R4 is a CI -6 alkyl optionally substituted by halogen, a C2-6 alkenyl optionally substituted by halogen or C2-12 alkynyl optionally substituted by halogen.

R3 is selected from the group consisting of 3H-imidazo[4.5-b]pyridin-3-yl;

1 H pyrrolo[3 ,2-b] pyridin- 1 -yi ;

I H-pyrro!-l-yl;

2-chioro- 1 H-benzimidazol- 1 -yl;

optionally substituted by trifluoromethyl, cyclopropyl, bromine, chlorine, fluorine, methoxy or cyano.

A further embodiment of the in vention consists in compounds of formula (I), their diastereomers and mixtures, or a pharmaceutically acceptable salt thereof.

Rl, R2 and R5 are hydrogen.

R3 is a substituted or unsubstituted heterocvcle linked to the rest of the molecule via one of its C atoms, said heterocvcle is selected from the group consisting of:

1 H-benzimidazol-6-yl;

1 H-benzimidazoi-7-y 1;

irnidazo[l ,2-a]pyridm-3~yl; imidazo[l .,2-a]pyrimidin-3-yl;

imidazofl ,2-b] [ 1 ,2,4]tr azm-7~yl;

imidazof 1 ,2-b]pyridazm-3-yl;

5,6,7,8-tetrahydroimidazo[l,2-b]pyridazin-3-yl:

imidazo[2,l-b][l ,3,4]thiadiazol-5-yl;

imidazo[2, 1 -b] [ 1 ,3 jthiazol-5-yl;

3 H-imi dazo [4,5 -b]pyri din-7-y 1; 1 H-imidazol-4-yl;

l H-imidazol-5-yl;

l H-indol-2-yl;

lH-indol-3-yl;

lH-indol-4-yl;

lH-indol-7-yl;

isoxazol-4-yl;

l H-pyrazoi-4-yl;

3 M-pyrazoi-5-yl;

1 H-pyrazolof 1 ,5-a]pyrimidin-3-yl; lH-pyrazolo[3,4-b]pyridin-3-yl: pyridazin-4-yl:

pyridin-2-y];

pyridin-3-yl;

pyridin-4-yl;

1 H-pyrrolo [2,3 -b] pyridin-3 -yl: l I-.l-pyrrolo[2,3-b]pyridm-4-yl; l H-pyrrolo[2,3-b]pyridin-5-yi;

! H-pyrrolo[2,3-c]pyridin-2-yl; 1 H-pyrrolo[2,3-c]pyridin-3-yl; lH-pyrrolo[3,2-b]pyridin-3-yl; lH-pyrrolo[3,2-c]pyridin-2-yl;

I H-pyrrolo[3,2-c]pyridin-3-yU l,3,4-thiadiazol-2-yl;

l,3-thiazoi-5-yl; [ 1 ,2,4]triazolo[4,3-b]pyridazin-7-yl;

[l,2,4]triazolo[4,3~b]pyrida.zin-8~yl;

indoiizin-3-yl;

Particularly preferred are imidazo[l,2-a]pyridin-3-yl; imidazo[l,2-a]pyrimidin-3- yl; imidazo[I.2-b]pyridazm-3-yl; lH-imidazol-4-yl; lH-imidazol-5-yl;

R4 is a substituted or unsubstituted phenyl moiety;

A further embodiment of the present invention consists in compounds of formula (I) wherein Rl is hydrogen or Cl-12 alkyl;

R2 is hydrogen;

R3 is an aromatic 5-mernbered heterocycle linked to the rest of the molecule via one of its C atoms;

R4 is hydrogen, Cl-12 alkyl or aryl;

R5 is hydrogen;

Alternatively, R4 can form together with 1 5 and the 2-oxo-l -pyrrolidine ring the following l ,3-dihydro-2H-indol-2-one cycle

wherein the asterisk * indicates the point of attachment of the subslituents;

R6 is hydrogen or halogen;

In this embodiment R4 may not be hydrogen when R3 is substituted lH-pyrazol-5- yl. Also this embodiment does not comprise 5-(2'-oxo- -pyrrolidinyl)metbyl- 1 ,3 ,4-tricarbomethoxy-pyrazole which is disclosed in A. Padwa et al J . Org. Chera. 2000, 65, 5223-5232 without any biological activity though.

In this embodiment wherein R3 is an aromatic 5-membered heterocycle linked to the rest of the molecule via one of its C atoms, specific moieties R3 may be selected from i,3-thiazol-5-yl, lH-imidazol-4-yl, lH-imidazol-5-yl, lH-pyrazol-4- yl, l H-pyrazol-5-yl, 2~oxo-2,3-dihydro~l,3~thiazol~5~yS, each of them being optionally substituted by 1 to 3 substituents independently selected from methyl, chlorine, bromine, amino, methylamino, dimethy!amino, (2-oxo-4-propyl- pyrrolidin- l-yl)methyl, 1-pyrrolidinyl, amido, cyano, methoxy, phenyl, 4- methylphenyl-sulfonyl, benzyl or 2-(benzylamino)~2-oxoethyL

In this embodiment, more specific moieties R3 are selected from 2-(methylamino)- 1 ,3 -thiazol-5 -yl; 2-pyrrolidin- 1 -y 1- 1 ,3 -thiazol-5 -yl; 5 -bromo- 1 H-imidazol-4 -yl: 5 - chloro-1 H-imidaz.ol-4-yl; 1 H-imidaz.ol-5-yl; 1 -methyl-l H-imidazol-5-yl; 4-bromo- 1 -methyl- 1 H-imidazol-5 -y 1; 4-chloro- 1 H-imidazol-5-yl; 4-chloro- 1 -methyl- 1 H- imidazoi-5-yl; 4-cyano-l -methyl-l H-imidazol-5-yl; 1 H-pyrazol-4-yl; 3,5- dimethyl- 1 H-pyrazol-4-y 1 ; 3 -methyl- 1 H-pyrazol-4-yl.

In this embodiment, most specific moieties R3 are selected from 5-bromo-lH- imidazol-4-yl; 5-chloro-l H-imidazol-4-yl; 1 H-imidazol-5-yl; 4-bromo- 1 -methyl- lH-imidazol-5-yl; 4-chloro- 1 -methyl-l H-imidazol-5-yl; lH-pyrazol-4-yl.

Still in this embodiment, a specific moiety Rl is selected from hydrogen or ethyl,

Still in this embodiment, a specific moiety R4 is selected from hydrogen, n-propyl, 2,3,5-trifluorophenyl or phenyl.

A further embodiment of the present invention consists in compounds having the specific formula (la),

In formula (la) the substituent RIO is hydrogen; halogen: CI -4 alkyl optionally substituted by at least one halogen; CI -4 alkoxy; methoxycarbonyl; nitro; amino; alkylamino; amido; or alkanoyl-aniino. Preferably RIO is hydrogen. Rl 1 is hydrogen; halogen; CI -4 alkyl optionally substituted by at least one halogen; CI -4 alkoxy; methoxycarbonyl; nitro; amino; aikyiamino; amido; or aikanoyiamino. Preferably Rl 1 is hydrogen.

R4 is C I -4 alkyl optionally substituted by at least one halogen; or C2-4 alkenyl optionally substituted by at least one halogen. Preferably 1 4 is n-propyl.

Still in this aspect of the invention a specific embodiment relates to an embodiment wherein RIO is selected from hydrogen; methyl; fluorine; chlorine; bromine;

methoxy; methoxycarbonyl; nitro; or trifluoromethyl, while Rl 1 is selected from hydrogen; methyl; fluorine; chlorine; bromine; methoxy; methoxycarbonyl; nitro; or trifluoromethyl; and R3 is n-propyl.

Specific compounds of the present invention are those selected from the group consisting of: l-[(l -methyi-l H-benzimidazol-6-yl)methyi]-4-propylpyrrolidin-2-one;

1 -( 1 H-benzimidazol-7-ylmethyl)-4-propylpyiTolidm-2-one;

1 -(imidazo[ 1 ,2-a]pyridin-3-ylmethyl)-4-propylpyrrolidm-2-one;

1 - {[6-chloro-2-(4-methyiphenyi)imidazo[ 1 ,2-a]pyridin-3-yl]methyl} -4- propy Spyrro li din -2-one ;

1 - { [2-(4-ch3orophenyl)-6-methylimidazo[ 1 ,2-a]pyridin-3-yl]methyl} -4- propylpyrrolidin-2-one;

l-[(5-methylimidazo[l,2-a]pyridin-3-yl)methyl]-4-phenylpy rro3idm-2-one;

1 -(imidazo[ 1 ,2-a]pyridin-3-ylmethyl)-4-ph.enylpyrrolidin-2-one;

l-[(6-methy[irrridazo[l,2-a]pyridin-3-yl)m

1 -[(6-bromoimidazo[ 1 ,2-a]pyridin-3-yl)methyi]-4-propylpyrroiidin-2-one;

l-[(8-nietiiylimidazo[l,2-a]py l -[(6-iodoimidazo[l ,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidm-2-one;

l -{[8-chloro-6-(trifluoroniethyl)imidazo[l,2-a]pyridin-3-yl]m ethyl}-4- propy lpyiTolidin-2 -one ; l-[(7-methylimidazo[l,2-a]pyridin-3-yl)methyl]-4-propy3pyrro lidin-2-one; l-[(6,8-dibromoimidazo[l,2-a]p>Tidin-3-yl)methyl]-4-propy lpyrrolidin-2-one: l-[(6,8-dichloroimidazo[l ,2-a]pyridin-3-y3)rafithyl]-4-propylpyTTolidin-2-one; l-[(6-chloroimidazo[l.,2-a]pyridin-3-yl)i ' nethyl]-4-propylpyrrolidin-2-one;

1 -[(2-chloroimidazo[ 1 ,2-a]pyridin-3-yl)methyl]-4-propylpyrrolidm-2-one;

1 -[(2-cyclopropyl-6-fluoroimidazo[ 1 ,2-a]pyridin-3-yl)methyl]-4-(2,2- difl uoro vinyl)pyrrol idin-2-one;

l -[(6-chloro-2-cyclopropylimidazo[l ,2-a]pyridin-3-yI)methyl]-4-(2,2- difluorovinyl)pyrrolidin-2-one;

1 -(imidazo[ 1 5 2-a]pyrimidin-3-ylmethyl)-4-propylpyrrolidin-2-one;

1- {[2-(4-chlorophenyl)imidazo[l,2-a]pyrimidin-3-yl]methyl} -4-propyl pyrrolidin-

2- one;

1 -(imidazo[ 1 ,2-a]pyrimidin-3-ylmethyl)-4-phenylpyrrolidin-2-one;

l -[(6-chloroimidazo[l ,2-a]pyrirnidin-3-yl)methylj-4-propylpyrrolidin-2-one; l -{[6-chloro-2-(trifluoroniethyl)imidazo[l,2-a]pyrimidin-3-yl ]methyl}-4- propylpyrrolidin-2-one;

1 -[(6-phenylimidazo[ 1 ,2-b] [ 1.2,4]triazin-7-yl)methyl]-4-propylpynOlidin-2-one;

1 - { [6-chloro-2-(4-methylphenyl)irnidazo[ 1 ,2-b]pyridazm-3-yl]metbyl} -4- propylpyrrolidin-2-οηε;

1 - { [6-chloro-2-(4-chlorophenyl)imidazo [ 1 ,2-b]pyridazin-3 -yl]methy 1 } -4- propylpyrrolidm-2-one;

l -[(6-chloroiiriidazo[l ,2-b]pyridazin-3-yl)niethyl]-4-propylpyrrolidin-2-one; l-[(6-c oroimidazo[l,2-b]pyri(iazin-3-yl)methyl]-4-phmy^^

1 - {[6-chloro-2-(trifluoromethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl} -4- propylpyrrolidin-2-one;

l- {[6-chloro-2-(trifluoromethyl)imidazo[l ,2-b]pyridazin-3-yl]niethyl}-4-(2,3,5- trifluoropheiiyl)pyrrolidin-2-one; 1- {[6-chlorQ-2-(trifluoromethyl)imid

2- one;

1 - { [6-chloro-2-(trifluoromethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl} -4-(2,2- difl uoro vinyl)pyrrol idin-2-one;

l -{[6-chloro-2-(trifluorom€thyl)ira azo[l,2-b]pyridazin-3-yl]methyl}-4- phenylpyiTolidin-2-one;

5-chloro-l-{[6-cWoro-2-(lTifluoromethyl)imidazo[l,2-b]pyrida zin-3-yl]methy 1 ,3 -dihydro-2 H -indo I -2-one;

1 - { [6-methoxy-2-(trifluoromethyi)imidazo[ 1 ,2-b]pyridazin-3-y 1 jmethyl} -4- propylpyrrolidin-2-οηε;

l -[(6-cbloro-2-cyclopropy3.imidazo[l ,2-b]pyridazin-3-yl)ni.ethyl]-4- propylpyrrolidin-2-one;

l-{[6-isQpropoxy-2-(trifiuoro^

propylpyrrolidin-2-one;

1- {[6-(benzyloxy)-2-(trifluoromethyl)imidazo[l ,2-b]pyridazin-3-yl]methyl}-4- propylpyrrolidin-2-οηε;

l -{[6-cyclopropyl-2-(triiluoromethyl)iraidazo[l,2-b]pyridazm- 3-yl]metbyl}-4- propylpyrrolidin-2-one;

1 - { [6-(dimethylamino)-2-(trifluoromethy l)imidazo [ 1 ,2-b jpyridazin-3 -yl jmethyl} - 4-propylpyrrolidin-2~one; -{[6-methoxy-2-(trM

3- yl]methyl}pyrrolidin-2-one;

4- (2-chloro-2,2-diiluoroethyl)-l-{[6-chloro-2-(trifluoromethyl )imidazo[l,2- b]pyridazm-3-yl]methyl}pyrrolidin-2-one;

1 - { [6-(methylamino)-2-(trifluoromethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl} -4- propylpyrrolidin-2-one;

1 - {[6-hydroxy-2-(trifluoromethyi)imidazo[ 1 ,2-b]pyridazin-3-y ljmethyl} -4- propylpyrrolidin-2-οηε; l-{[6-(methylthio)-2-(trifl^

propylpyrrolidin-2-one;

4-(2-bromo-2,2-difluoroethyl)- 1 - { [6-chloro-2-(trifluoromethyl)imidazo[ 1 ,2- b]pyridazin-3-yl]methyl}pyrrolidm-2-one;

1 - { [6-(methylsulfonyl)-2-(trifluorom.ethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl } - 4-propylpyrrolidin-2-one;

1 - { [6-(methylsulfiny l)-2-(trifluoromethyl)imidazo [ 1.2-b]pyridazin-3 -yljniethy 1 } -4- propylpyrrolidin-2-one;

1 - { [6-chloro-2-(trifluoromethy l)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl} -4-(2,2,2- trifluoroethyl)pyrrolidin-2-one;

l -[(6-chloro-2-cyclobutylimidazo[l,2-b]pyridazin-3-yl)methylj -4- propylpyrrolidin-2-one;

l-{ [6-chloro-2-(4-methylphenyl)imidazo[l ,2-b]pyridazin-3-yl]methyl} -4-(2,2- di fluorovi ny ! )pyrroli di n-2-one ;

1 - { [6-amino-2-(trifiuoromethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl} -4- propylpyrrolidin-2-οηε;

l -{[6-(ethylamino)-2-(triiluoromethyl)imidazo[l,2-b]pyri4azm- 3-yl]methyl}-4- propylpyrrolidin-2-one;

4-propyl- 1 - {[6-(propylamino)-2-(trifluoromethyl)imida.zo[ 1 ,2-b]pyridazin-3- yl]methyl}pyTrolidin-2-one;

4-( 2-bromo~2,2~difl oroethyl)~ 1 - { [6-(propylamino)-2- (trifluoromethyl)imidazo[l,2-b]pyridazin-3-yl]methyl}pyrroli din-2-one;

4-(2,2-difluoro vinyl)- l-{[6-(propylamino)-2-(trif3.uorometbyl)imidazo[ 1,2- b]pyridazin-3-yl]methyi}pyrrolidin-2-one;

4-(2,2-difluorovinyl)-l- {[6-methoxy-2-(4-methylphenyl)imidazo[l ,2-b]p>T"idazin-

3- yl]rneihyl}pyrrolidm-2~one;

4- propyl- - {[6-pyrrolidin-l-yl-2-(trifluoromethyl)imidazo[l,2-b]pyridaz in-3- yl]methyl}pyrrolidin-2-one; 4-(2-bromo-2.2-difluoroethyl)- 1 - { [6-methoxy-2-(trifluoromethyl)imidazo[ 1 ,2- b]pyridazin-3-yi]methyl}pyrrolidin-2-one;

1 - { [6-(cyclopropylamino)-2-(trifluoromethyl)imidazo [ 1 ,2-b]pyridazin-3 - yl]methyl}-4-(2,2-difl ' uorovinyl)pyrrolidin-2-one;

1 -[(6-cbloro-2-cyclopropyl.imidazo[ 1 ,2-b]pyridazin-3-yl)methyl]-4-(2,2- difluorovinyl)pyrrolidin-2-one;

4-(2,2-difluOTovinyl)-l-{[6-(isopropylamino)-2-(trifluOTomet hyl)imidazo[1.2- b]pyridazin-3-yi]methyl}pyrrolidin-2-one; -{[2-(trifluoromethyl)imidazo[l ,2-b]pyridazin-3- yl]methyl}pyrrolidin-2-one;

1 -{[2-cyclopropyl-6-(propylamino)imidazo[l ,2-b]pyridazin-3-yl]methyl}-4-(2,2~ difluorovinyl)pyrrolidin-2-one;

l-({2-cyclopropyl-6-[(2-fluoroethyl)aniin^

4-(2,2-difiuoro 'inyl)pyrrolidin-2-one;

l-( {2-cyclopropyl-6-[(2,2-difluoroethyl)amvno]imidazo[l ,2-b]pyridazin-3- yl}methyl)-4-(2,2-difluorovinyl)pyrrolidin-2-one;

1 ~( {2-cyclopropyl-6-[(2,2,2-trifluoroethyl)ammo]imidazo[ 1 ,2-b jpyridazin-3- yl}methyl)-4-(2,2-difluorovinyr)pyrrolidin-2-one;

4-(2 ,2-difluoroethyl)- 1 - { [2-(trifluoromethyl)imidazo [ 1 ,2-b]pyridazin-3 - yl]methyl}pyTrolidin-2-one;

l-{[2-cyc!opropyl-6-(cyclopropylamino)imidazo[l ,2-b]pyridazin-3-yl]methyS}-4- (2 5 2-diiluorovinyl)pyrrolidin-2-one;

l -[(6-cbloro-2-cyclobutylimidazo[l,2-b]pyridazin-3-yl)methylj -4-(2,2- difluorovinyl)pyrrolidin-2-one;

l-[(6-chloro-2-cyclopropylimidazo[l ,2-b]pyridazin-3-yl)methyl]-4-(3-ch[oro-4- fluorophenyl)pyrrolidin-2-one;

l- {[6-(butylaniino)-2-(trifluoromethyl)imidazo[l,2-b]pyridazin -3-yl]methyl}-4 (2 5 2-diiluorovinyl)pyrrolidin-2-one; 1 - { [6-(cyclobuty lamino)-2-(lTifluoromethy l)imidazo [ 1 ,2-b]pyridazin-3 - yi]methyl}-4-(2,2-difluorovinyl)pyrroiidin-2-one;

1 -[(2-cyclopropyl-6-methoxyimidazo[ 1 ,2-b]pyridazin-3-yl)methyl]-4-(2,2- difl uoro vinyl)pyrrol idin-2-one;

4-(2,2-difluorovmyl)-l-{[6-et oxy-2-(trifluoromethyl)imidazo[l,2-bjpyrida m-3- yljmethyl } pyrrol idm-2-one;

4-(2,2-difluOTovinyl)-l-{[6-isopropoxy-2-(trifluoromethyl)im idazo[l,2- b]pyridazin-3-yi]methyl}pyrrolidin-2-one;

l- {[6-(cyclopropylmethoxy)-2-(trifluoromethyl)imidazo[l,2-b]py ridazin-3- yljmethyl} -4-(2,2-difluorovinyl)pyrrolidin-2-one;

l -{[6-(cyclobutylmethoxy)-2-(trifluoromethyl)imidazo[l,2-bjpy ridazin-3- yl]methyl}-4-(2,2-difluorovinyl)pyrTolidin-2-one;

1 - { [6-(cyclopropyloxy)-2-(trifluoromethyl)imidazo[ 1 ,2-b]pyridazin-3-yl]methyl}- 4-(2,2-difiuoro 'inyl)pyrrolidin-2-one; - {[6-propoxy-2-(trifluoromethyl)imidazo[l ,2-b]pyridazin-3- yl]methyl}pyrrolidin-2-one;

3_ [4-(2,2-diiluorovinyl)-2-oxopyrrolidin- 1 -yljmethyl} -2- (trifluoromethyl)imidazo [ 1 ,2-b Jpyridazine-6-carbonitrile;

4-(2 ,2-difluoroviny 1)- 1 - { [6-thien-3 -yl-2-(trifluoromethy l)imidazo [ 1 ,2-b]pyridazin- 3-ylJm.ethyl}pyrrolidin-2-one; -{[6-phenyi-2-(trifluoromethyl)iniidazo[l,2-bJpyridazin-3- yl]methyl}pyrrolidin-2-one;

4-(2,2-difluorovmyl)-l-{[6-methyl-2-(lTifluoromethyl)imidazo [l ,2-bJpyridazin-3- yljmethyl } pyrro 1 idin-2-one;

4-(2,2-difluorovinyl)-l- {[6-pyridin-3-yl-2-(trifluoromethyl)imidazo[l ,2- bJpyridazm-3-ylJmethyl}pyrrolidin-2-one;

4-propyl- 1 - {[2-(trifluoromethyl)-5,6,7,8-tetrahydroimidazo[ 1 ,2-b]pyridazin-3- yl]methyl}pyrrolidin-2-one; 1 -[(6-methylimidazo[2, 1 -b] [ 1 ,3.4]thiadiazol-5-yl)methyl]-4-propylpyrrolidin-2- one;

1 - { [6-(4-methylphenyl)imidazo[2, 1 -b] [ 1 ,3 ,4]thiadiazol-5-yl]methyl } -4- propylpyrro3.idm-2-one;

l -[(2-cyclopropyl-6-phenylimidazo[2,l-b][l ,3,4]thiadiazol~5~yi)methyl]-4~ propylpyrrolidin-2-one;

1 -[(6-methylimidazo[2, 1 -b] [ 1 ,3]thiazol-5-yl)methyl]-4-propylp>Trolidin-2-one; l-[(6-chloroira azo[2,l -b][l,3]thiazol-5-yl)methyl]-4-propylpyrrolidin-2-one; l-[(2,6-dichloroimidazo[2,l-b][l ,3]thiazol-5-yl)methyl]-4-propylpyrrolidin-2-one; l-(3H-imidazo[4,5-b]pyridin-7-ylmethyl)-4-propylpyrrolidin-2 -one;

l-(3H-imidazo[4,5-b]pyridin-7-ylmethyl)-4-phenylpyrrolidi n-2-one;

4-phenyl-l-[(5-phenyl-3H-imidazo[4,5-b]p>Tidin-7-yl)methy l]pyrrolidin-2-one;

4-phenyl-l-{[5-(trifluororafithyl)-3H-irnidazo[4,5-b]pyri dm-7- yljmethyl } pyrrol idin-2-one; l-[(6-bromo-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpy rrolidin-2-one; l-[(2-pheiiyl-3H-imidazo[4,5-b]pyridiii-7-yl)methyl]-4-pro^ l-[(5-methyl-3H-imidazo[4,5-b]pyridin-7-y3)rafithyl]-4-propy lpyTTolidin-2-one; l-[(2-methyi-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylp yrrolidin-2-one;

4-propyl- l-{[5-(trifluoroniethyl)-3H-imidazo[4,5-b]pyridin-7- yl]methyl}pyrrolidin-2-one;

l -[(6-methyl-3H-imidazo[4,5-b]pyridin-7-yl)me1hyl]-4-propylpy rrolidm-2-one; l -[(6-phenyl-3H-imidazo[4,5-b]pyridin-7-yl)methyl]-4-propylpy rrolidin-2-one; l-[l-(lH-imidazol-4-yl)propyl]pyrrolidin-2-one;

1 - [(5 -methyl- 1 H-imidazol-4-yl)methyl]pyrrolidm-2-one;

1 -[(2 -methyl- lH-imidazol-4-yl)methyl]pyrrolidin-2-one;

l-(l H-imidazoi-4-y3methyl)-4~propy3pyrrolidin-2-one; 1 -( { 1 - [(2-oxo-4-propylpyrrolidin- 1 -yl)methyl] - 1 H-imidazol-4-yl} methyl)-4- propylpyrrolidin-2-one;

l-[(5-chloro- lH-iniidazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrro[idi n-2-one; l-[(5-bromo-lH-imidazol-4-yl)methyl]-4-(2,3,5-trifluoropheny l)pyrroli

1 -[(5 -bromo- 1 H-imidazol-4-y l)methyl] -5 -chloro- 1 ,3 -dihydro-2H-indol-2-one; l ~(lH-irmdazol-5-ylmethy{)pyrrolidin-2-one;

1 - [( 1 -methyl- 1 H-imidazo 1-5 -yl)methyl]pyrro 1 idin-2-one;

l-methyl-5-[(2-oxopyrrolidin-l-yl)methyl]-lH-inTidazole-4 -carbonitrile;

l-(lH-iniidazol-5-ylmethyl)-4-phenylpyrrolidin-2-one;

1 -[( 1 -methyl- 1 H-imidazol-5 -yl)methyl] -4-phenylpyrrolidin-2~one;

1 -[(4-metboxy- 1 -methyl- 1 H-imidazol-5-yl)methyl]pyrrolidin-2-one;

l-[(l -methyi-l H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one;

l-methyl-5-[(2-oxo-4-propylp Trolidin-l-yl)methyl]-lH-imidazole-4-carbonitri

1 -methyl- 5-[(2-oxo-4-propylpyrrolidin- 1 -yl)methyl] - 1 H-imidazole-4 -carboxamide;

N-benzyl-2- {5 - [(2-oxo-4-propylpyrrolidin- 1 -yl)methy 1] - 1 H-imidazol- 1 - yljacetamide;

1 -methyl-5-[(2-oxo-4-propylpyrro[idin- 1 -yl)methyl]- 1 H-imidazole-2-carbonitrile; l-[(4-chloro-lH-imidazol-5-yl)methyl]-4-propylp> r rrolidin-2-one;

1 -m.ethyl-5 - { [2-oxo-4-(2,3 ,5 -trill uorophenyl)pyrrolidin- 1 -vijmethy 1 } - 1 H- imidazole-4-carbonitrile;

1 - [(4-bromo- 1 -methyl- 1 H-imidazol-5 -yl)methyl] -4-propylpyrrolidin-2-one;

l-[(2.4-dichloro-l-methyl-lH-imidazol-5-yl)methyl]-4-prop ylpyrrolidin-2-one; benzyl l-methyl-5-[(2-oxo-4-propyipyrrolidin-l-yl)niethyl]-l.H-irni dazol-2- ylcarbamate;

1 - [(4-chloro- 1 -methyl- 1 H-imidazol-5 -yl)methyl] -4-propy lpyrrolidin-2-one;

l-[(2-chloro-l -meth 3-l H-imidazol-5-yl)methyl]-4-propylpyrrolidin-2-one; 5 -chloro- 1 -( 1 H-imidazol- 5 -ylmethyl)- 1 , 3 -dihydro-2H-indol-2-one;

l-[(2,4-dichloro-l H-imidazol-5-yl)methyl]-4-(2,3,5-triiluoropheny3)pyrrolidin- 2- one;

l-[(2,4Hlicliloro-l-methyl H midazol-5-yl)me l]-4~(2,3,5- trifiuorophenyl)pyrrolidm-2-one;

l -[(2-chloro-l-methyl-lH-imidazol-5-yl)methyi]-4-(2,3,5- trifluorophenyl)pyrrolidin-2-one;

l-[(4-bromo-l-rn.ethyl-l H-imidazol-5-yl)methyl]-4-(2,3,5- trifluorophenyl)pyrrolidin-2-one;

5-ch[oro-l-[(l-memyl-lH-irnidazol-5-y[)m

1 -[(4-chloro- 1 -methyl- lH-imidazol-5-yl)methyl]-4-(2,3,5- trifiuorophenyl)pyrrolidin-2-one;

1 -( 1 H-indol-2-ylmethy [)-4-propylp>Trolidin-2-one ;

l-(lH-indol-3-ylmethyl)-4-propylpyrrolidin-2-one;

3-[(2-oxo-4-propylpyrrolidin- 1 -yl)methyl]- lH-indole-5-carbonitrile;

1 -[(2 -methyl- lH-indol-3-yl)methyl]-4-propylpyrrolidm-2-one;

l-[(7-methoxy-l H-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;

l-[(6-nitro-lH-indol-3-yl)methyl]-4-propylpyrrolidin-2-on e;

4-propy 1- 1 - { [6-(trifluoromethyl)- 1 H-iiidol-3 -yl]methy 1 } pyrrolidin-2-one; l-[(5-mtro-lH-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;

l -[(7-fluoro-l H-indol-3-yl)m.ethyl]-4-propylpyrrolidin-2-one;

l -[(5-chloro-2-methyl-lH-indol-3-yl)methyl]-4-propylpyrrolidi n-2-one;

l-[lH-indol-3-yl(phenyl)methyl]-4-propylpyrro[idin-2-one; l-[l-(lH-indol-3-yl)propyl]-4-propy[pyrrolidin-2-one;

l-[2-mi l(lH-indol-3-yl)methyl]-4-propylpyrrolidin-2-one;

3-[(2-oxo-4-propylpyrrolidin-l-yl)(phenyl)methyl]-l H-indole-5-carbonitrile; l-(lH-indol-4-ylmethyl)-4-propylpyrrolidin-2-one;

l-(l H-indol-7-ylmethyl)-4-propylpyrrolidin-2-one;

l-(isoxazol-4-ylmethyl)-4-propylpyrrolidin-2-one;

1-[(1 -phenyl- lH-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrro[idin- 2-one; 1 -[( 1 -methyl- 1 H-pyrazol-4-yl)methyl]-4-(2,3 ,5-trifluoropheny i)pyrrolidin-2-one; l-[(l-benzyl-lH-pyrazol-4-yl)methyl]-4-(2,3.5-trifluoropheny l)pyrrolidin-2-one;

4-(2,3,5-trifluorophenyl)-l -[(l,3,5-1x ^

one;

4- phenyl- 1 -( 1 H-pyrazol-4-ylmethyl)pyTTolidin-2-one; l-({l-[(4-m(^ylphenyl)sulfonyl]-lH-pyrazol-4-yl}methyl)-4-(2 ,3,5- trifluorophenyl)pyrrolidin-2-one;

l-(lH-pyrazol-4-y[methyl)-4-(2,3,5-trifluorophenyl)pyrrol idin-2-one;

1 -[(5 -chloro- 1 ,3 -dimethyl- lH-pyrazol-4 -yl)methyl]-4-(2,3 , 5 - trifluorophenyl)pyrrolidin-2-one;

1 -[(1 -chloro- l H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyl)pyrrolidin-2 -one; l-[(3,5-dimethy[-lH-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorop heny[)pyrrolidin-2- one;

l-[(3-methyi-l H-pyrazol-4-yl)methyl]-4-(2,3,5-trifluorophenyi)pyrrolidin-2 -one; l-[(5-amino-l ,3-dimethyl-lH-pyrazo[-4-yr)methyl]-4-(2,3,5- trifluorophenyl)pyrrolidin-2-one;

1 -[(5-amino- 1 -methyl- 1 H-pyrazol-4-yl)methyl]-4-propylpyrrolidin-2-one;

(-)- 1 -( 1 H-pyrazo 1 -4-y lmethyl)-4-(2,3 ,5 -tri fi uorophenyl)pyrrolidin-2 -one;

(+)-l-(lH-pyrazol-4-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrro lidin-2-one;

1 -( 1 H-pyrazol-4-ylmethyl)- 1 ,3-dihydro-2H-indol-2-one;

5 -chloro- 1 -( 1 H-pyrazol-4-y lmethyl)- 1 , 3 -dihydro-2H-indol-2-one;

5- chloro- 1 -( { 1 -[(4-methylphenyl)sulfonyl]- 1 H-pyrazol-4-yl}methyl)- 1 ,3-dihydro- 2H-indol-2-one; l-{[5-chloro-l-methyl-3-(trifluoromethyl)-lH-p>Tazol-4-yl ]methyl}-4- propylpyrrolidin-2-one;

l-[(5-amino-lH-pyrazol-4-y[)methyl]-4-(2,3,5-trifluorophe nyl)pyrrolidin-2-one; l-[(l-benzyl-5-chloro-lH-pyrazol-4-yl)methyl]-4-propylpyrrol idin-2-one;

l-[(1 -dimethyl-lH~pyrazol-5-yl)methyl]-4-(23,5- one;

l-(lH-pyrazol-5-ylmethyl)-4-(2,3,5-trifluorophenyl)pyrro[ idin-2-one;

l-[(4-bromo-l-methyl-lH-pyrazol-5-yl)methyl]-4-(2,3,5- trifluoroph eny l)pyrrol i din -2-one ;

l-[(l -methyi-l H-pyrazol-5-yl)methyl]-4-(2,3,5-trifluorophenyi)pyrrolidin-2 -one; l-[(6-bromo-2-methylpyrazolo[l ,5-a]pyrimi&^

one;

l -[(2-methylpyrazolo[l ,5-a]pyriniidin-3-yi)methyl]-4-propylpyrrolidin-2-one;

1 - [(6-bromo-2-phenylpyrazolo [ 1 ,5 -a]pyrimidin-3 -yl)methyl] -4-propylpyrrolidin- one;

l-[(6-bromo-2-tbien-2-ylpyrazolo[l,5-a]pyrimidin-3-yl)met hyl]-4- propylpyrrolidin-2-οηε;

4-propyl- 1 -[(2-thien-2-ylpyrazolo[ 1 ,5-a]pyrimidin-3-yl)methyl]pyrrolidiii-2-one;

1 -[(6-bromo-2-cyclopropylpyrazolo[ 1 ,5-a]pyrimidin-3-yl)methyl]-4~

propy Ipyrro 1 i din -2-one ;

1 -[(6-bromo-2-tert-buty[pyrazolo[ 1 ,5-a]pyrimidin-3-yl)methyl]-4- propylpyrrolidin-2-one;

l-[(2-phenylpyrazolo[] ,5-a]pyrimidin-3-yl)niethyl]-4-propylpyrrolidin-2-one; l-[(2-tert-butyi-6-cyclopropylpyrazolo[l,5-a]pyrimidin-3-yl) methyl]-4- propylpyrrolidin-2-οηε;

l -{[2-(2-fuiyl)pyrazolo[i ,5-ajpyriraidm-3-yl]methyl}-4-propylpyrrolidin-2-one; l-[(2-methyl-6-thien-2-ylpyrazolo[l,5-a]pyrimidin-3-yl)methy l]-4- propylpyrrolidin-2-one;

1- [(2-methy[-6-phenylpyrazolo[l,5-a]pyrinu^in-3-yl)methyl]-4-p ropylpyrrolidin-

2 - one;

l -{[2-methyl-6-(lH-pyrrol-2-yl)pyrazolo[l,5-a]pyrimidin-3-yl] methyl}-4- propylpyrrolidin-2-one;

1 -( {6- [( 1 E)-hex- 1 -enyl]-2-methylpyrazolo [ 1 , 5 -a]pyrimidin-3 -yl } methyl)-4- propylpyrrolidin-2-one;

1- [(6-chloro-2-phenyipyrazolo[l,5-a]pyrimidin-3-yl)methyl]-4-p ropylpyrrolidin-2- one;

1 ~ { [2-methyl-6-(phenylethynyl)pyrazolo[ 1 ,5-ajpyrimidin-3-yl]m.ethyl} -4- propylpyrrolidin-2-one;

1 -[(6-bromo-2-phenylpyrazolo[ 1 ,5-a]pyrimidin-3-yl)methyl]-4-(2 5 2- di fluorovi ny ! )pyrroli di n-2-one ;

1 -[(6-hydroxy-2-methy lpyrazolo[ 1 ,5-a]pyriiTiidin-3-yl)methyi]-4-propylpyrroiidm-

2- one;

1 - [(6-methyl-2-phenylpyrazolo[l ,5-a]pyrimidin-3-y])rnethyl]~4-propylpyn , olidin- - one:

4-(2,2-difluorovinyl)- 1 -[(2-phenylpyrazolo[ 1 ,5-a]pyrimidin-3- yl)methyl]pyrrolidin-2-one;

1- [(6-methoxy-2-phenylpyrazolo[l,5-a]pyrimidin-3-yl)methyl]-4- propylpyrrolidin-

2- one;

l -[(5-chloropyrazolo[l,5-a]pyrimidm-3-yl)methyl]-4-propylpyrr oHdin-2-one;

4-(2,2-difluoro vinyl)- 1 -[(5 ,6-dimethyl-2-phenylpyrazolo[ 1.,5-a]pyrimidin-3- yl)methyl]pyrrolidin-2-one;

4-(2,2-difluorovinyl)-l-[(6-fluoro-5^

yi)methyl]pyrrolidin-2-one;

1 - [(5 -methoxypyrazolo [ 1 ,5 -a]pyrimidin-3 -yl)methyl] -4-propy [pyrrolidin-2-one; 1 - {[2-(4-bromophenyl)pyrazolo[ 1 ,5-a]pyrimidin-3-yl]methyl} -4-(2,2- di fluorovi ny ! )pyrroli di n-2-one ;

1 - { [2-(4-fluoropheny l)pyrazo io [ 1 , 5 -a]pyrimidin-3 -yljmethy 1 } -4-propylpyrro lidin 2 -one; 4-(2,2-difluorovmyl)~ 1 -[(6-methyl-2-phenylpyrazolo[ 1 ,5~a]pyrimidm-3~ yl)methyl]pyrrolidin-2-one;

4-(2,2-difluorovinyl)- 1 -[(5-methyl-2-phenylpyrazolo[ 1 ,5-a]pyrimidin-3- yi)methyl]pyrrolidin-2-one;

4-( 2 ,2-di ft uoroviny 1 )- 1 - [(2-thien-2-ylpyrazolo [1 ,5 -ajpyrimi din-3 - yl)methyl]pyrrolidin-2-one;

l -{[2-(4-chlorophenyl)-6-methylpyrazolo[l,5-a]pyrimidin-3-yl] methyl}-4- propylpyrrolidin-2-one;

1 - { [2-(4-c-hlorophenyl)pyra.zolo[l ,5-a]pyriniidin-3-yl]methyl} -4-(2,2- di fl uorovi ny 1 )pyrroli di n-2-one ;

l-[(6-chloro-2-phenyipyrazolo[l,5-a]pyrimidin-3-yl)methyl ]-4-(2,2- difluorovinyl)pyrrolidin-2-one:

l -{[6-chloro-2-(4-chlorophenyl)pyrazolo[l ,5-ajpyrimidin-3-yl]m.ethyl}-4-(2,2- difluorovinyl)pyrrolidin-2-one;

1 - [(2-cyclopropyl-5 -methy Ipyrazolo [ 1 ,5 -a]pyrimidin-3 -yljmethy 1] -4-(2 ,2- di fluoro v i ny 1 )pyrroli din-2 -one ;

l-[(5-chloro-2-cyclopropylpyrazolo[l,5-a]pyrimidin-3-yl)r aethyl]-4-(2,2- difluorovinyl)pyrrolidin-2-one:

l -[(5-chloro-2,6-dimethylpyrazolo[l,5-a]pyrimidin-3-yl)methyl j-4-(2,2- difluorovinyl)pyrrolidin-2-one;

l-[(5-bromo-lH-pyrazo[o[3,4-b]pyridin-3-yl)methy[]-4-(2,2 - di fl uoro v i ny 1 )pyrroli din-2 -one ;

4-propyl- l.-(pyridin-3-ylniethyl)pyrrolidin-2-one;

( -)- 1 -( 1 -pyridin-3 -ylpropyl)pyrrol idin-2-one; 5-chloro-l-[(2-fluoropyridin-3-yl)methyl]-l,3-dihydro-2H-ind ol-2-one;

l-[(6-chloropyridm-3~y{)methy]]-4-propy{pyrrolidm~2-one;

l- {[6-(benzylamino)pyridin-3-yl]niethyl}-4-propylpyrrolidin-2- one;

l-[(2-aminopyridin-3-yl)methyl]-4-propylpyrrolidin-2-one;

4-propyl- l-(lH-pyrro[o[2,3-b]pyridin-3-ylmethyl)pyrrolidin-2-one;

l-[(2-isopropyl-lH-p}Trolo[2 -b]pyridin-3-yl)methyl]-4-propylpyrrolidin-2-one; 1 -[(2-pheny 1 - 1 H-pyrrol o [2 ,3 -b]pyridin-3 -yl)methy 1 ] -4-propylpyrrol idin-2-one; 4-propy 1 - 1 - [(2-propy 1 - 1 H-pyrro ! o [2 ,3 -b ]pyridm-3 -yl)methy I jpyrroli din-2-one;

1 - [(6-bromo-l H-pyrrolo[2,3-b]pyridin-3-yi)rnethyl]-4-propylpyrrolidin-2-o ne; l-[(l-benzoy[-6-bromo-lH-pyrrolo[2,3-b]pyridin-3-yl)niethyl] -4-propylpyrroli

2- orie;

l-[(6-phenyl-^-pyrrolo[2,3-b]pyridin-3-yl)methyl]-4-propy lpyiTolidin-2-one; l-[(5-bromo-lH-pyrro[o[2,3-b]pyridin-3-yl)methy[]-4-(2,2- difl uoro vinyl)pyrrolidm-2~one; 1 -[(7-oxido- 1 H-pyrrolo[2,3-b]pyridm-3-yl)methylj-4-propylpyrrolidin-2-one ;

4-propyl-l-(l H-pyrrolo[2,3-b]pyridin-4-yirnethyl)pyrrolidin-2-one;

4-propyl-l-(lH-pyrrolo[2,3-b]pyridm-5-y[methyl)pyrrolidin-2- one;

4-propyl-l-(lH-pyn-olo[2,3-c]pyridin-2-ylmethyl)p} r rrolidin-2-one;

4-propyl-l-(lH-pyrrolo[23-c]pyridin-3-ylmethyl)pyrrolidin-2- one;

4-propyl-l-(lH-pyrrolo[3,2-bjpyridin-3-ylmethyl)pyrrolidm -2-one;

4-propyl-l-(lH-pyrroio[3,2-c]pyridin-2-ylmethyl)pyrrolidin-2 -one;

4-propyl- l-(lH-pyrro[o[3,2-c]pyridin-3-ylniethyl)pyrrolidin-2-one;

4-propy 1- 1 -( 1 ,3 ,4-thiadiazol-2-ylmethyl)pyrrolidin-2-one;

1 -[(2-amino- 1 ,3-thiazol-5-yl)methyljpyrrolidin-2-one;

1 ~(1 ,3-thiazol-5-ylmethyl)pyrrolidin-2-one;

1 -[(2-chloro-l -thiaaol-5-yl)me&^ 1- { [2-(dimethylamino)- 1 , 3 -thiazol-5 -yljmethyl } -4-(2 ,3 ,5 - trifluorophenyl)pyrrolidin-2-one;

1 - { [2-(methy[amino)- 1 ,3-thiazol-5-yl]methyl} -4-(2,3,5-trifluoropheny[)pyrrolidin-

2 - one;

1 -[(2-pyrrolidin- 1 -yl- 1 ,3-thiazol-5-yl)methyl]-4-(2,3,5-trif3.uorophenyl)pyrroHdin- -one:

5-{[2-oxo- -(23,5 rifluorophenyl)pyrc^^

4-phenyl-l-{[3-(tTifluoromethyl)[l,2,4]triazolo[4,3-b]pyrida zin-7- yi]methyl}pyrrolidin-2-one;

4-phenyl-l-[(3-pheny[[l,2,4]1riazolo[4,3-b]pyridazin-7-yl )methy[]pyrrolidin-2- one;

4-phenyl-l- {[3-(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyridazin-8- yl]methyl}pyiTolidin-2-one;

4-propyl-l-{[3-(triiluoromethyl)[l ,2,4]lTiazolo[4,3-b]pyridazin-8- yi]methyl}pyrrolidin-2-one;

4-phenyl-l -[(3-pheny[[l,2,4]1riazolo[4,3-b]pyridazin-8-yl)methy[]pyrro lidin-2- one;

l -[(6-chloro-3-phenyl[l ,2,4]triazoio[4,3-b]pyridazin-8-yl)methyl]-4- propy lpyrrolidin-2 -one ; 1 -[(6-chloro[ 1 ,2,4]triazolo[4,3-b]p>Tidazin-8-yl)methyl]-4-phenylpyrrol idin-2-one; l-{[6-chloro-3-(trifluoromethyl)[l,2,4]triazolo[4,3-b]pyrida zin-8-yl]methyl}-4- phenylpyrrolidin-2-one;

l-[(6-chloro-3-phenyl[l,2,4]lTiazolo[4,3-b]pyridazin-8-yl )methyl]-4- phenylpyrrolidin-2-one;

l -[(2-fiuoroindolizin-3-yl)methyl]-4-propylpyrrolidin-2-one;

1 -( 1 H- 1 ,2,3-benzotriazol- 1 -ylmethyl)-4-propylpyrrolidin-2-one;

1- [(6-bromo-2-chloro-lH-iniidazo[4,5-b]pyridin-l-yl)methyl]-4- propylpyiTolidin-

2- one; 1 - [(6-bromo-2 -phenyl- 1 H-imidazo [4,5 -bjpyridin- 1 -yl)methyl] -4-propylpyrrolidin 2-one;

1 -(3 H-imidazo [4, 5 -b]pyridin-3 -ylmethyl)-4-propylpyrrolidin-2-one;

l-[(6-bromo-3H-iniidazo[4,5-b]p>Tidin-3-yl)methyl]-4-p ropylpyrrolidin-2-one;

1- [(6-bromo-2-chloro-3H-imidazo[4,5-b]pyridin-3-yl)methyl]-4-p ropylpyiTolidin-

2- one;

1- [(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3-y[)niethyl]-4- propylpyrrolidin

2- one;

l-[(6-bromo-2-cyclopropyl-3H-imidazo[4,5-b]pyri.din-3-yl) methyl]-4-(2,2- difluorovinyl)pyrrolidin-2-one;

l-[(3-chloro-7H-imidazo[4,5-c]pyridazin-7-yl)methyl]-4-pr opylpyrrolM

1 -[(2-chloro- 1 H-indol- 1 -yl)methylj-4-propylpyrrolidin-2-one;

1 -[(5-methyl- 1 H-indo!- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1 -[(6-methyl- 1 H-indol- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1 -[(2 -phenyl- 1 H-indol- 1 -y[)methyl]-4-propy[pyrrolidin-2-one;

l-[(5-fluoro-lH-indol-l-yl)methyl]-4-propylpyrrolidin-2-o ne;

1 - [(5 -bromo- 1 H-indol- 1 -yl)methy 1 ] -4-propylpyrrol idin-2-one;

l-[(5-chloro-l.H-indol-l -yl)methyl]-4-propylpyrrolidin-2-one;

l-(2,3-dihydro-lH-indol-l -ylmethyl)-4-propylpyrrolidin-2-one;

1 -[(5-fluoro-2 -phenyl- 1 H-indol- 1 -y[)methyl]-4-propylpyrrolidin-2-one;

1 -[(2-oxo-4-propylpyrrolidin- 1 -yl)methyl]- 1 H-indole-2-carbonitrile;

l -[(2-bromo-l H-indol-l-y3)ra.ethyl]-4-propylpyTTolidin-2-one;

l -[(2,5-dichloro-l.H-indol-l -yl)methyl]-4-propylpyrrolidin-2-one;

l-[(6-amino-9H-purin-9-yl)methyl]-4-propy[pyrrolidin-2-on e;

4-propyl-l-(9H-purin-9-ylmethyl)pyrrolidm-2-one;

1 - { [6-(cyclopropylamino)-9H-purin-9-yl]methyl} -4-propylpyrrolidin-2-oiie; 1 - { [6-^enzylamino)-9H-purin-9-yl]methyl} -4-propylpyrrolidin-2-one;

4-propyl-l-{[6-(propylaraino)-9H-purin-9-yl]methyl}pyrrol idm-2-one;

1- ({6-[(cyclopropylmethyl)amino]-9H-p] rin-9-yl}methyl)-4-propylpyrrolidm-2- one;

4-propyl- 1 -[(6-pyrrolidin- 1 -yl-9H-purin-9-yl)methyl]p>Trolidin-2-one;

1 ~[(5 -bromo-3 -phenyl- 1 H-pyrazolo [3 ,4-b]pyridm- 1 ~yl)methy 1 ] -4-propylpyrrol idin- - one:

l-[(5-bromo-2H-pyrazolo[3,4-b]pyridin-2-yl)methyl]-4-prop ylpyrrolidin-2-one; l-[(5-bromo-3-phenyl-2H-pyrazolo[3.4-b]pyridm^

2-one;

1 -[(2-chloro- lH-pyrrolo[2,3-b]pyridin-l-yi^

4-propyl- 1 -( 1 H-pyrrolo[3 ,2-bJpyridin- 1 -ylmethyl)pyrrolidin-2-one;

l -(3,4-dihydroquino3.in-l(2Bl)-ylmethy3.)-4-propylpyrro3.idin -2-one;

l -(8H-isothiazolo[5,4-b]indoi-8-ylmethyl)-4-propylpyrrolidin- 2-one;

1 -(lH-1 ,2,4-triazol-l-ylmethyl)pyrroiidin-2-one;

l-[(2,5-dichloro- lH-pyrrol-l-yl)methyl]-4-propy[pyrrolidin-2-one;

1 -[(2-chloro- IH-pyrrol- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1- [(2-chloro-l.H-benzimidazol-l-y3.)rn.ethyl]-4-propy3.pyrroli din-2-one;

1 -[(2-chloro- l.H-benzimidazol-l-yl)niethyl]-4-phenyipyrrolidin-2-one;

2-chloro- 1 -[(2~oxo-4-propylpyrro3idin~ 1 -y I )m ethyl]- 1 H-benzimidazoie-5- carbonitrile;

2- chloro- 1 - [(2-oxo-4-propylpyrrolidin- 1 -yl)methyl]~ 1 H-benzimidazole-6- carbonitrile;

4-propyl- 1 -[(2,5 ,6-trichloro- 1 H-benzimidazol- 1 -yl)methyl]pyrro[idin-2-one;

1 -[(2-chloro-6-methoxy- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyrrolidiii-2-one; l-[(2-chloro-5-methoxy-lH-benzimidazol-l-yl)m.ethyl]-4-propy lpyrrolidin-2-one; 1 -[(2-chloro-6-nitro- 1 H-benziniidazoi- 1 -yl)methyl]-4-propylpyrrolidin-2-one; l-[(2-chloro-5-nitro-lH-benzimida ol-l-yl)ra.ethyl]-4-propylpyiTolidin-2-one;

1 -[(2-chloro-6-methyl- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1 -[(2-chloro- 1 H-benzimidazol- 1 -yl)methyl]-4-(2,2-difluorovinyl)pyrrolidin-2-one; 1 -[(6-bromo-2-chloro- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1 -[(5-bromo-2-chloro- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyirolidin-2-one; l -[(2-cbloro-6-fluoro-lH-benzimidazol-l-y3)mfithyl]-4-propylp yrrolidin-2-one; l -[(2-chloro-5-fluoro-lH-benzimidazol-l-yl)methyl]-4-propylpy rrolidin-2-one; l -[(2,6-dichloro-l.H-benzimidazol-l-yl)methyl]-4-propylpyrrol idin-2-one;

1 -[(2,5-dichloro- lH-benzimidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one;

1 - { [2-chloro-6-(trifluoromethyl)- 1 H-benzimidazol- 1 -yljmethyl} -4- propylpyrrolidin-2-one;

1 - { [2-chloro-5-(trifluoromethyl)- 1 H-benzimidazol- 1 -yljmethyl} -4- propylpyrrolidm-2-one;

1 -[(2-cbloro-l H-benzimidazol- 1 -yl)methyl]pyrrolidin-2-one;

1 -[(2-chloro-6-hydroxy- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyrrolidin-2-one; l-(pyridin-4-ylmethyl)pyrrolidin-2-one, and

1 -[(2-chloro-5-hydroxy- 1 H-benzimidazol- 1 -yl)methyl]-4-propylpyirolidm-2-one. viii) U.S. Patent 4,696,943

The present invention relates to the novel compound (S)-alpha-ethyl-2-oxo-l - pyrrolidineacetamide .

ix) U.S. Patent 4,696,942

The present invention relates to the novel compound, (R)-alpha~ethyl-2-oxo- 1 - pyrrolidineacetamide

x) U.S. Patent 5,334,720

According to this invention we provide novel compounds of the formula I,

wherein, Rl, R2, R3 and R4, which may be the same or different independently represent hydrogen, CI -6 alkyl, phenyl or phenyl substituted by one or more halogen, hydroxyl, nitro, amino, CI -6 alkyl or CI -C6 alkoxy groups;

R5 and R6 independently represent hydrogen, CI -C6 alkyl or C3 -C6 cycloalkvl , or R5 and R6 together with the nitrogen form a C4-6 N heterocyc!e;

m represents an integer from 1-2; and

n represents an integer from 1-3;

provided that,

two of the substituents Rl, R2, R3 and R4 independently represent phenyl or substituted phenyl and the other two independently represent hydrogen or CI -6 alkyl; or a pharmaceutically acceptable acid addition salt thereof,

Pharmaceutically acceptable acid addition salts of the compounds of formula I include salts of mineral acids, for example, hydrohalic acids, e.g. hydrochloric or hydrobromic; or organic acids, e.g. formic, acetic or lactic acids. The acid may be polybasic, for example sulphuric, fumaric, maleic or citric acid.

This invention also relates to all stereoisomeric forms and optical enantiomeric forms of the compounds of formula L

In the compounds of formula I: alkyl groups which Rl, R2, R3, R4, R5 and R6 may represent include methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl and s- butyl; cycloalkvl groups which R5 and R6 may represent include cyclopropyi, cyclobutyl, cyclopentyl and cyclohexyl; CI -6 alkoxy groups include methoxy. ethoxy and propoxy;

halogen groups include fluorine, chlorine, bromine or iodine;

We prefer compounds of formula I or a pharmaceutical ly acceptable acidaddition salt thereof, in which; Rl is hydrogen, phenyl or substituted phenyl, preferably phenyl;

R2 is hydrogen, phenyl or substituted phenyl, preferably phenyl;

R3 is hydrogen , phenyl or substituted phenyl, preferably hydrogen;

R4 is hydrogen, phenyl or substituted phenyl, preferably hydrogen;

R5 is hydrogen , Cl-3 alkyl or cyclopropyl , preferably hydrogen or methyl;

R6 is hydrogen , Cl-3 alkyl or cyclopropyl , preferably hydrogen or methyl:

m represents an integer from 1 -2 preferably 2;

n represents an integer from 1 -2, preferably 1.

We especially prefer compounds of formula I in which R l and R2 are both phenyl.

We especially prefer compounds of formula I in which one of R5 and R6 is hydrogen and the other is hydrogen or methyl

xi) International Patent Application Publication No. WO2005/054188

In one aspect the invention therefore provides a compound having the formula I or a pharmaceutically acceptable salt thereof,

wherein

RI is hydrogen, CI-20 alkyl, C3 23 cycloalkvl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro. amino, guanidine, amino derivative, alkylthio, aryithio, alkylsulfonyl, ar lsulfonyl, aikyisulfinyi, arylsulfinyi, aryl or heterocycle; R2 is hydrogen, CI 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl;

R3 is hydrogen, CI 20 alkyl, alkoxy, amino, halogen, hydroxy, ester, amido, nitro, cyano, carbamate, or aryl; or R2 and R3 can form, together with the imidazole ring the following 1 H- benzimidazole cycle

R4 is hydrogen, CI -20 alkyl, C2-12 alkenyl,C2-12 alkynyl, aryl, azido, alkoxycarbonylammo, arylsulfonyloxy or heterocycle; R4a is hydrogen or CI -20 alkyl; or R4 and R.4a can .form together a C3-8 cycloalkyl ; R.5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-l -pyrrolidine ring the following 1, 3- dihydro-2H-indol-2~one cycle

R6 is hydrogen or CI 20 alkyl ; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl ; R8 is hydrogen, halogen, nitro, cyano, CI 20 alkyl or alkoxy ; R9 is hydrogen, CI -20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylihio, alkylsuifonyl, arylsulfonyl, alkylsulfinyi or arylsulfinyl ;

RIO is hydrogen, CI 20 alkyl, halogen, hydroxy, alkoxy, aryloxy, ester, amido, cyano, nitro, amino, amino derivative, alkylthio, arylthio, alkylsuifonyl, arylsulfonyl, alkylsulfinyi or arylsulfinyl ; RI 1 is hydrogen, halogen, nitro, cyano, CI 20 alkyl or alkoxy ; R12 is hydrogen or halogen;

R13 is hydrogen, nitro, halogen, heterocycle, amino, aryl, CI -20 alkyl

unsubstituted or substituted by halogen, or alkoxy unsubstituted or substituted by halogen; R14 is hydrogen, CI -20 alkyl or halogen;

R15 is hydrogen, CI 20 alkyl or halogen;

with the proviso that R4 is different from hydrogen when

N represents a group of formula

The asterisk * indicates the point of attachment of the substituents.

In a preferred embodiment, the invention concerns a compound having the formula ], their tautomers, geometrical isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof

wherein Rl is hydrogen, CI -20 alkyl, C3-8 cycloalkyl, halogen, hydroxy, ester, aniido, cyano, nitro, amino, guanidine, a!ky!thio, aikylsulfonyl, aikyisu!fmyi, aryl or lieterocycle; R2 is hydrogen, CI 20 alkyl, halogen, cyano, ester, carbamate or amido; R3 is hydrogen, cyano, C 1 20 alkyl, halogen or ester; or R2 and R3 can form together with the imidazole ring the following 1H- benzimidazole cycle

hydrogen, CI 20 alkyl, C2 12 alkenyi or aryl; R4a is hydrogen;

R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo- 1 -pyrrolidine ring the following 1, 3-dihydro-2H-indol-2-one cycle

R6 is hydrogen or C 1 20 alkyl ; R7 is hydrogen; or R6 and R7 are linked together to form a C3-6 cycloalkyl ; R8 is hydrogen; R9 is hydrogen, C 1 -20 alkyl, halogen or alkoxy; RIO is hydrogen, CI 20 alkyl, halogen or cyano; Rl 1 is hydrogen; R12 is hydrogen or halogen; R13 is hydrogen, halogen, heterocycle or CI 20 alkyl ; R14 is hydrogen; R15 is hydrogen; with the proviso that R4 is different from hydrogen when

represents a group of formula

The term"alkyl", as used herein, represents saturated, monovalent hydrocarbon radicals having straight (unbranched) or branched or cyclic or combinations thereof and containing 1-20 carbon atoms, preferably 1-10 carbon atoms, more preferably 1-4 carbon atoms; most preterred alley! groups have 1 -3 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of halogen, hydroxy, cyano, azido, aryloxy, alkoxy, alkylthio, alkanoylamino, arylcarbonylamino, aminocarbonyl,

methyiaminocarbonyl, dirnethylaminocarbonyl or aryl. Usually alkyl groups, in the present case, are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1- ethylpropyl, n-heptyl, 2,4, 4-trimethylpentyl, ii-decyl, chloromethyl,

trifiuoromethyl, 2-bromo~2,2~difluoroethyi, 2,2, 2-trifluoroethy!, 3,3, 3- trifluoropropyl, hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl, (propiony!amino) methyl, (benzoylamino) methyl, (4-chlorophenoxy) methyl, benzyl, 2-phenylethyl or 2- (methylthio) ethyl, Preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, t-butyl, 1-ethylpropyl, 2,4, 4- trimethylpentyl, chloromethyl, trifluoromethyl, 2,2, 2-trifluoroethyl,

hydroxymethyl, cyanomethyl, azidomethyl, (acetylamino) methyl,

(propionylamino) methyl, (benzoylamino) methyl or 2- (methylthio) ethyl. More preferred alkyl groups are methyl, ethyl, n-propyl, i-propyl, n-butyl, azidomethyl or trifluoromethyl. Most preferred alkyl groups are methyl or n-propyl.

The term"cycloalkyl", as used herein, represents a monovalent group of 3 to 8 carbon atoms, usually 3-6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycioa!ky! groups are cyclopropyl and cyclohexyl. The terni"alkenyl"as used herein, represents straight, branched or cyclic unsaturated hydrocarbon radicals or combinations thereof having at least one carbon- carbon double bond, containing 2-12 carbon atoms, preferably usually 2-4 carbon atoms. Aikenyl groups are being optionally substituted with any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups. Usually an aikenyl group is ethenyl (vinyl) optionally substituted by 1 to 3 halogens. Preferred aikenyl group, in the present case, is 2,2- difluorovinyl.

The ierrn"alkynyi"as used herein, represents straight, branched or cyclic hydrocarbon radicals or combinations thereof containing at least one carbon- carbon triple bond, containing 2-12 carbon atoms, preferably 2-6 carbon atoms, and being optionally substituted by any suitable group, including but not limited to one or more moities selected from groups as described above for the alkyl groups, Preferably an alkynyl group is a halogenoalkynyl group (haloalkynyl group). Groups qualified by prefixes such as"s", "i","t"and the like (e, g."i-propyI","s- butyl") are branched derivatives.

The term"aryl"as used herein, is defined as phenyl optionally substituted by 1 to 4 substituents independently selected from halogen, cyaiio, alkoxy, alk lthio, CI 3 alkyl or azido, preferably halogen or azido. Usually aryl groups, in the present case are phenyl, 3-chlorophenyl, 3 -fluorophenyl, 4-chlorophenyl, 4-fluorophenyl, 3,4- difluorophenyl, 3, 5-difluorophenyl, 3-cbloro-4-fl.uorophenyl, 2,3, 4- trifluorophenyl, 2,4, 5-trifluorophenyl, 2,3, 5-trifluorophenyl, 3,4, 5- tri fluorophenyl, 3-azido-2,4- difluorophenyl or 3-azido-2,4, 6-trifluorophenyl. Preferably, aryl groups are phenyl, 3- chlorophenyl, 3-fluorophenyl, 4- chlorophenyl, 4-fluorophenyl, 3,4-difluorophenyl, 3,5- difluorophenyl, 3-c-hloro-4- fluorophenyl, 2,3, 4-trifluorophenyi, 2,4, 5-trifluorophenyl, 2,3, 5-trifluorophenyl, 3,4, 5 -trifluorophenyl or 3-azido-2, 4-difluorophenyl. Most preferred aryl groups are phenyl, 3-chlorophenyl, 3-fluorophenyl, 3, 5-difluorophenyl, 2,3, 4- trifluorophenyl, 2,4, 5-trifluorophenyl, 2,3, 5-trifluorophenyl, 3, 4, 5- trifluorophenyl or 3-azido-2,4~difluorophenyl, The terni"heierocycle", as used herein, is defined as including an aromatic or non aromatic cyeloaikyi moiety as defined above, having at least one O, S and/or atom interrupting the carbocyclic ring structure. Heterocyclic ring moities can be optionally substituted by aikyi groups or halogens and optionally, one of the carbon of the carbocyclic ring structure may be replaced by a carbonyl. Usually heterocycles are 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-thienyl, 3- thienyl, 2- tetrahydrofuranyl, IH-pyrrol-2-yI, 1 -methyl-lH-pyrrol-2-yl, lH-pyrazol- 2-yl, 1H- pyrazol-3-yl, 4-chloro-l-methyl-lH-pyrazol-3-yl, 5-chloro-l, 3-dimethyl- lH-pyrazol-4- yl, 1 , 2,3~thiadiazol-4-yi, 3, 5~dimethyl-4-isothiazyl, lH-imidazol-2- yl, 1 -methyl- 1H- imidazol-2-yl, 4-methyl-lH-imidazol-5-yl, or 2-methyl-l, 3- thiazol-4-yl. Preferred heterocycles are lH-imidazol-2-yi, 1 , 2,3-thiadiazol-4-yi, lH-pyrazol-3-yl, 2-furyl, 3- furyl, 2-thienyl, 1 -methyl- lH-pyrrol-2-yl, lH-pyrrol-2- yi.

The term"halogen", as used herein, includes an atom of chlorine, bromine, fluorine, iodine. Usually halogens are chlorine, bromine and fluorine, Preferred halogens are fluorine, bromine and chlorine.

The term" hydroxy", as used herein, represents a group of formula-OH.

The term"alkoxy", as used herein, represents a group of formula-ORa wherein Ra is an alkyl group, as defined above. Preferred aikoxy group is methoxy.

The ternf'aryloxy", as used herein, represents a group of formula-ORb wherein l b is an aryi group, as defined above. Preferred aryloxy group is phenoxy.

The ternY'ester", as used herein, represents a group of formula-COORC wherein Rc is an alkyl group or aryl group, as defined above. Preferred ester group is methoxy carbonyl. The term"amido", as used herein, represents a group of formula-CONH2.

The term" amino", as used herein, represents a group of formula-NH2.

The term"aminoderivative", as used herein, represents an alkylamino or an arylamino group, wherein the terms"alkyl"and"aryr'are defined as above.

The temfcyano", as used herein, represents a group of formula-CN, The term"nitro", as used herein, represents a group of formula-N02.

The temfazido", as used herein, represents a group of formula-N3.

The ternVguanidine", as used herein, represents a group of formula- NHC (=NH) NH2. The term"alkylthio", as used herein, represents a group of formula-SRd wherein Rd is an a!ky! group, as defined above. One aikyithio group is methyithio.

The term"alkylsulfonyl", as used herein, represents a group of formula- S (=0) 2Re wherein Re is an alkyl group, as defined above. One alkylsulfonyl group is methyisulfonyl. The term"alkylsulfiny[", as used herein, represents a group of formula-S (=0) Rf wherein Rf is an alkyl group, as defined above. One alkylsulfmyl group is methyisulfinyi.

The term"arylthio", as used herein, represents a group of formula-SRg wherein Rg is an aryl group, as defined above.

The temfary!sulfonyi", as used herein, represents a group of the formula- S (=0) 2Rh wherein Rh is an aryl group, as defined above.

The term"arylsulfinyl", as used herein, represents a group of the formula- S (~0) Ri wherein Ri is an aryl group, as defined above.

The term"carbamate"as used herein, represents a group of formula- N (H) C (O) ORl , wherein Ri is an alkyl or a aryl, as defined above. Usually carbamate groups are (propoxycarbonyl) amino or (benzyloaxycarbonyl) amino. One carbamate group is (benzyloaxycarbonyl) amino.

The term"alkanoylamino"as used herein, represents a group of the formula- NHC (=0) Rk wherein Rk is an alkyl group, as defined above.

The term"(arylcarbonyl) amino"as used herein, represents a group of the formula- NHC (=0) ffl wherein Rrn is an aryl group, as defined above. One (aryicarbonyl) amino is benzoylamino.

Usually, RI is hydrogen; CI lo alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methyithio, phenyl or 4-chlorophenoxy ; hydroxy ; C3-6 cycloalkyl ; halogen; ester; aniido; nitro ; cyano; amino ; phenyl; alkylthio;

alkylsulfony! ; alkylsulfinyl ; heterocycle unsubstituted or substituted by alkyl groups; or guanidine.

In some embodiments, RI is hydrogen; methyl; ethyl; i-propyl ; n-propyl ;

cyclopropyi ; n-butyl; i- butyl; t-butyl; 1-ethylpropyl ; 2,4, 4-trimethyipentyl; hydroxymethyl ; chloromethyl; trifluoromethyl ; 2,2, 2-trifluoroethyl ;

eyanomethyl; 2- (methylthio) ethyl; chloro; bromo; nitro ; cyano; amino;

aminocarbonyl; methoxycarbonyl ; methylthio; methylsulfinyl ; methyisulfonyl; phenyl; 2-furyl ; 3-furyl; IH-pyrrol-2-yI ; l-methyl-lH-pyrrol-2-yl ; 2~ thienyl; 1H- pyrazol-3-yl ; 1 , 2,3-thiadiazol-4-yl or lH-imidazol-2-yI. More preferably, RI is hydrogen; methyl; ethyl; i-propyl ; n-propyl ; n-butyl; methylthio; nitro ; cyano; amino; chloro or IH-pyrroI-2-yI. Most preferably, RI is hydrogen; methyl;

methylthio ; nitro; cyano; amino or chloro.

Usually, R2 is hydrogen; CI 4 alkyl unsubstituted or substituted by hydroxy, alkanoylamino or benzoylamino; halogen ; ester; cyano ; alkyl carbamate; [(N- methoxy- -methyl) amino] carbonyl. Preferably, R2 is hydrogen; methyl;

hydroxymethyl ; (acetylamino) methyl; (propionylamino) methyl; (benzoylamino) methyl; [(benzyloxy) carbonyl] amino ; chloro or cyano. In some embodiments, R2 is hydrogen; chloro or cyano.

Usually, R3 is hydrogen; C 1 4 alkyl unsubstituted or substituted by hydroxy; halogen; ester or cyano. In some embodiments, R3 is hydrogen; hydroxymethyl; chloro; cyano.

In some embodiments, R3 is hydrogen or cyano. In some embodiments R3 is hydrogen.

Usually, R4 is hydrogen; CI 4 alkyl tin substituted or substituted by halogens; C2 4 alkenyl substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens. Preferably, R4 is hydrogen; n-propyl ; 2,2-difluorovinyi ; phenyl; 3-chlorophenyl ; 3 -fluorophenyl ; 4-chlorophenyl; 4-fluorophenyl ; 3,5- difluorophenyl; 3,4-difluorophenyl ; 3-chloro-4-fluorophenyl ; 2,3, 4- trifluorophenyl ; 2,4, 5 -tri fluorophenyl ; 2,3, 5-trifluorophenyl ; 3,4, 5- trifluorophenyl ; 3~azido-2,4- diiluorophenyl or 3-azido-2,4, 6-trifluorophenyl. More preferably, R4 is hydrogen; n- propyl ; 2,2-difluorovinyl ; phenyl; 3- chlorophenyl; 3-fluorophenyS ; 4-chlorophenyS; 4- fluorophenyl ; 3, 5- difluorophenyl ; 3,4-difluorophenyl ; 3-chloro-4-fluorophenyl; 2,3, 4- tri fluorophenyl ; 2,4, 5 -tri fluorophenyl; 2,3, 5-trifluorophenyl; 3,4, 5- trifiuorophenyl or 3- azido-2,4-difluorophenyl. Most preferably, R4 is n-propyl ; 2,2-difluorovinyl ; phenyl; 3- chlorophenyl; 3 -fluorophenyl ; 3, -di fluorophenyl ; 2,3, 4-trifluorophenyl ; 2,4, 5- trifiuorophenyl ; 2,3, 5-trifluorophenyl ; 3,4, 5- tri fluorophenyl or 3~azido-2,4- difluorophenyl.

Usually, R4a is hydrogen, Usually, R5 is hydrogen.

Usually, R6 is hydrogen or C1-1~O alkyl unsubstituted or substituted by hydroxy or azido. Preferably, R6 is hydrogen or azidomethyl. More preferably R6 is hydrogen.

Usually R7 is hydrogen.

In other embodiments, R6 and R7 are linked to form a cyclopropyi.

In other embodiments, R2 and R3 can form together with the imidazole ring the following I H-benzimidaole cycle

Usually, R8 is hydrogen.

Usually, R9 is hydrogen; halogen ; 1-3 alkyl or alkoxy. In some embodiments, R9 is hydrogen; methyl; chloro or methoxy. In some embodiments R9 is hydrogen.

Usually, RIO is hydrogen; halogen; cyano; CI 3 alkyl unsubstituted or substituted by halogens; or alkoxy. In some embodiments, RIO is methyl; hydrogen;

trifluoromethyl ; fluoro; cyano or methoxy. In some embodiments RIO is hydrogen; trifluoromethyl ; fluoro or cyano.

Usually, RI 1 is hydrogen. In other embodiments, R4, R4a and R5 can form together with the 2- oxo-1- pyrrolidine ring the following 1, 3-dihydro-2H-indol-2-one cycle

Usually, R12 is hydrogen or halogen. In some embodiments R12 is hydrogen; chloro or fluoro. In some embodiments R12 is hydrogen.

Usually, R13 is hydrogen; CI 3 alkyl ; halogen or thiazolyl unsubstituted or substituted by alkyl groups, such as methylihiazolyl. In some embodiments R13 is hydrogen; chloro; bromo or methyl, in some embodiments R13 is chloro; bromo or methyl

Usually R14 is hydrogen.

Usually, I I 5 is hydrogen.

In a general embodiment of the invention, the compounds of formula I, or pharmaceutically acceptable salts thereof, are those wherein

RJ is selected from hydrogen; CI Io alkyl unsubstituted or substituted by halogen, hydroxy, cyano, methylthio, phenyl or 4-chlorophenoxy ; C3 6 cycloalkyl ;

hal ogen; ester; ami do; nitro; cy ano; amino; ph enyl; alkylthio ; alkylsulfon l ; alkylsulfmyl ; heterocycle unsubstituted or substituted by alkyl group; or guanidine; R2 is selected from hydrogen; C 1-4 alkyl unsubstituted or substituted by hydroxy, alkatioylamino or benzoylamino; halogen; ester; cyano; alkyl carbamate or [ (N-methoxy-N-methyl) amino] carbonyl.

R3 is selected from hydrogen; CI 4 alkyl unsubstituted or substituted by hydroxy ; halogen; ester or cyano; R4 is selected from hydrogen; CI 4 alkyl unsubstituted or substituted by halogens; C2 4 alkenyi substituted by halogens or phenyl group unsubstituted or substituted by azido or/and halogens;

R4a is hydrogen; R5 is hydrogen; R6 is selected from hydrogen or C 1-10 alkyl unsubstituted or substituted by hydroxy or azido; R7 is hydrogen; or R6 and R7 can be linked to form a cyclopropyl ; or R2 and R3 can form together with the imidazole ring the following 1H- benzimidazole cycle

R8 is hydrogen; R9 is selected from hydrogen; halogen; CI -3 alkyl ; alkoxy ; RIO is selected from hydrogen; halogen; cyano or Cil alkyl unsubstituted or substituted by halogens; or alkoxy ; R 1 is hydrogen; or 1 4, R4a and R5 can form together with the 2-oxo-l -pyrrolidine ring the following 1, 3-dihydro-2H-indol-2- one cycle

R12 is selected from hydrogen or halogen; R13 is selected from hydrogen; CI-3 alkyl ; halogen ; thiazolyi unsubstituted or substituted by alkyl groups, such as methylthiazoiyl; R14 is hydrogen; R15 is hydrogen; with the proviso that R4 is different from hydrogen when

represents a group of formula

In an embodiment of the invention, the compounds of formul a I, or

pharmaceutically acceptable salt thereof, are those wherein

RI is selected from hydrogen; methyl; ethyl; i-propyl ; n-propy ; cyclopropyl ; n- butyl; i-butyl; t-butyi; l-ethylpropyl ; 2,4, 4-trimethylpentyl; trifluoromethyl; 2,2, 2- irifluoroethyl; hydroxymethyl; chloromethyl; cyanomethyl ; 2~ (methylihio) ethyl; chloro; bromo; nitro; cyano ; amino; aminocarbonyl; methoxycarbonyi ; methylihio; methylsulfinyl; methyisulfonyl; phenyl; 2-furyl ; 3-furyl ; lH-pyrrol-2 yl ; 1 -methyl- 1H- pyrrol-2-yl ; 2-thienyi; lH-pyrazol-3-yl ; 1 , 2, 3-thiadiazol~4~yl ; or lH-imidazol-2-yl ; R2 is selected from hydrogen; methyl ; hydroxymethyl; (acetyl amino) methyl; (propionylamino) methyl ; (benzoylamino) methyl;

(benzyloxycarbonyl) amino; chloro; or cyano; R3 is selected from hydrogen; hydroxymethyl; chloro; cyano; or R2 and R3 can form together with the imidazole ring the following 1H- benzimidazole cycle

1 8 is hydrogen; R9 is selected from hydrogen; methyl; choro ; methoxy;

RIO is selected from methyl; hydrogen; trifluoromethyl; fluoro; cyano; or methoxy; R is hydrogen; R4 is selected from hydrogen; n-propyl ; 2,2- difluorovinyl ; phenyl; 3- chlorophenyl; 3-fluorophenyl; 4-chlorophenyl; 4- fiuoropherryl ; 3,5-dilIuorophenyl ; 3,4- difluoropheny!; 3-chloro-4-fluorophenyl ; 2,3, 4-trifluorophenyl; 2,4, 5 -tri fluorophenyl ; 2,3, 5 -tri fluorophenyl; 3,4, 5- trifluoropiienyl ; 3-azido-2,4-difluorophenyi ; or 3-azido- 2,4, 6-trifluorophenyi, R4a is hydrogen: R5 is hydrogen; or R4, R4a and R5 can form together vvith the 2- 0X0-1 -pyrrolidine ring the following 1 , 3-dihydro-2H-indol-2-one cycle

R12 is selected from hydrogen; chloro; fluoro; R13 is selected from hydrogen; chloro; bromo; methyl; R14 is hydrogen; R15 hydrogen; R6 is selected from hydrogen; azidomethyl; R7 is hydrogen; or R6 and R7 are linked to form a cyciopropyl ; with the proviso that R4 is different from hydrogen when

represents a group of formula

In one embodiment of the invention, the compounds of formula I, or

pharmaceutically acceptable salt thereof, are those wherein

RI is selected from hydrogen; methyl; ethyl; i -propyl ; n-propyl ; n-butyl;

methyithio; niiro; cyano ; amino; chloro ; or lH-pyrrol-2-yl ; R2 is selected from hydrogen; chloro; cyano; R3 is selected from hydrogen; cyano; or R2 and R3 can form together with the imidazole ring the following 1 H- benzimidazole cycle

R8 is hydrogen; R9 is hydrogen;

RIO is selected from hydrogen ; trifluoromethyl ; fluoro ; cyano;

RJ 1 is hydrogen; R4 is selected from hydrogen; n-prop l ; 2, 2-difluorovinyl ; phenyl; 3- chlorophenyl; 3 -fluorophenyl; 4-chiorophenyl; 4-fluorophenyl ; 3, 5- difiuorophenyl ; 3,4- difluorophenyl ; 3-chloro-4-fluorophenyl; 2,3, 4- tri fluorophenyl ; 2,4, 5 -tri fluorophenyl ; 2,3, 5 -tri fluorophenyl; 3,4, 5- trifluorophenyi; or 3-azido-2, 4-difluorophenyl ; R4a is hydrogen; R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-l. -pyrrolidine ring the following 1, 3-dihydro-2H-indol-2-one cycle

wherein R12 is hydrogen; R13 is selected from methyl; chloro; hromo; R14 is hydrogen; R15 hydrogen; R6 is hydrogen; R7 is hydrogen; with the proviso that R.4 is different from hydrogen when

Rl 1 represents a group of formula

In one embodiment of the invention, the compounds of formula I, or

pharmaceutically acceptable salt thereof!, are those wherein

RI is selected from hydrogen; methyl; methylthio; nitro ; cyano; amino; chloro; R2 is selected from hydrogen; chloro; cyano; R3 is hydrogen; R4 is selected from n- propyl ; 2, 2-difluorovinyl ; phenyl; 3-chlorophenyi; 3- fluorophenyl; 3,5- difluorophenyl ; 2,3, 4-trifluorophenyl ; 2,4, 5 -triiluorophenyl ; 2,3, 5- tri fluorophenyl; 3,4, 5~iriiluorophenyl; 3~azido-2,4-difi.uorophenyl ; R4a is hydrogen;

R5 is hydrogen; or R4, R4a and R5 can form together with the 2-oxo-l -pyrrolidine ring the following 1 , 3-dihydro-2H-indol-2-one cycle

R12 is hydrogen; R13 is selected from chloro; bromo; methyl ; R14 is hydrogen; R15 hydrogen; R6 is hydrogen; R7 is hydrogen,

In some embodiments, compounds are: l-(lIi-irnida.zoi-l-ylmethyl) pyrrolidin-2- one ; 1 - (1H- imidazol-l -ylmethyl)-4-phenylpyrrolidin-2-one ; 4- (3-azido-2, 4, 6- trifluorophenyi)-!- (lH-imidazol-1 -ylmethyl) pyrrolidin-2-one; 1- (IH-imidazol-1- yimethyl)-4- propylpyrrolidin-2-one; (-)-4-(3-azido-2,4-difluorophenyl)-l-(lH- imidazol-i- ylmethyl) pyrrolidin-2-one; (+)-4- (3-azido-2, 4-difluorophenyl)-l- (IH-imidazol-i- ylmethyl) pyrrolidin-2-one ; 1 -[(2-ethyl-lH-iniidazol-l -yl)methyi]- 4-propylpyrrolidin-2- one; 1 -[(2-isopropyl-lH-imidazol-l-yl) methyl] -4- propylpyrrolidin-2-one ; l-[(2-methyl- IH-imidazol-l-yl) methyl]-4- propylpyrrolidin-2-one ; 1 -[(2-phenyl-l H-imidazol-l- yl) methyl]-4- propylpyrrolidin-2-one ; 4-propyl- l-[(2-propyl-lH-imidazol-l- yl) methyl] pyrrolidin-2-one ; (+)- l -(lH-imidazol-l-ylmethyl)-4-propylpyrrolidm-2-one ; (- )~1 ~ (lH-imidazol-l-ylmethyl)-4-propylp>TTolidin-2-one ; 4- (2, 2-difluorovinyl)- l - (1H- imidazoi- 1 - lmethyl) pyrroli din-2-one ; 4-(3 -chlorophenyl)- 1 -( 1 H -imi dazol- 1 - yimethyl) pyrrolidin-2-one; 1 - { [2-(methylthio)-lH-imi dazol- 1 -yl] methyl} -4- propyipyrro] idin-2-orie; 1 - { [2-(methylsulfinyl)-lH~imidazol~ 1 -yl] methyl } -4- propyipyrrolidin-2-one ; 1 -[(2-tert-butyl-lH-imidazol-i-yi) methyS]-4- propylpyrrolidin-2- one; 1- [1- (IH-imidazol-l-yl) cyclopropyl] pyrrolidin-2-οηε ; 1- [ (2-methyl-l H-imidazol-l- yl) methyl]-4-phenylpyrrolidin-2-one ; 1- {[2- (methylsulfonyl)-lH-iniidazol-l-yl] methyl} -4- propylpyrrolidin-2-οηε; 1 -[(2-oxo- 4-propylpyrrolidin- l-yl)methyl]-l.H-iniidazole-2- carboxamide; 4-(4- fluorophenyl)-l-(lH-imidazol-l-ylmethyl)pyrrolidin-2-one ; 1- (1H- imidazol-1- yimethyl)-4-(3, 4, 5-trifluorophenyl.) pyrrolidm-2-one; 4- (3-fluorophenyl)~l- (1H- imidazo i - 1 -yimethyl) pyrrolidin-2-one; 4-(3 ,5 -difluoropheny [)- 1 -( 1 H-imidazo i- 1 - yimethyl) pyrrolidin-2-one; 4-(3,4-difluoropheriyl)-l -(lH-irmdazol- l -yimethyl) pyrrolidin- 2-one; 4-(3-chloro-4-fluorophenyl)- 1 -(1 H-imidazol- 1 -yimelthyi) pyrrolidin-2-one; 4- (4- chlorophenyl)- 1-(1 H-imidazol- 1 -yimelthyi) pyrrolidin-2- one; l -(lH-imidazol-l-ylmethyi)- 4- (2, 3, 4-trifluorophenyl) pyrroiidin-2-one; 1 - (lH-imidazol-l-ylmethyl)-4-(2, 3,5-trifluorophenyi) pyrrolidin-2-one ; 1-(1H- imidazol-l-ylmethyl)-4-(2, 4,5- trifluorophenyl) pyrrolidin-2-one; l.-{[2- (hydroxymethyl)-l H-imidazol- l-yl]methyl} -4- propylpyrrolidin-2-οηε ; methyl 1- [ (2-oxo-4-propylpyrrolidin- 1 -yl) methyl]-iH-imidazole- 2-carboxylate ; 1- [ (2- nitro-lH-imidazol- l-yl) methyll-4- (3, 4,5 -trifluorophenyl) pyrrolidin- 2-one; 1- {[2~oxo-4-(3, 4, 5-trifluorophenyl) pyrrolidm-l-yl] methyl}-lH-imidazole-2- carbonitrile; l-[(2-aniino-lH-imidazol- l-yl)methyl]-4-propylpyrroiidin-2-one ; 1- [ (2, 4- dichloro-IH-imidazol-l-yl) methyl]-4- (3, 4, 5-trifluorophenyl) pyrrolidin-2- one ; 1- [ (5- chloro-IH-imi dazol- 1 -yl) methyl]-4- (3, 4, 5-trifluorophenyl) pyrrolidin-2-one ; l-{[2-oxo-4- (3,4, 5-trifluorophenyl) pyrrolidin- 1-yl] methyl} - lH-imidazole-4-carbomtrile ; l -{ [2-oxo-4- (3,4, 5-trifluorophenyl) pyrrolidin-l-yl] methyl} -lH-imidazole-5-carbonitrile ; (+)-!- (1H- imidazol- 1 -ylmethyl)-4- phenylpyrrolidin-2-one : (-)- 1 -( 1 H-imidazol- 1 -ylmethyl)-4- phenylpyrrolidin-2- one ; 1- { [2-oxo-4- (2, 3, 5 -tri fluorophenyl) pyrrolidin-l-yl] methyl} -1 H- imidazole-5-carbonitrile ; (-)-l-{[2-oxo-4-(2, 3, 4-trifluorophenyl) pyrrolidin-I- yljmethyS}- lH-imidazole-5-carbonitrile ; (+)-l- {[2-oxo-4-(2, 3, 4-trifluorophenyl) pyrrolidin-1- yl] methyl}-lH-imidazole-5-carbonitrile ; (-)-l -{[2-oxo-4-(2, 3,4- trifluorophenyi) pyrrolidin- 1-yl] methyl}-lH-imidazole-4-carbonitrile ; (+)-l -{[2- oxo-4-(2, 3, 4-trifluorophenyl)- 1- pyrrolidinyl] methyl } -lH-imi dazole-4- carbonitrile ; (-)-l - { [2-oxo-4- (3, 4,5- triiluorophenyi) pyrrolidin- l-yl]methyl.}- l H-imidazole-4-carbonitrile; (+)-l- {[2-oxo-4- (3,4, 5-trifluorophenyl) pyrrolidin-l- yl] methyl}-lH-imidazole-4-carbomtrile ; (+)-l-{[2-oxo- 4- (2, 4, 5- trifluorophenyi) pyrrolidin-l -yl] methyl}-lH-irnidazole-4-carbonitriie ; (~)~1~ {[2- oxo-4- (2, 4,5-irifluorophenyl) pyrrolidin- 1-yl] methyl} -lH-iniidazole-4- carbonitrile ; (-)-l - {[2-oxo-4-(2, 3, 5-trifluorophenyl) pyrrolidin-1 -ylmethyi}-l H- imidazole-4-carbonitrile ; (-)- l-{[2-oxo-4-(3, 4, 5=trifluorophenyl) pyrrolidin- 1- yl] methylj-lH-irnidazole-S-carbonitrile ; l-{[2-oxo-4-(2, 3, 5-trifluorophenyl) pyrrolidin-l-yl]methyl}- lH-iriiidazole-5-carbonitrile ; l- {[2-oxo-4-(2, 3,5- tri fluorophenyl) pyrrolidin- methyl }-lH-imidazole-5-carbonitrile ; l-[(5-methyl-2- phenyl- lH-imidazol-l-yl)methyl]-4-propylpyrrolidin-2-one ; 1- [ (5- methyl-IH- imidazol-l-yl) methyl]-4-propylpyrrolidin-2-one ; 1- [(5 -phenyl- 1 H-imidazol- 1-yl) methyl] -4-propy lpyrrolidin-2-one ; 1 -[(2-ethyl-5-methy 1- 1 H-imidazol- 1 - yl)methyl]- 4-propylpyrrolidin-2-one; l-[(2,5-dimethyl-lH-imidazol-l-yl)methyl]- 4- propylpyrroiidin-2-one; 1- [ (2-chloro- ( H-imidazol- 1 -yl) methyll-4- (3, 4,5- trifluorophenyl) pyrrolidin-2-one; l-[2-azido-l-(lH-imidazol-l-yl) ethyl] -4- propylpyrrolidin-2-one ; 1 - [ (4-chloro-IH~imidazol~l-yl) methyll-4- (3, 4,5- triiluorophenyi) pyrrolidin-2-one; l-[(2-bromo-4,5-dichloro-lH-imidazol-l - yl)methyl]-4- propylpyrrolidin-2-one; 1- [(2~chloro~l H-imidazol- 1 -yl)methyl]-4- propylpyrrolidin-2- one; (+)-l-l [2-oxo-4- (3, 4, 5 -tri fluorophenyl) pyrrolidin- 1- yl]methyl} - 1 H-imidazole-5 - carbonitrile; 1 - { [5 -(hydroxymethyl)- 1 H-imidazol- 1 - yi]methyl}-4-propyipyrrolidin-2-one ; l- {[4-(hydroxymethyS)-lH-imidazol-i-yi] methyl} -4-propylpyrrolidin-2-one ; benzyl 1- [ (2- oxo-4-propylpyrrolidin-l-yl) methyi]-SH-imidazol-5-ylearbamate ; N-[(l -{[2-oxo-4-(3, 4,5-trifluorophenyi) pyrrolidin-l-yl] methyl} -lH-imidazol-5-yl) methyl] acetamide ; N- [(l-{[2- oxo-4- (3, 4, 5-trifluorophenyl) pyrrolidin-l-yl] methyl}-lH-imidazol-5- yl) methyl] benzamide; N-l (1 -1 [2-oxo-4- (3, 4, 5-trifluorophenyl) pyrroldin-1. -yl]methyl}- 1H- imida.zol-5-yl) methyl] propanamide ; 1- (IH-benzimidazol-l-ylmethyl)-4- propyipyrrolidin- 2-one; 1 -[(2 -methyl- 1 H-benzimidazoi-1 -yl)methyl]-4- propylpyrrolidin-2-one ; 4-propyl- l-[(2-propyl-lH-benzimidazol-l- yl)methyl]pyrrolidin-2-one ; 1 -[(2-isopropyl-lH- benzimidazol-l-yl) metbyl]-4- propylpyrrolidin-2-one ; 4-propyl- 1 - { [2-(trifluoromethyl)- 1 H-benzimidazo 1- 1 -y 1 ] methyl} pyrrolidin-2-one; l-{[2-(methylthio)~lH-benzimidazol~l- yl] methyl} -4- propylpyrrolidin-2-one ; l-[(2-amino-l H-benzimidazol-l-yl)methyl]-4- propylpyrrolidin-2-one ; 1 - { [2-(chloromethyl)- 1 H-benzimidazol- 1 -yl]melthyl} -4- propylpyrrolidin-2-one ; (l -[(2-oxo-4-propylpyrrolidin-l-yl) methyl]- 1 H- benzimidazol-2- yl} acetonitrile ; 1- [ (5-methoxy-lH-beiizimidazol- 1 -yl) methyl]- 4-propylpyrrolidin-2-one ; 1 -[(5-methyl-lH-benzimidazol-l -yl) methyl]-4- propylpyrrolidin-2-one ; 1- [ (5, 6-dimethyl- IH-benzimidazol-i-yl) methyl]-4- propyipyrrolidin-2-one ; l ~{[2-isopropyl~5~ (trifluoromethyl)~lH-benzimidazol~l~ yl] methyl} -4-propylpyrrolidin-2-one ; l-[(6-chloro- IH-benzimidazol-l-yl) methyl]-4-propyipyrrolidm-2~one ; l-[(2-oxo-4-propylpyrrolidin-l- yl) methyl]-2- propyl-lH-benzimidazole-5-carbonitrile ; l- {[2-ethyl-5-(trifiuoromethyl)- 1H- benzimidazol- 1 -yl] methyl} -4-propylpyrrolidin-2-one ; 4-propyl- 1 - { [2-(l H-pyrrol- 2-yl)- IH-benzimidazol-l -yl] methyl} pyrrolidin-2-one ; 1- [ (5-fluoro-2-propyl- 1 H-benzimidazol- 1-yl) methyl]-4-propylpyrrolidin-2-one ; l-{[6-methyl-2-(lH- pyrrol-2-yl)-l H- benzimidazol-l-yl] methyl} -4-propylpyrrolidin-2-one ; l-[(6- methoxy-2-propyl-lH- benzimidazol-l-yl) methyl]-4-propylpyrrolidin-2-one ; 2- butyl-1- [ (2-ΟΧΟ-4- propylpyrroiidin-l-yl) methyl]-l.H-benzim.idazoie-5- carbonitrile ; l-{[2-[2- (methylthio) ethyl]-5-(trifluoromethyl)-lH-benzimidazol-l- yl] methyl} -4-propylpyrrolidin- 2-one; l-[(5-fluoro-2-isobutyi-l H-benzimidazol-1- yl)methyl]-4-propylpyrrolidin-2-one ; l-{[5-fluoro-2-(2, 4, 4-trimethylpentyl)-l H- benzimidazol- 1 -yl] methyl} -4-propylpyrrolidin- 2-one; 2-cyclopropyl- 1 ~[(2-oxo-4- propylpyrrolidin-l -yl)methyl]-l H-benzimidazole-5- carbonitrile ; 1- [ (2-oxo-4- propylpyrrolidin-l-yl) methyl]-2- (lH-pyrazol-3-yl)-lH- benzimidazole-5- carbonitrile; 1.-[(2-cyclopropyl-5-fluoro-l .H-benzimidazol- 1-yl jmethyl]- 4- propylpyrrolidin-2-one ; l-[(5-fluoro-2-isopropyl-lH-benzimidazol-l-yl)methyl]- 4- propylpyrrolidin-2-one : l-{[2-(3-furyl)-6-methoxy-lH-benzimidazol-l- yimethyl } -4- propylpyrrolidm-2-one; 1 - [(2-cyclopropyi-6-m.ethoxy-iH- benzimidazol-l-yl) methyl]-4- propylpyrrolidin-2-one; 1- [(2-isopropyl-6- methoxy-lH-benzimidazol-l -yl) methyl]-4- propylpyrrolidin-2-one ; 1- [(2-oxo-4- propylpyrrolidm-l-yl) methyl]-2-(l , 2,3-thiadiazol-4- y[)-lH-benzimidazole-5- carbonitrile ; l~ {[2-(lH-irnidazol-2~yl)~5-(trifluoromethy])-l H- benzimidazol-l-yl] niethyl}-4-propyipyrrolidin-2-one ; l- {[5-fluoro-2-(2, 2,2- trifluoroethyl)-lH- benzimidazol-! -yl] methyl} -4-propylpyrrolidin-2-one ; 1- { [2- (1 - ethylpropyl)-6- methoxy-IH-benzimidazol-l-yi] methyi} -4-propylpyrrolidin-2-one ; 1 ~ {[6- methoxy-2- (l-methyl-lH-pyrrol-2-yl)-IH-benzimidazol-l-yl] methyl} -4- propylpyrrolidin- 2-one; 1 - {[2-(2-fuiyl)-5-(trifluoromethyl)-l H-benzimidazol-1 - yljmethyl} -4- propylpyrrolidin-2-one; 4-propyl- 1 - { [2-thien-2-yl-5- (trifluoromethyl)-lH-benziniidazol- l-yi]melthyl}pyrrolidin-2-one ; 1 -1 [2- (3- fury )-5 - (trifluoromethyl)-IH-benzimidazol-l- y 1] methyl } -4-propylpyrrolidin-2- one ; 1- { [2-cyclopropyl-5- (triiiuoromethyl)-lH- benzimidazol-l-yl] methyl} -4- propylpyrrolidin-2-one ; 4-propyl- 1 - { [2-( 1 H-pyrrol-2-yl)-5 - (trifluoromethy [)- 1 H- benzimidazol-l -yl] methyl} pyrrolidm-2-one ; 1- (IH-irnidazol-1 - ylmethyl)-l , 3- dihydro-2H-indol-2-one ; 5-bromo-l-(lH-imidazol-l-ylmethyl)- l , 3- dihydro-2H- indol-2-one; 5-chloro-l- (IH-imidazol-l-ylmethyl)-l , 3-dihydro-2H-indol-2~ one; 4-fluoro-l-(lH-imidazol-i-yimethyl)- 1 , 3-dihydro-2H-indoi-2-one; 4-chioro- 1 -(1 H- imidazol- 1 -ylmethyi)- 1 , 3 -dihydro-2H~indol-2-one ; 1 -(lH-imidazol-l-y lmethyl)-5 - methyl- 1 , 3-dihydro-2H-indol-2-one ; 1 - [ (2-oxo-2, 3-dihydro-lH-indol-l-y!) methyl] -1H- imidazole-5-carbonitrile; and 1- [ (5-chloro-2-oxo-2, 3-dihydro-lH- indol-l-yl) methyl]-! H~ irmdazole-5-carbonitrile. In some embodiments, compounds are: 1 - (lH-imidazol-1.-ylmethyi) pyrro!idin-2- one, 1- (lH-imidazol-l-ylmethyl)-4-phenylpyrrolidin-2-one ; l-(lH-imidazol-l- yimethyl)-4- propy!pyrrolidin-2-one; (-)-4- (3-azido-2, 4-difluorophenyl )-.l - (!H- imidazol- 1 - ylmethyi) pyrrolidin-2-one; (+)-4-(3 -azido-2,4-difluorophenyl)- 1 -( 1 H- imi.dazol-1- ylmethyi) pyrrolidin-2-one ; l ~[(2-ethyl~lH-i.rnidazol-l ~yi)methyl]-4- propylpyrrolidin-2- one; l-[(2-isopropyl- lH-imidazol-l -yl)methy[]-4- propylpyrrolidin-2-one ; 1- [ (2-methyl- I H-imidazol-! -ylj methyl]-4~

propylpyrrolidin-2-one ; 4-propyl- 1 -[(2 -propyl-1 H-imidazol- 1-yl) methyl] pyrrolidin-2-one : (lH-imidazol-l-ylmethyl)-4-propylpyrrolidin-2-one : (-)- l-(lH-imidazol-1 -ylmethyi)-4-propylpyrroiidin-2-one ; 4-(2, 2-difluorovinyl)-l -(l H- imidazol-l-ylmethyl) pyrrolidin-2-one: 4- (3-chlorophenyl)-l- (lH-imidazol-1- ylmethyl) pyrrolidin-2-one ; 1 -{[2-(niethylthio)-l H-imidazoi-l-yl]niethyl}-4- propyipyrrolidin-2-one ; l-[(2-methy[-lH-imidazol- l-yl)methyl]-4- phenylpyrro idin-2- one; 4-(4-fluorophenyl)-l-(l H-imidazol-l-y3.m.ethyl) pyrrolidin-2-one; l-(lH-imidazol-l- ylmethyl)-4- (3, 4, 5-trifluorophenyl) pyrrolidin-2-one; 4-(3-fluorophenyl)-l-(l H-imidazoi- 1 -ylmethyi) pyrrolidm-2- one; 4-(3,5-difluorophenyl)-l -(lH-imidazol-l - yimethyl) pyrrolidin-2-one ; 4-(3,4- difluorophenyl)-l-(lH-imidazol-l -ylmethyi) pyrrolidin- 2-one; 4-(3-chloro-4- fluorophenyI)-l -(lH-imidazol-l -ylmethyi) pyrrolidin-2-one; 4- (4- chlorophenyl)- l-(lH-imidazol-l -ylmethyi) pyrrolidin-2-one; 1- (lH-imidazol-1 -ylmethyi)- 4- (2, 3, 4-trifluorophenyl) pyrrolidin-2-one; l-(l H-imidazoi-l-ylmethyl)-4-(2, 3,5- trifluorophenyl) pyrrolidiii-2-one; 1- (1 H-imidazol- 1 -y lmethyl)-4- (2, 4,5- trifluorophenyi) pyrrolidin-2-one; l-[(2-nitro-lJi~imidazol~l-yl) methyl]-4-(3, 4,5- trifluorophenyi) pyrrolidin-2-one ; 1- { [2-oxo-4- (3, 4, 5 -trifluorophenyi) pyrrolidin- 1- yl] meth l} -lH-imidazole-2-carbonitrile ; l-[(2-amino-l H-imidazol- l-yl)methyl]-4- propylpyrrolidin-2-οηε; 1-1 (5-chloro-SH-imidazoH-yl) methyl] - 4- (3, 4,5-trifluorophenyl) pyrrolidin-2-one; l-{[2-oxo-4-(3, 4, 5 -trifluorophenyi) pyrrolidin-1- yl] methyl} -lH-imidazoie-4-carbonitrile ; l-{[2-oxo-4-(3, 4, 5- trifluoropiieiiyl) pyrrolidin-1- yl] methyl}-lH-imidazole-5-carbonitrile ; (H-)-l-(lH- imidazol-1 -ylmethyl)-4- phenylpyrro!idin-2-one ; (-)-l-(lH-imidazol-l-ylmethyl)-4- phenyipyrrolidin-2-oiie : (+); l-{[2-oxo-4-(3, 4,5-trifluorophenyl) pyrrolidin-1- yl]methyl} - 1 H-imidazole-4-carbonitrile ; 1 -[(2-chloro-l H-imidazol-l-yl) methyl]-4- (3, 4, 5-trifluorophenyl) pyrrolidin-2-οηε ; 1 - [2- azido-l-(lH-imidazol-l-yl) ethyl]- 4-propylpyrrolidin-2-one ; l-[(2-chloro-lH-imidazol-l- yl) methyl]-4- propylpyrrolidin-2-one ; (+)-l-l [2-oxo-4- (3, 4, 5-trifluorophenyl) pyrrolidin-1 - yi]methyl} - 1 H-imidazoie-5 -carbonitriie ; 1 - [(2-oxo-4-propylpyrrolidin- 1 -yl) methyl]-2- propyl-lH-benzimidazole-5-carbonitrile ; l -{[2-ethyl-5- (trif uoromethyl)-lH- benzimidazol-l-yi]metiiyl}-4-propyipyrrolidin-2-one ; 4- propyl-1- {[2-(l H-pyrrol-2-yl)-lH- benzimidazol-l-yi]methyl}pyrrolidin-2-one ; 1 - [(5-fluoro-2-propyl- 1 H-benzimidazol- 1 - yl) methyl]-4-propylpyrrolidin-2-one ; 2- butyl- 1- [(2-oxo-4-propylpyrrolidin-l-yl) methyl]- lH-berizimidazole-5-carbonitrile ; 1 - [ (5-fluoro-2-isopropyl-lH-benzimidazol- 1 - yl) methyl]-4-propylpyrrolidin-2~ one ; 1-(1 H-imidazol- 1 -ylmethyl)- 1. 3-dihydro-2H-indol- 2-one; 5-bromo-l- (1H- miidazol-1 -ylmethyl)- 1, 3-dihydro-2H-indol-2-one ; 5-chloro-l- (lH-imidazoi-1- ylmethyl)-!, 3-dihydro-2H-indol-2-one ; 1 -( 1 H-imidazol- 1 -ylmethy I)-5 - methyl- l,3-dihydro-2H-indol-2-one ; l-[(5-chloro-2-oxo-2 5 3-dihydro-lH-indol-l - yl) methyl] -lH-imidazole-5-carbonitrile.

In some embodiments, compounds are: 1-(1 H-imidazol- l-ylmethyl)-4- phenylpyrrolidin- 2-one; 1 -(IH-imidazol- 1 -ylmethyl)-4-propylpyrrolidin-2-one ; (- )_4_ (3-azido-2, 4- difluorophenyl)-l-(l H-imidazol- 1 -ylmethy 1} pyrrolidin-2-one; (+)-4- (3~azido-2, 4- dif3.uorophenyl)-l-(lH-imidazol-l -ylmethy 1) pyrrol idin-2-one; 4-(2,2-difluorovinyl)-l - (iH-imidazol-l -ylmethyl) pyrrolidin-2-one; 4-(3- chlorophenyl)- 1 ~( 1 H-imidazol- 1 - ylmethyl) pyrrolidm-2-one; 1 - { [2-(methylthio)- IH-imidazol-l-yi] methyl} -4- propylpyrrolidin-2-one; 1 -[(2 -methyl- lH-imidazol- l-yl)methyl]-4-phenylpyrrolidin-2- one: 1- (lH-imidazol-l-ylmethyl)-4- (3, 4,5- trifluorophenyl) pyrrolidin-2-one; 4- (3- fluorophenyl)-l-(l H-imidazol-l-ylmethyl) pyrrolidin-2-one; 4-(3 , 5-difluoromethyl)- 1 - (IH-imidazol- 1 -vlmetliyl) pyrrolidin-2- one; 1-(1 H-imidazol- l-ySmethyl)-4-(2, 3,4- tri fluorophenyl) pyrrolidin-2-otie; 1 - (lH-imidazol-l-ylmethyl)-4-(2, 3,5- trifluorophenyl) pyrrolidin-2-οηε; 1- H- imidazol-l-ylmethyl)-4-(2, 4,5- trifluorophenyl) pyrrolidin-2-one; l -[(2-nitro-lH- imidazoi-l-yl) methyl]-4-(3, 4,5- trifluorophenyl) pyrroiidin-2-one; l-{[2-oxo-4- (3, 4, 5-trifluorophenyl) pyrrolidm-1- yl] methyl}-lH-imidazole-2-carbonitrile ; 1 - [(2-amino-lH-imidazol-l-y[) methyl]-4- propylpyrrolidin-2-one ; l-[(5-chloro-lH- imidazol-I-yl)methyl]-4-(3, 4,5- trifluorophenyl) pyrrolidin-2-one; (+)-i-(lH- imidazoi-l-ylmethyi)-4-phenylpyrrolidin-2- one; (-)- 1 -(iH-imidazol-l-ylmethyl)-4- phenylpyrrolidin-2-one ; l-[(2-chloro-lH- imidazol-l-yl) methyl] -4- (3, 4,5- tri fluorophenyl) pyrrolidin-2-one l-[(2-chloro-l H-imidazol-i-yl) methyl]-4- propylpyrrolidin-2-one ; (+)-l-l [2-oxo-4- (3, 4,5- trifluorophenyl) pyrrolidin-l-yl] methyl} -lH-imidazole-5-carbonitrile ; 5-bromo-l- (1H- imidazoi- 1 -ylmethyl)- 1 , 3- dihydro-2H-indol-2-one; 5-chloro-l-(lH-imidazol-l-ylmethyl)- 1, 3-dihydro-2H- indol-2-one; 1- (lH-imidazol-l-ylm.ethyl)-5-methyl-l , 3~dihydro-2H- indol-2-one; 1 -[(5-chloro-2-oxo-2, 3-dihydro-lH-mdol-l-yl) methyl]-lH-imidazole-5- carbonitri!e.

Some compounds are: (-)-4- (3-azido-2, 4-difluorophenyl)-l- (lH-imidazol-1- ylmethyl) pyrrolidin-2-orie ; (+)-4-(3~azido-2, 4-difluorophenyl)- 1 -(IH-imidazol- 1 - ylmethyl) pyrrolidin-2-one; 4-(3-azido-2, 4-difluorophenyl)- l-(iH-imidazol-l - yimethyl) pyrrolidin-2-one.

The acid addition salt form of a compound of formula I that occurs in its tree form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or iiydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, maionic, succinic, maieic, fumaric, malic, tartaric, citric, m ethanes ulfonic, ethanesulfonic,

beiizenesulfonic, p-toiuenesulfonic, cyclic, salicylic, p-a.niiiiosalicylic, pamoic and the like.

The compounds of formula I containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e. g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e. g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e. g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.

Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.

Compounds of the formula I and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.

Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Cheni. , 45 (1976) 11-30. The invention also relates to ail stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).

Some of the compounds of formula I may also exist in tautomeric forms, Such forms although not expiicity indicated in the above formula are intended to be included within the scope of the present invention.

In another preferred embodiment, the present invention concerns also compounds of formula IA and their tautomeric form IB

With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically.

Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention.

The invention also includes within its scope pro-drug forms of the compounds of formula i and its various sub-scopes and sub-groups.

xii) U.S. Patent Application Publication No. 20090018148

In one aspect the invention provides compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,

wherein

Rl is hydrogen or CI -6 alkyl;

R2 is hydrogen or CI. -4 alkyl;

R3 is a group of formula CHR5R6 or a benzyl group;

R4 is C I -8 alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle;

R5 is C2-4 alkyl;

R6 is C2-4 alkyl, amido or—COOR7;

R7 is CI -4 alkyl;

In one aspect, the invention provides compounds:

When Rl is hydrogen, R2 is methyl, R3 is— CHR5R6, R.6 is ethoxycarbonyl and R5 is ethyl, then R4 is different from methyl, n-propyl, i-propyl, n-pentyl, n-heptyl, 3-bromobenzyl, 4-chiorobenzyl, 4-methylbenzyl or 2-phenylethyl;

When R l is hydrogen, R2 is methyl, R3 is benzyl, then R.4 is different from i- propyl, n-butyl, 3-methylbutyl, benzyl, phenylethyl-, or 3-phenylpropyl;

When Rl and R2 are methyl, R3 is benzyl, R4 is different from methyl, 3- methyibutyl, benzyl, 3-phenylpropyl or 4-chlorophenylmethyl;

Finally 8-(2-chioro ^enzylsulfaxTyl)-3-methyl-7-octyl-3,7-dihydro-purme is considered.

Usually when R3 is a benzyl group, then R4 is CI -8 alkyl optionally substituted by alkoxycarbonyl.

Usually when R3 is a group of formula— CHR5R6, then R4 is CI -8 alkyl optionally substituted by C3-6 cycloalkyl, aryl or heterocycle. The term "alkyl", as used herein, is a group which represents saturated,

monovalent hydrocarbon radicals having straight (unbranched) or branched moieties, or combinations thereof, and containing 1-8 carbon atoms, preferably 1-6 carbon atoms; more preferably alkyl groups have 1-4 carbon atoms. Alkyl moieties may optionally be substituted by 1 to 5 substituents independently selected from the group consisting of hydroxy, alkoxy, cyano, ethynyl, alkoxycarbonyl, acyl, aryl or heterocycle. Alkyl moieties may be optionally substituted by a cycloalkyl as defined hereafter, Preferred alkyl groups according to the present invention are methyl, cyano ethyl, ethyl, 2-ethoxy-2-oxoethyi, 2-methoxyethyl, n-propyl, 2- oxopropyl, 3-hydroxypropyl, 2-propynyi, n-butyl, i-butyi, n-pentyi, 3-pentyl, n- hexyi, cyciohexyimethyl, benzyl, 2-brom.obenzyl, 3-bromobenzyi 4-bromobenzyi,

3- methoxybenzyl, 3-nitrobenzyL 3-a.miiiobenzyl, 4-(aminosulfonyi)benzyl, 1- phenylethyl, 2-phenyiethyl, (3,5-dimethylisoxazol-4-yl)methyl or (5-nitro-2- furyl)methyl. More preferred alkyl groups are methyl, ethyl, cyanomethyl, 2- methoxyethyl, n-propyl, 3-hydroxypropyl, 2-propynyl, n-butyl, 3-pentyl, n-hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyL 3-aminobenzyl, (3,5- dimethylisoxazol-4~yl)methyl or (5~nitro-2~furyl)methyl. Most preferred alkyl groups are methyl, ethyl, 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2- furyl)methyl.

The term "cycloalkyl", as used herein, represents a monovalent group of 3 to 8, preferably 3 to 6 carbon atoms derived from a saturated cyclic hydrocarbon, which may be substituted by any suitable group including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferred cycloalkyl group according to the present invention is cyclohexyl.

The term "aryl" as used herein, is defined as a phenyl group optionally substituted by 1 to 4 substituents independently selected from halogen, amino, nitro, alkoxy or aminosuifonyl. Preferred aryl groups are phenyl, 2~hrom.oph.enyl, 3-bromoph.enyl,

4- bromophenyl, 3-methoxyphenyi 3-nitrophenyi, 3-aminophenyl or 4- (aminosuifonyl)piienyl.

The term "phenyl", as used herein, represents an aromatic hydrocarbon group of formula— C6H5. The term "benzyl group", as used herein, represents a group of formula— CH2- aryl. Preferred benzyl groups are benzyl, 2-bromobenzyl, 3-bromobenzyl, 4- bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl or 4- (aminosulfonyl)benzyl. More preferred benzyl groups are benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl or 3-aminobenzyl. In some embodiments alkyl groups are 3-methoxybenzyl or 3-nitrobenzyl.

The term "halogen", as used herein, represents an atom of fluorine, chlorine, bromine, or iodine. In some embodiments the halogen is bromine.

The term "hydroxy", as used herein, represents a group of formula— OH.

The term "cyano", as used herein, represents a group of formula— CN.

The term "amino", as used herein, represents a group of formula NH2.

The term "ethynyl", as used herein, represents a group of formula— C≡CH.

The term "alkoxy", as used herein, represents a group of formula— QRa wherein a is an alkyl group, as defined above. In some embodiments the alkoxy group is methoxy.

The term "nitro", as used herein, represents a group of formula— N02.

The term "amido", as used herein, represents a group of formula— C(==0)NH2.

The term "aeyl", as used herein, represents a group of formula C(=0)Rb wherein Rb is an alkyl group, as defined here above. In some embodiments the aeyl group is acetyl (— C(= )Me).

The term "alkoxycarbonyi (or ester )", as used herein, represents a group of formula — COORc wherein Rc is an alkyl group; with the proviso that Rc does not represent an alkyl alpha-substituted by hydroxy. In some embodiments the alkoxycarbonyi group is ethoxycarbonyl.

The term "heterocycle", as used herein, represents a 5-membered ring containing one or two heteroatoms selected from O or N. The heterocycle may be substituted by one or two Cl-4 alkyl or nitro. In some embodiments the heterocvcles are (3,5- dimethylisoxazol-4-yl) or (5-nitro-2-furyl). Most preferred heterocycle is (5-nitro- 2-furyl). Generally Rl is hydrogen or CI -6 alkyl. Usually Rl is hydrogen or CI -6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethynyi, alkoxycarbonyl or aeyl. In some embodiments Rl is hydrogen, methyl, cyaiiomethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n-propyS, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl. In some embodiments Rl is hydrogen, methyl, cyaiiomethyl, 2- methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl. In some embodiments Rl is hydrogen.

Generally R2 is hydrogen or CI -4 alkyl. Usually R2 is hydrogen or unsubstituted CI -4 alky], In some embodiments R2 is hydrogen, methyl or n-butyl. In some embodiments, R2 is methyl.

Generally R3 is a group of formula— CHR5R6 or a benzyl group. In some embodiments R3 is 3-pentyl, l-(arninocarbonyl)propyi, l-(ethoxycarbonyl)propyl or 3-bromobenzyl. In some embodiments R3 is I-(ethoxycarbonyl)propyl.

Generally R4 is CI -8 alkyl optionally substituted by alkoxycarbonyl, C3-6 cycloalkyl, aryl or heterocycle. Usually R4 is CI -8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyi, aminophenyl, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5-dimethylisoxazol-4-yl, 5-nitro-2-furyl or

ethoxycarbonyl. In some embodiments R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2-bromobenzyl, 3-bromobenzyl, 4-brornobenzyl, 3- methoxy benzyl, 3-nitrobenzyl, 3-aminobenzyl, 4-(aminosulfonyl)benzyl, 1- phenylethyl, 2-phenylethyl, (3,5-dimethylisoxazol-4-yl)methyl, (5-nitro-2- furyl)methyl or l-(ethoxycarbonyl)propyl. In some embodiments R4 is n-butyl, n- hexyl, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3-aminobenzyl, (3,5-dimethylisoxazol-4-yl)rnethyl, (5-nitro-2-furyl)methyl or 1 - (ethoxycarbonyi)propyl. In some embodiments R4 is 3-methoxybenzyl, 3- nitrobenzyl or (5-nitro-2-furyl)methyl.

Generally R5 is C2-4 alkyl. Usually R5 is unsubstituted C2-4 alkyl. In some embodiments R5 is ethyl.

Generally R6 is C2-4 alkyl, amido or— COOR7. Usually R6 is unsubstituted C2-4 alkyl, amido or— COOR7. In some embodiments R6 is ethyl, amido or ethoxycarbonyl. In some embodiments R6 is ethoxycarbonyl. Generally R7 is CI -4 alkvl. Usually R7 is unsubstituted CI -4 alkyl. In some embodiments, R7 is ethyl.

Usually the invention provides compounds having formula I, their enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers), or pharmaceutically acceptable salts thereof,

wherein

Rl is hydrogen, CI -6 alkyl optionally substituted by hydroxy, alkoxy, cyano, ethyrryl, alkoxycarbonyl or acyl;

R2 is hydrogen or unsubstituted CI -4 alkyl;

R3 is a group of formula— CHR5R6 or a benzyl group;

R4 is CI -8 alkyl optionally substituted by cyclohexyl, phenyl, bromophenyi, aminophenyi, methoxyphenyl, nitrophenyl, aminosulfonylphenyl, 3,5- dimethylisoxazol-4-yl, 5-nitro-2-furyl or ethoxycarbonyl;

R5 is unsubstituted C2-4 alkyl;

R6 is unsubstituted C2-4 alkyl, ami do or— COOR.7;

R7 is unsubstituted CI -4 alkyl;

with the proviso that when Rl is hydrogen, R2 is methyl, 1 3 is CHR5R.6, R6 is ethoxycarbonyl and R5 is ethyl, then R4 is different from n-propyl, i-propyl, n- pentyl, n-heptyl, 3-bromobenzyl, 4-chlorobenzyS, 4-methyibenzyl or 2-phenylethyl.

In the above embodiment, sometimes, when R3 is a benzyl group, then R4 is CI -8 alkyl optionally substituted by alkoxycarbonyl. In the above embodiment, sometimes, when R3 is a group of formula— CHR5R6, then R4 is Cl-8 alkyl optionally substituted by C3-6 cyeloalkyl, aryl or heterocycle.

In one embodiment, Rl is hydrogen, methyl, cyanornethyl, 2-ethoxy-2-oxoethyl, 2-methoxyethyl, n- propyl, 2-oxopropyl, 3-hydroxypropyl, 2-propynyl, n-pentyl or n-hexyl;

R2 is hydrogen, methyl or n-butyi;

R3 is 3-pentyl, 1 -(aminocarbonyl)propyl, l-(ethoxycarbonyl)propyl or 3~ bromobenzyi;

R4 is n-butyl, i-butyl, n-pentyl, n-hexyl, cyclohexylmethyl, benzyl, 2- bromobenzyl, 3 -bromobenzyi, 4-bromobenzyl, 3-methoxybenzyi, 3-nitrobenzyl, 3- aminobenzyl, 4-(aminosulfonyl)benzy[, 1-phenylethyl, 2-phenylethyl, (3,5- dimethy]isoxazol-4-y])methyi, (5-nitro-2-furyl)m.ethyl or 1 - (ethoxycarbonyi)propyl;

with the proviso that when Rl is hydrogen, R2 is methyl and R3 is 1-

(ethoxycarbonyi)propyi, then R4 is different from n-pentyl, 3~bromobenzy{ or 2- phenylethyl.

In the above embodiment, sometimes, when R3 is 3 -bromobenzyi, then R4 is Cl-8 alkyl optionally substituted by aikoxycarbonyl. In the above embodiment, sometimes, when R3 is 3-pentyl, 1 -

(aminocarbonyi)propyl or l-(ethoxycarbonyl)propyl, then R4 is different from 1- (ethoxycarbonyl)propyi.

In a more preferred embodiment, Rl is hydrogen, methyl, cyanomethyl, 2- methoxyethyl, n-propyl, 3-hydroxypropyl or 2-propynyl;

R2 is methyl;

R3 is 3-pentyl, 1 -(aminocarbonyl)propyl, l-(ethoxycarbonyl)propyl or 3- bromobenzyl; R4 is n-butyl, n-hexyi, benzyl, 3-bromobenzyl, 3-methoxybenzyl, 3-nitrobenzyl, 3- aminobenzyl, (3,5-dimethylisoxazol-4-yl)methyi, (5-nitro-2-fuxyl)methy! or 1 - (ethoxycarbonyl)propyl:

with the proviso that when Rl is hydrogen, R2 is methyl and R3 is 1- (ethoxycarbonyl)propyl, then R4 is different from 3-brornobenzyl.

In the above embodiment, sometimes, when R3 is 3-bromobenzyl, then R4 is 1- (ethoxycarbonyl)propyl;

In the above embodiment, sometimes, when R3 is 3-pentyl, 1 - (aminocarbonyl)propyi or l-(ethoxycarbonyl)propyl, then R4 is different from 1- (ethoxycarbonyl)propy ί ;

In one embodiment, Rl is hydrogen; R2 is methyl; R3 is 1 - (ethoxycarbonyi)propyl; and R4 is 3-methoxybenzyl, 3-nitrobenzyl or (5-nitro-2- furyi)methyl.

A further embodiment consists in compounds wherein R2 is methyl, R3 is a group of formula— CHR5R6 with R5 being C2-4 alkyl, R6 being amido or— COOR7 and 1 7 being methyl or ethyl.

In some embodiments, compounds are ethyl 2-[(7-benzyl-l,3-dimethyl-2,6-dioxo- 2,3 ,6,7-tetrahydro- 1 H- urin-8- 3.)thio]butanoate; ethyl 2~ { [7-(3-bromobenzyl)- 1 ~ (2-ethoxy-2-oxoethyl)-3-methyl-2,6-dioxo-2 ,6 etrahydro-l H-p riti-8- yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-l-(2-methoxyethyl)-3-methyl-2,6- dioxo-2,3,6,7-tetrahydro-lH-purin-8-yi]thio}butanoate; ethyl 2- {[7-(3- bromobenzyi)~2,6-dioxo-2,3,6,7-tetrahydro-lH-purin-8-yl]thio }butanoate; ethyl 2- {[7-(3-bromobenz}d)-l.,3-dimethyl-2,6-dioxo-23,6 5 7-tetrahydro-l H-purin-8- y ijthio } butanoate; ethyl 2- { [7-(2-bromobenzyl)- 1 ,3-dimethyl-2,6-dioxo-2,3 ,6,7- tetrahydro-1 Bl-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-l- (cyanomethy[)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-purin -8- yljthiojbutanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-l-propyl- 2,3,6,7-tetrahydro-lH-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3- methyl-2.6-dioxo- 1 -(2-oxopropyl)-2,3 ,6,7-tetrahydro- 1 H-purin-8- yijthiojbutanoate; ethyl 2- {[7-(3-bromobenzyl)-l .-(3-hydroxypropyl)-3-methyl- 2,6-dioxo-2,3 ,6,7-tetrahydro- 1 H-puriii-8-yl]thio } butanoate; ethyl 2-{[7-(3- bromobenzyl)-3-methyl-2,6-dioxo- 1 -(2-propynyl)-2,3 ,6,7-tetrahydro- 1 H-purin-8- yl]thio}butanoate; ethyl 2-{[7-(3-methoxybenzyl)-3-methyl-2,6-dioxo-2,3,6,7- tetrahyd.ro- lH-purin-8-yl]thio}buta.noate; ethyl 2-{[3-methyl-7-(3-nitrobenzyl)- 2,6-dioxo-2,3 ,6,7-tetrahydro- 1 H-purin-8-yl]thio} butanoate; ethyl 2-{[7-(3- aminobenzyl)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-purin- 8- yljthiojbutanoate; ethyl 2-({7-[4-(aminosulfonyl)benzyl]-3-methyl-2,6-dioxo- 2,3 ,6,7-tetrahydro- 1 H-purin-8-yl} thio)butanoate; ethyl 2- { [7-(4-bromobenzyl)- 1 ,3 dimethyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-purin-8-yl]thio}but anoate; ethyl 2-{[7- (cyclohexylmethyl)-1 ,3-diraethyl-2,6-dioxo-23,6,7-tetrahydro-lH-p-urin-8- yl]thio}butanoate; ethyl 2-{[l,3-dimethyl-2,6-dioxo-7-(l-phenylethyl)--2,3,6,7- tetrahydro-lH-piirm-8-yl]thio}butanoate; ethyl 2-{[1 ,3-diraethyl-2,6-dioxo-7-(2- phenylethyl)-2,3,6J-tetrahydro-lH-purin-8-yl]thio}butanoate; ethyl 2-({7-[(3,5- dimethylisoxazol~4~yl)methyl]-3-m

yl} thio)butanoate; ethyl 2-( {3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo- 2,3,6,7-tetrahydro-lH-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methyl-2,6- dioxo-2,3,6,7-tetrahydro-lH-purin-8-yl)thio]butanoate; ethyl 2- {[7-(3- bromobenzyl)-2,6-dioxo-2,3,6,7-tetrahydro-ni-purm-8-yl]thio} butanoate; ethyl 2- [(l,7-dihexyl-3-methy[-2,6-dioxo-2,3,6,7-te1rahydro-lH-purin -8-yl)thio]butanoate ethyl 2-[(7-hexyl-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-pmin-8- yi)thio]butanoate; ethyl 2-[(3-methyl-2,6-dioxo-l ,7-dipentyi-2,3,6,7-tetrahydro- lH-purin-8-yl)thio]butanoate; 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-2,3,6,7- tetrahydro-l H-purin-8-yl]thio}butanarnide; 2-[(7-butyi-3-methyl-2,6-dioxo- 2,3,6,7-tetrahydro-lH-purin-8-yl)thio]butanamide; 7-(3-bromobenzyl)-8-[(l- ethylpropyl)thio]-3-rnethyl-3,7-dihydro-lH-purine-2,6-dione; ethyl 2-{8-[(3- bromoberizy[)thio]-l,3-dimethyl-2,6-dioxo-l,2,3,6-te1rahydro -7H-purin-7- yljbutanoate; and ethyl 2~[(7-isobutyl-3-methyl-2,6~dioxo-2,3,6,7 etrahydro-l H- purin-8-y[)thio]butanoate.

In some embodiments compounds are: ethyl 2-[(7-benzyi- 1 ,3-dimethyl-2,6-dioxo- 2,3 ,6,7-tetrahydro- 1 H-purin-8- l)tbio]butanoate; eth l 2~ { [7-(3-bromobenzyl)- 1 - (2-methoxyethyl)-3-niethyl-2,6-dioxo-2,3,6 7-tetrahydro- lH-purin-8- yl]thio}butanoate; ethyl 2- {[7-(3-bromobenzyl)-l ,3-dimethyl~2,6-dioxo-2,3,6,7- tetrahydro-lH-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)- l- (cyanomethyl)-3-methyl-2,6-dioxo

yl]thio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-l-propyl- 23,6J-tetrahydro-lH-purin-8-yl]tliio}butanoate; ethyl 2-{[7-(3-bromobenzyl)-l- (3-hydroxypropyi)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-p urvn-8- yl]thio}butanoate; ethyl 2- {[7-(3-bromobenzyl)-3-methyl-2,6-dioxo-l-(2- propynyl)~2,3,6,7-tetrahydro~lH-purin~8-yl]thio}butar jate; ethyl 2-{[7-(3- niethoxyben¾4)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-pur in-8- yl]thio}butanoate; ethyl 2-{[3-rnethyl~7~(3-nitrobenzyl)~2,6~dioxo-2,3,6,7- tetrahydro-l H-purin-8-yl]thio}butanoate; ethyl 2-{[7-(3-aminobenzyl)-3-rnethyl- 2,6-dioxo-2,3,6J-tetrahydro-lH-purin-'8-yl]thio}butanoate; ethyl 2-({7-[(3,5- dimethylisoxazol-4-yl)methyl]-3-methyl-2,6-dioxo-2,3,6,7-tet rahydro-l H-purin-8- yl}thio)butanoate; ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo- 2,3,6,7-tetrahydro-l H-purin-8-yl}thio)butanoate; ethyl 2-[(7-butyl-3-methy 1-2,6- dioxo-2 ,6,7-tetrahyd.ro-lH~purin-8-yl)thio]buta.noate; ethyl 2-[(7-hexyl-3- methyl-2 5 6-dioxo-2,3,6,7-tetrahydro-lH-purin-8-yl)thio]butanoat e; 2-{[7-(3- bromobenzy[)-3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-lH-purin- 8- yljthi o } butanamide; 7~(3 -bromobenz l)-8 - [( 1 ~ethylpropyl)thio] -3 -meth l -3 ,7- dihydro- lH-purine-2,6-dione; and ethyl 2- { 8-[(3-bromobenzyl)thio]- 1 ,3-dimethyl- 2,6-dioxo-l,2.3 5 6-telTahydro-7H-purin-7-yl}buta.noate.

In some embodiments compounds are: ethyl 2- {[7-(3-methoxybenzyl)-3 -methyl - 2,6-dioxo-2,3,6,7-tetrahydro-lH-purin-8-yl]thio}butanoate; ethyl 2-{[3-methyl-7- (3-nitrobenzyl)-2,6-dioxo-23,6,7-tetrahydro-lH-purin-8-yl]th io}buto and ethyl 2-({3-methyl-7-[(5-nitro-2-furyl)methyl]-2,6-dioxo-2,3,6,7-t etrahydro-lH- purin- 8 -y 1} thio)butanoa te .

The acid addition salt form of a compound of formula I that occurs in its tree form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalie such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, maionic, succinic, maieic, fumaric, malic, tartaric, citric, methanesulfomc, ethanesulfonic, beiizenesulfonic, p-toiuenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like. The compounds of formula I containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.

Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.

Compounds of the formula I and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like.

Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Ciiem., 45 (1976) 11-30.

The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).

With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically.

Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention.

xiii) U.S. Patent 7,465,549

In some embodiments, the compound includes optionally substituted N-alkylated 2-oxo-pyrroiidine derivatives. In some embodiments, those compounds are alkyl amides derivatives substituted on the positions 4 and/or 5 of the pyrrolidone ring. Examples of optionally substituted N-alkylated 2-oxo-pyrrolidine derivatives include, but are not limited to, compounds such as (2S)-2-[(4S)-4-(2,2- difluoroviny!)-2-oxopyrro!idinyl]butanamide, (2S)-2-[(4R)-2-oxo-4- piOpylpyrrolidinyljbutanamide, (2S)-2-[(4S)-2-oxo-4- propylpyrrolidinyljbutanamide, and (2S)-2-[4-(3-azidophenyl)-2-oxopyrrolidin- 1 - yljbutanamide,

In some embodiments, the compounds further include optionally substituted N- alkylated 2-oxo-piperidinyl derivatives. In some embodiments, those compounds are alkyl amides derivatives substituted on the position 4 and/or 5 and/or 6 of the 2-oxo-piperidinyl ring. Examples of optionally substituted N-alkylated 2-oxo- pyrrolidine derivatives include, but are not limited to, compounds such as those referred to in international patent application PCT/EP02/05503 such as (2S)-2~[5- (iodomethyl)-2-oxo- 1 -piperidinyl] butanamide, (2S)-2-[5-(azidomethyl)-2-oxo- 1 - piperidinyljbutanamide, 2-(2-oxo-5-phenyl- 1 -piperidin ljbutanamide, (2S)-2-[4- (iodomethyl)-2-oxo- 1 -piperidinyljbutanamide, and (2S)-2-[4-(2-fiuoro-2- methylpropyl)-2-oxo- 1 -pyrroiidinyljbutanamide.

In some embodiments, the compounds include any acetam compound of formula I, in racemic or isomeric form, or a pharmaceutically acceptable salt thereof,

wherein

R represents hydrogen or hydroxy;

Rl and R2 represent independently hydrogen or an alkyl group of 1-4 carbon atoms; and R3 and R4 represent independently hydrogen, an alkyl group of 1-4 carbon atoms or (CH2)n R5R6 wherein n is 1 , 2 or 3 and R5 and R6 represent

independently hydrogen or an alkyl group of 1-4 carbon atoms.

An example of such an acetam compound includes, but is not limited to, a compound of formula I wherein R, Rl , R2, 1 3 and R4 are hydrogen, 2-oxo- pyrrolidineacetamide, known by the generic name piracetam as described in UK Patents Nos. 1,039,1 13 and 1,309,692.

In some embodiments, the compounds also include optionally substituted N- aikyiated 2-oxo-azepanyl derivatives. Preferably, those compounds are alkyl amides derivatives substituted on the positions 4 and/or 5 and/or 6 and/or 7 of the 2-oxo-azepanyl ring. Examples of optionally substituted N-alkylated 2-oxo- azepanyl derivatives include, but are not limited to, compounds such as those referred to in international patent application PCT/EP02/05503 such as 2-[5- (iodomethy l)-2-oxo- 1 -azepanyljbutanamide.

xiv) U.S. Patent Application Publication No. 2006258704

This invention provides novel compounds of the formula I

wherein n represents 0 or 1 whereby R<1 > is not existent when n :::: 0 and R<1 > is existent when n=l ;

A<1 > represents an oxygen or a sulfur atom:

X is -CONR<7> R<8> , -COOR<9> , -CO-R<10 > or CN;

R<1 > when existent, R<2> , R<3> , R<4 > and R<5 > are the same or different and each is independently hydrogen, halogen, hydroxy, thiol, amino, nitro, nitrooxy, cyano, azido, carboxy, amido, sulfonic acid, sulfonamide, alkyl, alkenyl, alkynyi, ester, ether, ary , heterocycle, or an oxy derivative, thio derivative, amino derivative, acyl derivative, sulfonyl derivative or sulfinyl derivative,

provided that at least one of the suhstituents R chosen from R<1 > when existent, R<2> , R<3> , R<4 > or R<5 > is not hydrogen;

R<6 > is hydrogen, alkyl, aryl or -CH2-R<6a > wherein R<6a > is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;

R<7> , R<8 > and R<9 > are the same or different and each is independently hydrogen, hydroxy, alkyl, aryl, heterocycle or an oxy derivative; and

R 10 > is hydrogen, hydroxy, thiol, halogen, alkyl, aryl, heterocycle or a thio derivative;

their pharmaceutically acceptable salts, geometrical Isomers (including cis and trans, Z and E isomers), enantiomers, diastereoisomers and mixtures thereof (including all possible mixtures of stereoisomers).

In the above formula, at least one substituent R<1 > to R<5 > is different from hydrogen. Some non-substituted compounds are referred to in U.S. Pat. Nos.

5,468,733 and 5,516,759. U.S. Pat. No. 5,468,733 discloses non-ring substituted 2~ oxo-l-pyrrolidinyl and 2-oxo-l-piperidinyl derivatives as inhibitors of the oncogene Ras protein. In particular, these compounds block the ability of Ras to transform normal cells to cancer cells, and therefore can be included in several chemo therapeutic compositions for treating cancer.

US Patent No. 5,516,759 discloses non-ring substituted 2-oxo-l-pyrrolidinyl, 2- oxo-1 -piperidinyl and azepanyl derivatives present at the N-terminus of dodecapeptides possessing LHRH (luteinizing honnone-releasing hormone) antagonistic acti vity . Such LH IIH antagonists are useful in the treatment of a variety of conditions in which suppression of sex steroids plays a key role including contraception, delay of puberty, treatment of benign prostatic hyperplasia a.o.

In the definitions set forth below, unless otherwise stated, R<11 > and R<! 2 > are the same or different and each is independently amido, alkyl, alkenyl, alkynyi, acyl, ester, ether, aryl, aralkyl, heterocycle or an oxy derivative, thio derivative, acyl derivative, amino derivative, sulfonyl derivative, or sulfinyl derivative, each optionally substituted with any suitable group, including, but not limited to, one or more moieties selected from lower alkyl or other groups as described below as substituents for alkyl.

The term "oxy derivative", as used herein, is defined as including -0-R<l 1 > groups wherein R<11 > is as defined above except for "oxy derivative". Non- limiting examples are alkoxy, alkenyloxy, alkynyioxy, acyioxy, oxyester, oxyamido, alkylsulfoiiyloxy, alkylsuifiiiyloxy, aryisulfonyloxy, arylsulfinyloxy, aryloxy, araikoxy or heierocyclooxy such as peniyloxy, aliyloxy, methoxy, ethoxy, phenoxy, benzyloxy, 2-naphthyloxy, 2-pyridyloxy, methylenedioxy, carbonate.

The term "thio derivative", as used herein, is defined as including -S-R<11 > groups wherein R<11 > is as defined above except for "thio derivative". Non- limiting examples are alkylthio, aikenyithio, alkynylthio and arylthio.

The term "amino derivative", as used herein, is defined as including -NHR<1 1 > or -NR<11> R<12 > groups wherein R<11 > and R<12 > are as defined above. Non- limiting examples are mono- or di-alkyl-, alkenyl-, alkynyl- and arylamino or mixed amino.

The term "acyl derivative", as used herein, represents a radical derived from carboxylic acid and thus is defined as including groups of the formula R<11> -CO- , wherein R<11 > is as defined above and may also be hydrogen. Preferred are acyl derivatives of formula -C0R<11 > wherein R<11 > is selected from hydrogen, Cl- 12 alkyl, C2-12 alkenyl, C2-12 alkenyl, heterocyle and aryl. Non-limiting examples are formyl, acetyl, propionyl, isobutyryl, valeryl, lauroyL heptanedioyl, cyciohexanecarbonyi, crotonoyl, fumaroyl, acryloyl, benzoyl, naphthoyl, furoyl, nicotinoyl, 4-carboxybutanoyl, oxalyl, ethoxalyl, cysteinyl, oxamoyl.

The term "sulfonyl derivative", as used herein, is defined as including a group of the formula ~SO~R<1 1 > , wherein R<1 1 > is as defined above except for "sulfonyl derivative". Non-limiting examples are alkylsulfonyl, alkenylsulfoiiyl,

alkynylsuifonyl and arylsulfonyl ,

The term "sulfinyl derivative", as used herein, is defined as including a group of the formula -SO-R<l i> , wherein R<11 > is as defined above except for "sulfinyl derivative". Non-limiting examples are alkylsulfinyl, alkenylsulfinyl,

alkynylsulfinyl and arylsulfinyl.

The term "alkyl", as used herein, is defined as including saturated, monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof and generally containing 1-20 carbon atoms, most often 1 to 12 carbon atoms, preferably 1-7 carbon atoms for non-cyclic alkyl and 3-7 carbon atoms for cycloalkyl (in these two preferred cases, unless otherwise specified, "lower alkyl"), each optionally substituted by, preferably 1 to 5, substituents independently selected from the group consisting of halogen, hydroxy, thiol, amino, nitro, cyano, thiocyanato, acyl, acyloxy, sulfonyl derivative, sulfinyl derivative, aikylamino, carboxy, ester, ether, amido, azido, cycloalkyl, sulfonic acid, sulfonamide, thio derivative, alkylthio, oxyester, ox amido, heterocycle, vinyl, alkoxy (preferably CI -5), aryloxy (preferably C6-10) and aryl(preierably C6-10).

In some embodiments are alkyl groups containing 1 to 7 carbon atoms, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkylthio, cyclopropyl, acyl and phenyl. Most preferred are CI -4 alkyl and C3-7 cycloalkyl, each optionally substituted by one or more hydroxy, halogen, lower alkyl or/and azido.

In some embodiments are alkyl groups are hydroxymethyl, propyl, butyl, 2,2,2- trifluoroethyl, 2-bromo-2,2-difluoroethyl, 2-chloro-2,2-difluoroethyl, 3,3,3- trifluoropropyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2,2-diiIuoropropyl, 2-iodo-2,2-difluoroethyl.

The term "lower alkyl", as used herein, and unless otherwise specified, refers to CI to C7 saturated straight, branched or cyclic hydrocarbon, Non limiting examples are methyl, ethyl, propyl, isopropyl, butyl, tertiobutyl, pentyl, cyclopropyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, 3-methypentyl, 2,2- dimethylbutyl, optionally substituted with any suitable group, including but not limited to one or more moieties selected from groups as described above for the alkyl groups. Preferably, lower alkyl is methyl.

The term "alkenyl", as used herein, is defined as including both branched and unbranched, unsaturated hydrocarbon radicals having at least one double bond, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol, amino, thiocyanato, azido, alkylthio, cycioalkyl, acyl, nitro, cyano, aryl and heterocycie.

In some embodiments are alkenyl groups are C2-C12 alkenyls, especially C2- 6alkenyls, such as ethenyS (=vinyl), 1 -methyl- 1 -ethenyf 2,2-dimethyl-l -ethenyl, 1- propenyl, 2-propenyl (=allyi), 1-butenyl, 2-butenyl, 3-butenyl, 4-pentenyl, 1- methyl-4-pentenyl, 3-methyS-l-pentenyl, 1-hexenyl, 2-hexenyi and the like, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycioalkyl, phenyl and acyl. Most prefered is vinyl, optionally substituted by one or more halogen or/ and lower alkyl, and especially 2,2-difluorovinyl, 2,2-dibromovinyl and 2,2-dicmorovinyl.

The term "alkynyl" as used herein, is defined as including a monovalent branched or unbranciied hydrocarbon radical containing at least one carbon-carbon triple bond, for example ethynyl, 2-propynyl ( ; =propargyS), and the like, and being optionally substituted by at least one substituent selected from the group consisting of halogen, hydroxy, thiol , amino, nitro, cyano, aryl, heterocycie, thiocyanato, azido, alkylthio, alkyl and acyl.

In some embodiments are alkynyl groups are C2-I2 alkynyl, especially C2-6 alkynyl, optionally being substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, acyl, aryl such as phenyl and alkyl, preferably cycioalkyl.

In some embodiments are ethynyl, propynyl and butynyl, optionally substituted by lower alkyl or/and halogen, and especially I -propynyl, cyclopropylethynyl, 3- methyl- 1 -butynyl and 3 ,3 ,3-trifluoro~ 1 -propynyl,

When present as bridging groups, alkyl, alkenyl and alkynyl represent straight- or branched chains, C1-I2, preferably Cl-4-alkylene or C2-12-, preferably C2-4- alkenyiene or -a!kynylene moieties respectively .

Groups where branched derivatives are conventionally qualified by prefixes such as "n", "sec", "iso" and the like (e.g. "n-propyl", "sec-butyl") are in the n-form unless otherwise stated. The term "aryl", as used herein, is defined as including an organic radical derived from an aromatic hydrocarbon consisting of at least one ring, most often 1 to 3 rings and generally containing 6-30 carbon atoms by removal of one hydrogen, such as phenyl and naphthyl, each optionally substituted by one or more substituents independently selected from halogen, hydroxy, thiol, amino, nitro, cyano, acyl, acyloxy, sulfonyi, sulfinyi, alkylamino, carboxy, ester, ether, amido, azido, sulfonic acid, sulfonamide, alkylsulfonyl, alkylsulfinyl, Cl-6-alkylthio, oxyester, oxyamido, aryl, Cl-6-alkoxy, C6-10-aryloxy, Cl -6-alkyl, Cl-6- haloalkyl. Aryl radicals are preferably monocyclic or bicyclic containing 6-10 carbon atoms. Preferred aryl groups are phenyl and naphthyl each optionally substituted by one or more substituents independently selected from halogen, nitro, amino, azido, Cl-6-alkoxy, Cl-6-alkyl, Cl-6-haioalkyl, sulfonyi and phenyl.

In some embodiments the ar l is phenyl, optionally substituted by one or more halogen, lower alkyl, azido or nitro, such as 3-chlorophenyl and 3-azidophenyl.

The term "halogen", as used herein, includes an atom of CI, Br, F, L

The term "hydroxy", as used herein, represents a group of the formula -OH.

The term "thiol", as used herein, represents a group of the formula -SH.

The term "cyano", as used herein, represents a group of the formula -CN.

The term "nitro", as used herein, represents a group of the formula -NG2.

The term "nitrooxy", as used herein, represents a group of the formula -ON 02.

The term "amino", as used herein, represents a group of the formula -NH2.

The term "azido", as used herein, represents a group of the formula ~N3.

The term "carboxy", as used herein, represents a group of the formula -COOH.

The term "sulfonic acid", as used herein, represents a group of the formula -S03H.

The term "sulfonamide", as used herein, represents a group of the formula - S02NH2.

The term "ester", as used herein, is defined as including a group of formula -COO- R<11 > wherein R<11 > is as defined above except oxy derivative, thio derivative or amino derivative. Preferred are esters of formula -COQR<! 1 > wherein R<11 > is selected from Cl-12 alkyl, C2-12 alkenyl, C2-12 alkynyl and aiyl. Most preferred are esters where R<11 > is a lower alkyl especially methyl.

The term "ether" is defined as including a group selected from Cl -50-straight or branched alkyl, or C2-50-straigbt or branched alkenyl or alkynyl groups or a combination of the same, interrupted by one or more oxygen atoms.

The term "amido" is defined as including a group of formula -CONH2 or - CQNHR<! 1 > or -C0NR<11> R<12 > wherein R<1 1 > rand R<12 > are as defined above.

The term "heterocycie", as used herein, is defined as including an aromatic or non aromatic cyclic alkyl, alkenyl, or alkynyl moiety as defined above, having at least one O, S and/or atom interrupting the carbocyciic ring structure and optionally, one of the carbon of the carbocyciic ring structure may be replaced by a carbonyl, and optionally being substituted with any suitable group, including but not limited to one or more moieties selected from lower alkyl, or other groups as described above for the alkyl groups. Non-limiting examples of heterocycies are pyridyi, furyl, pyrrolyl, thienyi, isothiazolyl, triazolyl, imidazolyi, benzimidazolyl, tetrazolyl, quinazolinyl, quinolizinyl, naphthyridinyl, pyridaziny!, pyrimidinyi, pyrazinyl, quinolyi, isoquinolyl, isobenzofuranyl, benzotiiienyi, pyrazolyl, indolyl, indolizinyi, purinyl, isoindolyl, carbazolyl, thiazolyl, 1,2,4-thiadiazolyl, thiomorpholinyl, thieno(2,3-b)furanyl, furopyranyl, benzofuranvl, benzoxepinyl, isooxazolyl, oxazolyl, thianthrenyl, benzothiazolyl, or benzoxazolyl, cinnolinyl, phthalazinvl, quinoxalinyl, 1-oxidopyridyl, phenanthridinvl, acridinyl, perimidinyl, phenanmrolirryl, phenothiazinyl, furazanyl, benzodioxolyl, isochromanyl, indolinyl, xanthenyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazoiopyridinyi, imidazoiopyridinyi, pyrrolopyrimidinyi, pyrazoiopyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, piperidyl, piperazinyl, imidazolidinyi, morpholino, morpiiolinyl, l-oxaspiro(4.5)dec-2-yl, pyrrolidinyl, 2- oxo-pyrrolidinyl, sugar moieties (i.e. glucose, pentose, hexose, ribose, fructose, which may also be substituted) optionally substituted by alkyl or as described above for the alkyl groups. The term "heterocycie" also includes bicyclic, tricyclic and tetracyclic, spiro groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring or another monocyclic heterocyclic ring or where a monocyclic heterocyclic group is bridged by an aikyiene group, such as quinuclidinyl, 7-azabicyclo(2.2.1)heptanyl, 7- oxabicyclo(2.2. l)heptanyl, 8-azabicyclo(3.2. i)octanyl.

The heterocycle may be selected from triazolyi tetrazo!yl, pyrrolidinyl, pyridy!, 1- oxidopyridyl, thiomorpholinyi, benzodioxoiyi, furyl, oxazoiyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyl, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alkyl, alkoxy, nitro, amino, acyl and phenyl. In some embodiments the heterocycle is selected from tetrazolyl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each option ally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyl, alkoxy, nitro, amino and phenyl, and especially from 2-and 3-thienyl, optionally substituted by one or more halogen, acyl such as formyl, cyano and/or lower alkyl, such as methyl. In the above definitions it is to be understood that when a substituent such as R<1> , R<2> , R<3> , R<4> , R<5> , R<7> , R<8> , R<9> , R<10 > is attached to the rest of the molecule via a heteroatom or a carbonyl, a straight- or branched chain, CI .-12-, preferably Cl-4-aIkylene or C2-12, preferably C2-4-aIkenylene or - alkynylene bridge may optionally be interposed between the heteroatom or the carbonyl and the point of attachment to the rest of the molecule.

The acid addition salt form of a compound of formula (I) that occurs in its tree form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maieic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like.

The compounds of formula (I) containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt form, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.

Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.

Compounds of the formula ] and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, aicohoiates and the like.

Many of the compounds of formula I and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. ( 1976), 45, 1 1-30.

The invention also relates to all stereoisomeiic forms such as enantiomeric and diastereoisomeric forms of the compounds of formula I or mixtures thereof (including all possible mixtures of stereoisomers).

Furthermore, certain compounds of formula I which contain alkenyl groups may exist as Z (zusammen) or E (entgegen) isomers, in each instance, the invention includes both mixture and separate individual isomers.

Multiple substituents on the piperidinyi or the azepanyl ring can also stand in either cis or trans relationship to each other with respect to the plane of the piperidinyi or the azepanyl ring.

Some of the compounds of formula I may also exist in tautomeric form s. Such forms although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.

With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof unless the particular isomeric form is referred to specifically.

The invention also includes within its scope prodrug forms of the compounds of formula I and Its various sub-scopes and sub-groups. The term "prodrug" as used herein includes compound forms which are rapidly transformed in vivo to the parent compound according to the invention, for example, by hydrolysis in blood. Prodrugs are compounds bearing groups which are modified by biotransformation prior to exhibiting their pharmacological action. Such groups include moieties which are readily oxidised, cyclised or cleaved, which compound after biotransformation remains or becomes pharmacologically active. For example, metabolically cleavable groups form a class of groups well known to practitioners of the art. They include, but are not limited to such groups as alkanoyl (i.e. acetyl, propiony!, butyryl, and the like), unsubstituted and substituted carbocyciic aroyl (such as benzoyl, substituted benzoyl and 1- and 2- naphthoyl), alkoxycarbony! (such as ethoxycarbonyl), trialkylsilyl (such as trimethyl- and triethylsilyl), moiioesters formed with dicarboxylic acids (such as suecinyi), phosphate, sulfate, sulfonate, sulfonyl, sulfinyl and the like. The compounds bearing the biotransiormable groups have the advantage that they may exhibit improved bioavailability as a result of enhanced solubility and/or rate of absorption conferred upon the parent compound by virtue of the presence of the biotransiormable group. T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery System", Vol. 14 of the A.C.S. Symposium Series; "Bioreversible Carriers in Drug Design", ed. Edward B. Roche, American Pharmaceutical Association and

Pergamon Press, 1987.

The term "R substituent" refers to R< 1> , R<2> , R<3> , R<4 > or R<5 > , independently.

According to one embodiment, the present invention relates to a compound of formula I as defined above wherein n represents 0, The compound is a 6-ring structure (2-thioxo- or 2-oxo-piperidinyl derivative) wherein R<1 > is not existent since n=0, and is depicted by the formula (I-A).

(T-A)

According to a following embodiment, the present invention relates to a compound of formula I according to the invention as defined above wherein n represents 1 . The compound is a 7-ring structure (2-thioxo- or 2-oxo-azepanyl derivative) wherein R<1 > is existent since n=l and depicted by the formula (I-B).

According to one embodiment, the invention relates to said compound as defined above wherein n 0, R<3 > and/or R<4 > are different from hydrogen and R<2 > and R<5 > represent hydrogen.

According to another embodiment, the invention relates to said compound as defined above wherein n=l, R<2> , R<3 > and/or R<4 > are different from hydrogen and wherein R<1 > and R<5 > represent hydrogen.

According to another embodiment, the invention relates to said compound as defined above wherein only one R substituent chosen from R<3 > or R<4 > when n=0 or from R<2> , R<3 > or R<4 > when n=l, is different from hydrogen and the remaining R substituent(s) is/are hydrogen. We hereby refer to a mono-substituted 2-thioxo- or 2-oxo-piperidinyl or 2-thioxo- or 2-oxo-azepanyl derivatives.

According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein A<1 > represents an oxygen atom. We hereby refer to 2-oxo-piperidinyl or 2-oxo-azepanyl derivatives. According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein X is CONR<7> R<8> , especially CONH2. We hereby refer to amido derivatives of 2-oxo(or thioxo)-piperidinyl or 2-oxo(or thioxo)-azepanyl.

According to another embodiment, the present invention relates to compounds of formula I according to the invention as defined above wherein R<6 > represents hydrogen, CI -4 alkyi, or a CH2-R<6a > group wherein R<6a > represents a heterocycle. Most preferably R<6 > is a C 1-4 alkyl, especially ethyl. When R<6 > is ethyl we refer to 2-(2-oxo(or thioxo)-l -piperidinyi)butanamide or 2-(2-oxo(or thioxo)- 1 -azepa.nyl)butananiide derivatives.

According to another embodiment, the present invention relates to compounds of formula ί according to the invention as defined above wherein the carbon atom to which R<6 > is attached is of the S configuration. In case where R<6 > is ethyl, A is oxygen and X is CON R<7 > R<8> , we refer then to (2S)-2-(2-oxo-l- piperidinyi)butanamide or (2S)-2-(2-oxo-l-azepanyl)butanamide derivatives.

According to one embodiment, the present invention relates to a compound as defined above wherein R<2 > w r hen n=l , R<3 > and R<4 > are the same or different and each is independently hydrogen, halogen, nitro, nitrooxy. cyano, carboxy, aniido, sulfonic acid, sulfonamide, alkyl, alkenyi, alkynyl, ester, ether, aryl, heterocycle, acyl derivative, sulfonyl derivative or sulfinyl derivative:

R<1 > when existent, R<2 > when n=0 and R<5 > are hydrogen;

R<6 > is hydrogen, alkyl, aryl or -CH2~R<6a > wherein R<6a > is aryl, heterocycle, halogen, hydroxy, amino, nitro or cyano;

provided that, when R<6 > is hydrogen, X is -CONR<7> R<8 > and that the compound is neither methyl (2R)-2-[(6R.)-6-methyl-2-oxoazepanyl]-3-phenyipropanoate nor methyl (2S)-2-[(4R)-4-methyl-2-oxoazepanyl]-3-phenylpropanoate.

According to this embodiment, the compound is generally such that when R<6 > is benzyl, X is -COOCH3 and n=l, R<2 > is different from methyl when R<3 > and R<4 > are both hydrogen and R<4 > is different from methyl when R<2 > and R<3 > are both hydrogen.

According to another embodiment, the present invention relates to a compound as defined above wherein R<2 > when n=l, R<3 > and R<4 > are the same or different and each is independently hydrogen; cyano; carboxy; amido; Cl-12 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cycloalkyl, acyl, aryl and heterocycle;

C2-12 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl;

C2-12 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, alkyl, aryl and acyl; acyl derivative of formula -C0-R<11> , wherein R<11 > is selected from Cl-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, heterocycle and aryl;

ester of formula -CO-0-R<l 1 > wherein R<! 1 > is selected from Cl -12 alkyl, C2- 12 alkenyl, C2-12 alkynyl and aryl;

heterocycle selected from triazolyl, tetrazolyl, pyrrolidinyl, pyridyl, 1- oxidopyridyl, thiornorpholinyl, benzodioxo!yl, fury}. oxazolyS, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl and piperazinyi, each optionally substituted by one or more substituents selected from halogen, alkyl, substituted alky], alkoxy, nitro, amino, acyl and phenyl;

aryl, each optionally substitued by one or more substituents selected from Cl-6 alkyl, Cl-6 haloa!kyl, Cl -6 alkoxy, Cl -6 alkylthio, amino, azido, sulfonyl, aryl and nitro,

According to another embodiment, the present invention relates to a compound as defined above, wherein R<2 > when n=l, R<3 > and R<4 > are the same or different and each is independently hydrogen;

CI -7 alkyl, each optionally substituted by one or more substituents selected from hydroxy, halogen, cyano, thiocyanato, alkoxy, azido, alkyltio, cyclopropyl, acyl and phenyl;

C2-6 alkenyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl:

C2-6 alkynyl, each optionally substituted by one or more substituents selected from halogen, cyano, thiocyanato, azido, alkylthio, cycloalkyl, phenyl and acyl:

Ζζ, ί heterocycle selected from tetrazolvl, pyrrolidinyl, pyridyl, furyl, pyrrolyl, thiazolyl and thienyl, each optionally substituted by one or more substituents selected from halogen, alkyl, halogen substituted alkyl, acyi, alkoxy, nitro, amino and phenyl: phenyl, each optionally substitued by one or more substituents selected from CI -6 alkyl, halogen substituted alkyl, halogen, alkoxy, amino, azido, suifonyl, phenyl and nitro.

According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2> , R<3 > and R<4 > when n=l or from the group R<3 > and R<4 > when n=0, represents independently CI -4-aikyi or C3-7-eyeloaikyi, optionally substituted by one or more halogen, hydroxy, lower alkyl and/or azido.

According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2> , R<3 > and R<4 > when η=1 or from the group R<3 > and R<4 > when n-0, represents independently vinyl, optionally substituted by one or more halogen or/and lower alkyl.

According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2> , R<3 > and R<4 > when n=l or from the group R3 and R<4 > when n=0, represents independently ethynyl, propynyl or butynyl, optionally substituted by one or more halogen and/or lower alkyl.

According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2> , R<3 > and R<4 > when n=l or from the group R<3 > and R<4 > when n=0, represents independently phenyl, optionally substituted by one or more halogen, lower alkyl, azido and/ or nitro.

According to another embodiment, the present invention relates to a compound as defined above wherein at least one of the R substituents chosen from the group R<2> , R<3 > and R<4 > when n=l or from the group R<3 > and R<4 > when n=0, represents independently 2- or 3-thienyi, optionally substituted by one or more halogen, acyl, cyano or/and lower alkyl. According to a particular embodiment, the present invention rela tes to a compound as defined above wherein at l east one of the R substituents chosen from the group R<3> , R<4 > and R<2 > when n=l or from the group R<3 > and R<4 > when n=0„ is hydroxymethyi, propyl, butyl, 3,3,3-trifiuoropropyl, 2,2,2-trifluoroethyl, cyclopropylmethyl, iodomethyl, azidomethyl, 2-thienyl, 3-thienyl, phenyl, 3- chlorophenyl, 3-azidophenyi, 2,2-difluorovinyl, 2,2-dibromovinyl, 2,2- dichlorovinyl, 2-ethynyl, 5-niethyl-2-thienyl, 5-formyl-2-ethynyl, 5-cyano-2- thienyi, 3-bromo-2~thierryl, 4-methyl-2-thienyl, 3,3,3-triiluoro-l-propynyl, 1- propynyl, cyclopropylethynyS, 3-methyi-l -butynyS, 1-butynyl, 2,2-difluoropropyl, 2-chloro-2,2-difiuoroethyl, 2-bromo-2,2-difiuoroethyl and 2-iodo-2„2- difluoroethyl.

According to yet another embodiment, the present invention relates to a compound as defined above wherein R<1> , R<2> , R<4 > and R<5 > are hydrogen,

According to another embodiment, the present invention relates to a compound as defined above wherein R<1> , R<2> , R<3 > and R<5 > are hydrogen.

According to another embodiment, the present invention relates to a compound as defined above wherein n=l and R<1> , R<3> , R<4 > and R<5 > are hydrogen.

In all the above-mentioned scopes when the carbon atom to which R<6 > is attached is asymmetric it may be in the "S"-configuration,

Representative compounds of this invention as defined above are selected from the group consisting of 2-[5-(hydroxymethyl)-2-oxo-l-piperidinyl]butanamide, 2-(2- oxo-5-propyl-l-piperidinyl)butanamide, 2-12-oxo-5-(3,3,3-trifluoropropyl)-l- piperidinyljbutanamide, 2-[5-(cyclopropylmethyl)-2-oxo-l- piperidinyijbutanamide, 2-[5-(iodomethyl)-2-oxo-l-piperidinyl]butanamide, 2-[5- (azidomethyi)-2-oxo- 1 -piperidinyijbutanamide, 2-(2-oxo-5-phenyl- 1 - piperidiiiyl)butanamide, 2-[2-oxo-5-(2-thienyl)-l -piperidinyijbutanamide, 2-[2- oxo-5-(3-thienyl)-l -piperidinyijbutanamide, 2-[5-(3-chlorophenyl)-2-oxo-l- piperidinyijbutanamide, 2-[5-(3-azidophenyl)-2-oxo- 1 -piperidinyijbutanamide, 2- [5~(2,2~diiluorovinyl)~2-oxo-l -piperidinyijbutanamide, 2~[5-(2,2-dibromovinyl)-2- oxo-1 -piperidinyijbutanamide, 2-[5-(2,2-dichlorovinyl)-2-oxo-l- piperidinyljbutanamide, 2-(5-ethynyl-2-oxo- 1 -piperidinyijbutanamide, 2-[5-(5- methyl-2-thienyl)-2-oxo- 1 -piperidinyljbutanamide,, 2-[5-(5-formyl-2-thienyl)-2- 0X0-1 -piperidinyljbutanamide, 2-[5-(5-cyano-2-thienyl)-2-oxo-l - piperidinyljbutanamide, 2-[5-(3-bromo-2-thienyl)-2-oxo-l -piperidinyljbutanamide, 2-[5-(4-methyl-2-thienyl)-2-oxo-l -piperidinyljbutanamide, 2-[2-oxo-5-(3,3,3- trif iuoro- 1 -propynyl)- 1 -piperidinyljbutanamide, 2- [2-oxo-5 -( 1 -propynyl)- 1 - piperidinyljbutanamide, 2-[5-(cyclopropylethynyl)-2-oxo-l- piperidinyljbutanamide, 2-[5-(3-methy[-l -butynyl)-2-oxo- 1 - piperidinyljbutanamide, 2-[5-(l -butynyl)-2-oxo- 1 -piperidinyljbutanamide, 2-[5- (2,2-difluoropropyi)-2-oxo 1 -piperidinyljbutanamide, 2-[5-(2-chloro-2,2- difluoroethyl)-2-oxo-l -piperidinyljbutanamide, 2-[5-(2-bromo-2,2-difluoroethyl)- 2-oxo- 1 -piperidinyl] butanamide, 2-[4-(hydroxymethyl)-2-oxo- 1 - piperidinyljbutanamide, 2-(2-oxo-4-propyl- 1 -piperidinyljbutanamide, 2-[2-oxo-4- (3,3,3-trifluoroproyl)-l -piperidinyljbutanamide, 2-14-(cyclopropylmethyl)-2-oxo- 1 -piperidinyljbutanamide, 2-[4-(iodomethyl)-2-oxo- 1 -piperidinyljbutanamide, 2- [4-(azidomethyl)-2-oxo-l -piperidinyljbutanamide, 2-(2-oxo-4-phenyl-l- piperidinyl)butanamide, 2- 12-oxo-4-(2-thienyl)-l -piperidinyljbutanamide, 2-[2- oxo-4-(3 -thi enyl)- 1 -pi peridmyljb u tanamide, 2- [4-(3 -chlorophenyl)-2-oxo- 1 ~ piperidinyljbutanamide, 2-[4-(3-azidophenyl)-2-oxo- 1 -piperidinyljbutanamide, 2- [4-(2,2-diiluorovinyl)-2-oxo-l-piperidinyljbutanamide, 2-[4-(2,2-dibromovinyl)-2- 0X0-1 -piperidinyljbutanamide, 2-[4-(2,2-dichlorovinyl)-2-oxo-l - piperidinyljbutanamide, 2-(4-ethynyl-2-oxo- 1 -piperidinyi)butanamide, 2-[4-(5- methyi-2-thienyl)-2-oxo-l -piperidinyljbutanamide, 2-[4-(5-formyl-2-thienyl)-2- oxo- 1 -piperidinyljbutanamide, 2 - [4-(5 -cyano-2-thienyl)-2-oxo- 1 - piperidinyljbutanamide, 2-[4-(3-bromo-2-thienyl)-2-oxo-l -piperidinyljbutanamide, 2-[4-(4-methyl-2-thienyl)-2-oxo-l -piperidinyljbutanamide, 2-[2-oxo-4-(3,3,3- trifluoro- 1 -propynyl)- 1 -piperidinyljbutanamide, 2-[2-oxo-4-( 1 -propynyl)- 1 - piperidinyljbutanamide, 2-[4-(cyclopropylethynyl)-2-oxo-l- piperidinyljbutanamide, 2-[4-(3-methyl-l-butynyl)-2-oxo-l- piperidinyijbutanamide, 2-[4-(l -butynyl)-2-oxo-l -piperidinyljbutanamide, 2-[4- (2,2-diiluoropropyl)-2-oxo- 1 -piperidinyljbutanamide, 2-[4-(2-chloro-2,2- difluoroethyl)-2-oxo-l -piperidinyljbutanamide, 2-14-(2-bromo-2,2-difluoroethyl)- 2-oxo- 1 -piperidinyljbutanamide, 2-[4-(2,2,2-trifluoroethyl)-2-oxo- 1 - piperidiiiyijbuianamide, 2-[5-(hydroxymethyl)-2-oxo- 1 -azepanyljbutanamide, 2- (2-0X0-5 -propyl- 1 -azepanyl)butanami de, 2- [2 -oxo-5 -(3 ,3 ,3 -irifluoropropy !)- 1 - azepanyljbutanamide, 2-(5-(cyclopropylmethyl)-2-oxo- 1 -azepanyljbutanamide, 2- [5-(iodomethyl)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(azidomethyl)-2-oxo- 1 - azepanyljbutanamide, 2-(2-oxo-5-phenyl-l-azepanyl)butanamide, 2-[2-oxo-5-(2- thienyl)- 1 -azepanyljbutanamide, 2-[2-oxo-5-(3-thienyl)- 1 -azepanyljbutanamide, 2- [5 -(3 -chlorophenyl)-2-oxo-l -azepanyljbutanamide, 2-[5-(3-azidophenyl)-2-oxo-l- azepanyl jbutanamide, 2-[5-(2,2-difluorovmyl)-2~oxo- 1 -azepanyljbutanamide, 2- [5-(2,2-dibromovinyl)-2-oxo-l -azepanyljbutanamide, 2-[5-(2,2-dichlorovinyi)-2~ oxo- 1 -azepanyljbutanamide, 2-(5-ethynyl-2-oxo-l -azepanyljbutanamide, 2-[5-(5- methyi-2 -thieny l)-2-oxo- 1 -azepanyljbutanamide, 2- [5 -( 5 -formyl-2 -thieny l)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(5-cyano-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(3-bromo-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[5-(4-methyi-2- thienyl)-2-oxo- 1 -azepanyl Jbu tanamide, 2-[2-oxo-5-(3 ,3 ,3 - trifluoro- 1 -propynyl)- 1 - azepanyljbutanamide, 2-[2-oxo-5-(l -propynyl)- 1 -azepanyljbutanamide, 2-[5-

(cyclopropylethynyl)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(3-methyl- 1 -butynyl)-2- oxo-1 -azepanyljbutanamide, 2-[5-(l-butynyl)-2-oxo-l -azepanyljbutanamide, 2-[5- (2,2-difluoropropy[)-2-oxo- 1 -azepanyljbutanamide, 2-[5-(2-chioro-2,2- difluoroethyl)-2-oxo- 1 -azepanyljbutanamide, 2- [5 ~(2-bromo-2 ,2-difluoroethyl)-2- oxo-1 -azepanyljbutanamide, 2-[5-(2,2,2-trifluoroethyl)-2-oxo-l - azepanyljbutanamide, 2-[6-(hydroxymethyl)-2-oxo-l -azepanyljbutanamide, 2-(2- oxo-6-propyl-l -azepanyl jbutanamide, 2-[2-oxo-6-(3,3,3-trifluoropropyl)-l - azepanyljbutanamide, 2-[6-(cyclopropylmethyl)-2-oxo- 1 -azepanyljbutanamide, 2- [6-(iodomethyl)-2-oxo- 1 -azepanyljbuta amide, 2- 16-(azidomethyl)-2-oxo- 1 - azepanyljbutanamide, 2-(2-oxo-6-phenyl- 1 -azepanyl butanamide, 2-[2-oxo-6-(2- thienyl)- 1 -azepanyljbuianamide, 2-[2-oxo-6-(3-thienyl)- 1 -azepanyljbutanamide, 2- [6-(3-chlorophenyl)-2-oxo- 1 -azepanyljbutanamide, 2-[6-(3-azidopheny[)-2-oxo- 1 - azepanyljbutanamide, 2-[6-(2,2-difluorovinyl)-2-oxo- 1 -azepanyljbutanamide, 2- [6-(2,2-dibromovinyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(2,2-dichlorovinyi)-2- oxo- 1 -azepanyljbutanamide, 2-(6-ethyny 1-2-oxo- 1 -azepan l)butanamide, 2-[6-(5- methyi-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(5-formyi-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[6-(5-cyano-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[6-(3-bromo-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(4-methyl-2- thienyl]-2-oxo-l -azepanyljbutanamide, 2-[2-oxo-6-(3,3,3-trifluoro-l-propynyl)-1 - azepanyljbutanamide, 2-[2-oxo-6-(l-propynyl)- 1 -azepanyljbutanamide, 2-[6- (cyclopropylethynyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(3-methyi-l-butynyl)-2- oxo- 1 -azepanyljbutanamide, 2-[6-( 1 -butynyl)-2-oxo- 1 -azepanyljbutanamide, 2-[6- (2,2-diiluoropropyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(2~chloro~2,2~ difluoroethyl)-2-oxo-l -azepanyljbutanamide, 2-[6-(2-bromo-2,2-difluoroethy[)-2- oxo-1 -azepanyljbutanamide, 2-[6-(2,2,2-trifluoroethyl)-2-oxo-l- azepanyljbutanamide, 2-[4-(hydroxymethyl)-2-oxo-l -azepanyljbutanamide, 2-(2- oxo-4-propyl- 1 -azepanyljbutanamide, 2-[2-oxo-4-(3 ,3,3 -trifluoropropyl)- 1 - azepanyljbutanamide, 2- 14-(cyclopropylmethyl)-2-oxo-l -azepanyljbutanamide, 2- [4-(iodomethyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(azidomethyl)-2-oxo- 1 - azepanyljbutanamide, 2-(2-oxo-4-phenyl-l -azepanyl)butanamide, 2-[2-oxo-4-(2- thienyl)- 1 -azepanyljbutanamide, 2-[2-oxo-4-(3-thienyl)- 1 -azepanyljbutanamide, 2- f4-(3-chlorophenyl)-2-oxo-l -azepanyljbutanamide, 2-[4-(3~azidophenyl)-2-oxo-l- azepanyljbutanamide, 2-[4-(2,2-difluorovinyi)-2-oxo- 1 -azepanyljbutanamide, 2- [4-(2,2-dibromovinyl)-2-oxo-l -azepanyljbutanamide, 2-[4~(2,2~dichlorovinyl)-2~ oxo- 1 -azepanyljbutanamide, 2-(4-ethynyl-2-oxo- 1 -azepanyljbutanamide, 2-[4-(5- methyl-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(5-formyl-2-thienyl)-2-oxo- 1 -azepanyljbutanamide, 2-[<4> -(5-cyano-<2> -thienyl)-2-oxo-l- azepanyljbutanamide, 2-[4-(3-bromo-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2- [4-(4-methyl-2-thienyl)-2-oxo-l -azepanyljbutanamide, 2-[2-oxo-4-(3,3,3-trifiuoro- 1 -propvnyl)- 1 -azepanyljbutanamide, 2-[2-oxo-4-( 1 -propvnyl)- 1 - azepanyljbutanamide, 2-[4-(cyclopropylethynyl)-2-oxo- 1 -azepanyljbutanamide, 2- [4-(3-methy[- 1 -butynyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-( 1 -butynyl)-2-oxo- 1 - azepanyljbutanamide, 2-[4-(2,2-difluoropropyl)-2-oxo- 1 -azepanyljbutanamide, 2- [4-(2-chloro-2,2-difluoroethylj-2-oxo-l -azepanyljbutanamide, 2-[4-(2-bromo-2,2- difluoroethyl)-2-oxo- 1 -azepanyljbutanamide, 2-[4-(2,2,2-trifluoroethyl)-2-oxo- 1 - azep any 1 Jbutanamide .

Results have been obtained with the following compounds:

(2S)-2-[5-(iodomethyl)-2-oxo-l-piperidinyljbutanamide.

(2S)-2-[5-(azidomethyl)-2-oxo-l -piperidinyl jbutanamide, 2-(2-oxo-5-phenyl- 1 -piperidinyijbutanamide,

(2S)-2-[4-(iodomethyl)-2-oxo- 1 -piperidinyijbutanamide,

2-[5-(iodomethy !)-2-oxo~ 1 -azepanyljbutanamide.

xv) International Patent Application Publication No. WO2008/132139 In some embodiments, the compounds are of formula (I) as follows:

wherein

Y is O or S. in some embodiments Y is O. Rl is hydrogen or C-j.g alkyl;

R2 is hydrogen;

R3 is -CONR5R6, -COR7, an imidazolyi, an irnidazopyridinyl, an

imidazopyridazinyl; R5, R6 are the same or different and are independently selected from hydrogen and C-| 6 alkyl;

R7 is </ ; 6 alkyl;

A is a monocyclic or bicyciic heterocyclic moiety selected from the group consisting of imidazolidin-l-yl, 1 ,3-oxazolidin-3-yl, 2,5-dihydro-l H-pyrrol-l -yl, 1 ,3-thiazol-3(2H)-yl, 1 ,3- thiazolidin-3-yl, piperidin-l-yl, azepan-l-yl, 5,6- dihydro-4H-thieno[3,2-b]pyrrol-4-yl, hexahydro-4H-thieno[3,2-b]pyrrol-4-yl, 2,3- dihydro-1 H-thieno[3,4-b]pyrrol-l -yl, 1 ,3- benzothiazol-3(2H)-yl, 1 ,3- benzoxazol-3(2H)-yl, pyrazoIo[l ,5-a]pyridin-l (2H)-yi, 3,4- dihydroisoquinolin- 2(1 H)-yl, 3,4-dihydroquinoiin-l (2H)-yl, 1 ,3,4,5-tetrahydro-2H-2- benzazepin-2- yl, 1 ,2,4,5-tetrahydro-3H-3-benzazepin-3-yS; R4 is either R A a or R A b depending on whether A being is a monocyclic or a bicyciic heterocycle:

where A is a monocyclic heterocyclic moiety, R A is R A a which is selected from the group consisting of hydrogen; C-j.g alkyl optionally substituted by a substituent selected from halogen, C-1.4 alkoxy, C-1.4 alkylthio, azido, nitrooxv or an aryl; C2-6 alkenvl optionally substituted by halogen; C2-6 alkynyl optionally substituted by halogen; azido; aikoxycarbonylamino; arylsulfonyloxy; a substituted or unsubstituted ary ; or a 3-8 membered substituted or unsubstituted heterocycle; where A is a bicyclic heterocyclic moiety R A is R A which is selected from the group comprising or consisting of hydrogen; nitro; cyano; halogen; heterocycle; amino; aryl; C-j.g alkyl optionally substituted by at least one halogen; or C-j.g alkoxy optionally substituted by at least one halogen;

In some embodiments the compounds are as follows:

For compounds where A=Y is selected from a 2-oxo-piperidin- 1 -yl, a 2-oxo- azepan-l-yl, a 2-oxo-l ,3-benzothiazol-3(2H)-yl or a 2-oxo-l ,3-benzoxazol- 3(2H)- yl, R3 must beselected from an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl.

For compounds where A=Y is a 5-oxoimidazolidin-l-yi, R A and R A are hydrogen, R3 is -CONR5R6, R5 ancj R6 are as above defined, then R A a may not be an alkyl, araikyi or substituted aralkyl.

Where A=Y is either of a 2-oxo-piperidin- 1 -yl and a 2-oxo-azepati-l -yl, R A , R A and R A a are all hydrogen, then R A could not be a 2-phenylimidazo[ 1 ,2-a]pyridin- 3-yl.

In a specific embodiment A=Y is selected from the list consisting of:

wherein X is O or S, in a more specific embodiment O; in another embodiment, X is S.

The asterisks in the above illustration indicate the attachment sites of the substituent R A a,

In a specific embodiment, when R A is -CONR5R6 and R A is C^g alkyl, the carbon atom to which R-I and R A are attached is preferably in the "S" -configuration.

In a specific embodiment R A is hydrogen, methyl, ethyl and R A is hydrogen. In a specific embodiment R3 is -CONH2.

In a further specific embodiment R A is 1 H-imidazol-l-yl, 1 H-imidazol-4-yl, 1 H~ imidazoi-5- yl, imidazo[l ,2-a]pyridin-3-yl or imidazo[l ,2-b]pyridazin-3-yl. In a specific embodiment R A a is a C-j.g alkyl which may optionally be substituted by a halogen; or a phenyl.

In another specific embodiment R A b is hydrogen, halogen, nitro, cyano or a€-μ alkyl optionally substituted by a halogen. In still a further embodiment compounds may be used in the treatment of the above mentioned disorders, in particular of epilepsy, having the formula (I-E), as wells as its geometrical isomers, enantiomers, diastereomers and mixtures, or a

pharmaceutically acceptable salt thereof!,

wherein

X is O or S;

R-I is hydrogen or C-|,g alkyl, in a more specific embodiment hydrogen;

R3 is an imidazoiyl, an imidazopyridinyl, an imidazopyridazinyl; R A h is hydrogen; nitro; cyano; halogen; C~].g alkyl optionally substituted by halogen; C~j.g alkoxy optionally substituted by halogen.

A further aspect of the present invention consists in novel compounds having the formula (I-A), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

wherein

Rl is hydrogen or C-j.g alkyl, preferably hydrogen, methyl or ethyl; in a more specific embodiment R A is ethyl.

R3 is -CONH2, an imidazoiyl, an imidazopyridinyl, an imidazopyridazinyl, preferably R A is••COM 12.

R A a is either hydrogen or an aryl; with the proviso that 2-(5-oxoimidazo3.idm-l- yljacetamide is excluded. Preferably R A a is an aryl, e.g. a phenyl which may be substituted preferably by halogen, nitro, alkoxy, in particular by nitro. In a particular embodiment, when R A is -CONH2 and R A is C-j.g alkyl, the carbon atom to which Rl and R A are attached is preferably in the '^"-configuration.

A further aspect of the present invention consists in novel compounds having the formula (I-Bl or I-B2), their geometrical isomers, enaniiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

0-B1) (i-B2) wherein X in formula (I-B2) is either S or O, in a more specific embodiment 8;

Rl is hydrogen or C-j.g alkyl, preferably hydrogen, methyl or ethyl; in a more specific embodiment R A is ethyl

R3 is -CONH2, an imidazolyl, an irmdazopyridinyl, an imidazopyridazinyl; preferably R A is -CONH2

R A a is hydrogen; C-j.g alkyl optionally substituted by halogen or C-1.4 alkoxy; an aryl; or C2.g alkenyl optionally substituted by halogen, Preferably, R A a is C-j.g alkyl optionally substituted by halogen or C2-6 alkenyl optionally substituted by halogen or a aryl. In a more specific embodiment R A a is C~j.g alkyl optionally substituted by halogen or aryl.

In a particular embodiment, when R A is -CONH2 and R A is C-j.g alkyl, the carbon atom to which R-] and R A are attached is preferably in the -configuration.

A further aspect of the present in vention consists in novel compounds having the formula (I-B3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

wherein

Rl is either hydrogen or C- g alky], preferably hydrogen, methyl or ethyl; more preferably Rl is ethyl.

R3 is -CONH2. an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl;

preferably R A is -CONH2 R A a is C-| 5 alkyl optionally substituted by halogen or C-1.4 alkoxy; an aryl; or C2_g alkenyl optionally substituted by halogen.

Preferably, R A a is C-j.g alkyl optionally substituted by halogen or C2 g alkenyl optionally substituted by halogen.

In a particular embodiment, when R A is -CONH2 and R A is C-j,g alkyl, the carbon atom to which R-] and R A are attached is preferably in the -configuration.

A further aspect of the present in vention consists in novel compounds having the formula (I-C), their geometrical isomers, eiiantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

wherein

Rl is hydrogen or C~j,g alkyl, in particular hydrogen, methyl or ethyl,

1 3 is -CO H2, an imidazolyl, an imidazopyridinyl, an imidazopyridazinyl; in particular R A is -CONH2

R A a is methyl, ethyl, butyl optionally substituted by halogen or C-1.4 alkoxy, an unsubstituted phenyl or a phenyl substituted by halogen, a C-j.g alkyl optionally substituted by halogen or a C-1.4 alkoxy; or R A a is a C2-6 alkenyl optionally substituted by halogen. Preferably, R A a is methyl, optionally substituted by halogen, an unsubstituted phenyl or a phenyl substituted by halogen.

In a particular embodiment, when R A is -CONH2 and R A is C-j.g alkyl, the carbon atom to which R l and R|3 are attached is preferably in the "S"-configuration. A further aspect of the present invention consists in compounds having the formula (1-Dl or I-D2), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmace tically acceptable salt thereof,

(I-D1 ) il-D2) wherein

R~I is hydrogen or C-j.g alkyl, in particular hydrogen; R3 is an imidazolyl, a imidazopyridinyl or an imidazopyridazinyi. In one embodiment, R A is 1 H- imidazol-l-yl, 1 H-im.idazol-4-yl, 1 H-imidazol-5-yl, imidazo[l ,2-a]pyridin-3~yl or imidazo[l ,2-b]pyridazin-3-yl. In a more specific embodiment, R A is 1 H- imidazol-l-yl, 1 H- imidazol-4-yl, 1 H-imidazol-5-yl, imidazo[l ,2-a]pyridin-3-yl; R A a is hydrogen, C~].g alkyl optionally substituted by halogen or C-1.4 aSkoxy; aryl: or C2- g alkenyl optionally substituted by halogen. In a specific embodiment, R A a is C-j.g alkyl optionally substituted by halogen; aryl; or C2-6 alkenyl optionally substituted by halogen. In a more specific embodiment R A a is C-j.g alkyl optionally substituted by halogen; or aryl; e.g, propyl or phenyl;

with the proviso that when R A and R A a are hydrogen, R A is not 2-phenyiimidazo[] ,2- a]pyridin-3-yl.

A further aspect of the present invention consists in compounds having the formula (I-Fl , I-F2 or I-F3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

(M¾} (I-F3) wherein R-I is hydrogen or C-|,g alkyl, preferably hydrogen, methyl or ethyl; more preferably, R A is hydrogen.

R3 is -CO H2, an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl; in a more specific embodiment R3 is -CONH2, 1 H-imidazol-l -yl, 1 H-imidazol-4-yl, 1 H-imidazoi-5-yl, imidazo[l ,2-a]pyridin-3-yl or imidazo[l ,2-b]pyridazin-3-yl. R A b is hydrogen; halogen; nitro; cyano; CI . alkyl optionally substituted by halogen; C-l .4 alkoxy optionally substituted by halogen. In a more specific embodiment R A is hydrogen, halogen or cyano, more specifically halogen.

In a particular embodiment, when R A is -CONH2 and R A is C-j.g alkyl, the carbon atom to which R l and R|3 are attached is preferably in the "S"-configuration.

A further aspect of the present invention consists in compounds having the formula (I-F4), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

wherein

R-I is hydrogen or C-j.g alkyl, preferably hydrogen;

R3 is an imidazolyl, an imidazopyridinyl or an imidazopyridazinyl; more specifically R A is I H-imidazol-l- l, 1 H-imidazol-4-yl, 1 H-imidazol-5-yl, imidazo[l ,2-a]pyridin-3-yl or imidazo[l ,2-b]pyridazin-3-yl. More specifically R is 1 H-imidazol-4-yl or irnidazoj ,2- a]pyridin-3-yi.

R A b is hydrogen; halogen; nitro; cyano; C-l .4 alkyl optionally substituted by halogen; C-l .4 alkoxy optionally substituted by halogen; specifically R A is hydrogen, halogen or cyano,.

In a particular embodiment, when R A is -CONH2 and R A is C-j.g alky], the carbon atom to which R-I and R A are attached is preferably in the ' " -configuration. A further aspect of the present invention consists in compounds having either of the formula (I-Gl , 1-G2 or 1-G3), their geometrical isomers, enantiomers, diastereomers and mixtures, or a pharmaceutically acceptable salt thereof,

{«-61} (f-G2> (I-G3)

wherein

R-I is hydrogen or C-j.g alkyl; preferably hydrogen;

R3 is -CONH2, an imidazoiyl, an imidazopyridinyl, an iniidazopyridazinyl; in a more specific embodiment R A is -CO H2, 1 H-imidazol-l-yi, 1 H-imidazol-4-yl, 1 H-imidazol-5-yl, imidazo[l ,2-a]pyridin-3-yl or imidazo[l ,2-b]pyridazin-3-yL In a even more specific embodiment R3 is an 1 H-imidazol-4-yl or imidazo[l ,2- a]pyridin-3-yl;

R4D js hydrogen; halogen; C-1.4 alkyl optionally substituted by halogen; C-1.4 alkoxy optionally substituted by halogen.

Specific compounds of the present in vention are those selected from the group consisting of: (2S)-2-[3-(4-nitrophenyl)-5-oxoimidazolidin-l -yljbutanamide; (2Sj- 2-[3-(2,4- dinitrophenyl)-5-oxoimidazolidin-l -yljbutanamide; (2S)-2-(5-oxo-3- phenyiimidazoiidin- 1 - yljbutanamide; 2-[5-(iodomethyl)-2-oxo- 1 ,3-oxazolidin-3- yijbutanamide; 2-(2~oxo-2,5- dihydro-1 H-pyrrol-l-yl)butanamide; 2-(2-oxo-4- phenyl-2,5-dihydro- 1 H-pyrrol- 1 - yljbutanamide; 2-(4-methyl-2-oxo-2,5-dihydro- 1 H -pyrrol- 1 -yljbutanamide; (2S)-2-(2-oxo-5- propyl-1 ,3-thiazol-3(2H)- yljbutanamide; 2-(2-oxo-5 -propyl-1 ,3-thiazol-3(2Hj- yljpropanamide; 2-(5-butyl- 2-oxo-l ,3 -thiazolidin-3 -yljbutanamide; 2-(5-butyl-2-oxo-l ,3- thiazolidin-3- yljpropanamide; 2-(2-oxo-5-phenyl-l ,3-thiazolidin-3-yl)propanamide; 2-(2- oxo- 5-propyi- 1 ,3-thiazolidin-3-yljbutanamide; 2-(2-oxo-5 -phenyl- 1 ,3-thiazolidin-3- yljbutanamide; 2-(2-oxo-5-propyl-l ,3-thiazoiidin-3-yl)propanamide; (2S)-2-[2- oxo-5-(2,2,2- trifluoroethyl)-l ,3-thiazolidin-3-yl]butanamide; l-{[6-chloro-2- (triiluoromethyljimidazo[l ,2- b]pyridazin-3-yi]methyi}piperidin-2~one; 1 ~(1 H- imidazol-4 -y Imethy l)-5 -propy lpiperidin-2- one; 1 -( 1 H-imidazol- 1 -ylmethyl)-5 - propylpiperidin-2-one; 1 -(imidazo[l ,2-a]pyridin-3- ylmethyl)-5-propylpiperidin- 2-one; 1-(1 H-imidazol-l-ylmethyl)-5-phenylpiperidin-2-one; 1- (imidazo[l ,2- a] pyridm-3-ylmethyi)-5-phenylpiperidin-2-one; 1 -(imidazo[ 1 ,2-ajpyridin-3- ylmethyl)-4-phenylpiperidin-2-one; 1-(1 H-imidazol- l-yimethyl)-4- phenylpiperidin-2-one; 1 - (imidazo[ 1 ,2-a]pyridin-3-ylmethyl)-4-propylpiperidin-

2- one; 1-(1 H-imidazoI-5-ylniethyl)-4- propylpiperidin-2-one; 1-(1 H-imidazol- 1- ylmethyl)-4-propylpiperidin-2-one; 1 -{[6-chloro-2- (trifluoromethyl)imidazo[l ,2- b] pyridazin-3-yl]methyi}azepan-2-one; 1 -(1 H-imidazol-5- ylmethyl)-5- propylazepan-2-οηε; 5 -propyl- 1 - { [2-(trifluorometliyl)imidazo [ 1 ,2-a jpyridin-3 - y Ijmethyl} azepan-2-one; 5-phenyl- 1 - {[2-(trifluoromethyl)imidazo[ 1 ,2-ajpyridin-

3- y Ijmethyl} azepan-2-one; 1-(1 H-imidazol-5-ylmethyl)-6-propylazepan-2-one; 1- (1 H- imidazol-4-ylmethyl)-4-propylazepan-2-one; 4-(l H-imidazol-4-yimethyl)- 4,6-dihydro-5H- tiiieno[3,2-b]pyrrol-5-one; 2-(5-oxo-5,6-dihydro-4H-thieno[3,2- bjpyrrol-4-yl)acetamide; 4- {[2-(trifluorom.ethyl)imidazo[l ,2-ajpyridin-3- y[jmethyl}-4,6-dihydro-5H-thieno[3,2-bjpyrrol-5- one; 4- {[2- (trifluoromethyl)imidazo[l ,2-a]pyridm-3~yl]methyl}hexahydro-5H-thieno[3,2~ bjpyrrol-5-one; 1-(1 H-imidazol-4-ylmethyl)- 1 H-thieno[3,4-bjpyrrol-2(3H)-one; 2-(6-chloro- 2-0X0-1 ,3-benzothiazol-3(2H)-yl)acetamide; 6-bromo-3-(l H- imidazol-1 -y lmethyi)- 1 ,3- benzothiazol-2(3H)-one; 2-(6-bromo-2-oxo- 1 ,3- benzothiazol-3(2H)-yl)propanamide; 2-(6- bromo-2-oxo-l ,3-benzothiazol-3(2H)- yl)propanamide; 2-(6-fluoro-2-oxo-l ,3-benzothiazol- 3(2H)-yS)acetamide; 2-(6- methyl-2-oxo-l ,3-benzothiazol-3(2H)-yl)acetamide; 6-fluoro-3- (1 H-imidazol- 1 - yimethyl)-] ,3-benzoxazol-2(3H)-one; H-imidazol-4- ylmethyl)pyrazolo[l ,5- a]pyridin-2(l H)-one; 2-(6-chloro-3-oxo-3,4-dihydroisoquinolin- 2(1 H)- yljpropanamide; 5-chloro-2-(l H-imidazol-4-y lmethyi)- 1 ,4-dihydroisoquinolin- 3(21 1 )· one; 2-(6-chloro-2-oxo-3,4-dihydroquino[in-l (2H)-yl)acetamide; 2-(6- bromo-2-oxo-3,4- diiiydroquinoiiii-1 (2H)-yl)acetamide; 1-(1 H-imidazol-4- yimethyl)-3,4-dihydroquinolin-2(l H)- one; 2-(6-iodo-2-oxo-3,4-dihydroquinolin-l (2H)-yl)acetamide; 2-(6-cyano-2-oxo-3,4- diiiydroquinoiiii-1 (2H)-yl)acetamide; 7- chloro-2-{[2-(trifluoromethyl)imidazo[l ,2-ajpyridin- 3-yljmethyl} -l ,2,4,5- tetrahydro-3H-2-benzazepin-3-one; 7-chloro-2-(l H-imidazol-4- yimethyl)-! ,2,4,5-tetrahydro-3H-2-benzazepin-3-one; 7-chloro-3-(l H-imidazol-4-ylmethyl)- 1 ,3,4,5-tetrahydro-2H-3-benzazepin-2-one; and 7-ch!oro-3-{[2- (trifluoromethyl)imidazo[ 1 ,2-a]pyridm-3-yl]methyl}-l ,3,4,5-tetrahydro-2H-3- benzazepin-2- one.

In some embodiments, compounds of the present invention are those selected from the group consisting of: 1-(1 H-imidazol-4-ylmethyl)-5-propylpiperidin-2-one; 1- ( 1 H-imidazol-1 - ylmethyl)-5-propylpiperidin-2-one; l-(imidazo[l ,2-a]pyridin-3- ylmethyl)-5-propylpiperidin-2- one: l-(i H-imidazol-l-ylmethyl)-5- phenylpiperidin-2-one; 1 ~(imidazo[ 1 ,2-a]pyridin-3~ ylm.ethyl)-4-phenylpiperidin- 2-one; 1-(1 H-imidazol-1 -ylmethy[)-4-phenylpiperidin-2-one; 1 - (imidazo[l ,2- a]pyridin-3-y3methyl)-4-propy3piperidm-2-one; 1-(1 H~imidazol-5-ylmethyl.)-4- propylpiperidin-2-one; 1 -( 1 H-imidazoi- 1 -ylmethyl)-4-propylpiperidin-2-one; 1 -( 1 H-imidazol- 4-ylmethyl)-l H-thieno[3,4-b]pyrrol-2(3H)-one; 6-bromo-3-(l H- imidazol-l -ylmethyl)-l ,3- benzothiazol-2(3H)-one; 2-(6-bromo-2-oxo-l ,3- benzothiazol-3(2H)-yl)propanamide; and 5-c-hloro-2-(l H-imidazol-4-ylmethyl)-l ,4-dihydroisoquinolin-3(2H)-one.

The foll owing paragraphs provide definitions of the various chemical moieti es that make up the compounds according to the invention and are intended to apply uniformly through- out the specification and claims unless an otherwise expressly set out definition provides a broader definition.

"C~j β alkyl" refers to alkyl groups having 1 to 6, or 1 to 4 carbon atoms. This term is exemplified by groups such as methyl, ethyl, n-propyi, isopropyl, n-butyl, isobutyl, tert- butyl, n-penlyl, n-hexyl, trifiuoromethyl and the like. "Aryl" refers to an unsaturated aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl). Preferred aryl include phenyl, naphthyl, phenantrenyl and the like.

"Heterocycle" refers to a saturated or unsaturated ring system containing, in addition to carbon atoms, at least one hetero atom, such as nitrogen, oxygen and/or sulfur. "Heterocycle" includes both "heteroaryl" and "heterocycloalkyS".

"Heteroaryl" refers to a monocyclic heteroaromatic, or a bicyclic or a tricyclic fused-ring heteroaromatic group. Particular examples of heteroaromatic groups

"> ' ¾7 include optionally substituted pvridyl, pyrrolyl, fur I. thienvl, imidazolvl, oxazolyl, isoxazoiyl, thiazolyl, isothiazolyl, pyrazoiyl, 1 ,2,3-triazolyS, 1 ,2,4-triazo!yl, 1 ,2,3-oxadiazolyl, 1 ,2,4-oxadia-zolyl, 1 ,2,5-oxadiazolyi, 1 ,3,4-oxadiazolyl,l ,3,4- triazinyl, 1 ,2,3-triazinyi, benzofuryl, [2,3- dihydrojbenzofuryl, isobenzofuryl, benzothienyl, benzotriazoiyl, isobenzothienyl, indolyl, isoindolyl, 3H-indoIyi, benzimidazoiyi, imidazopyridmyl, benzothiazolyl, benzoxazolyl , quinol izinyl, qiiinazolinyl, pthaiazinyl, quinoxalinyi, cinnolinyl, napthyridinyl, pyrido[3,4- bjpyridyl, pyrido[3,2-b]pyridyl, pyrido[4,3~b]pyridyl, quinolyl, isoquinolyl, tetrazolyl, 5,6,7,8- tetrahydroquinolyl, 5,6,7,8-tetrahydroisoquinoiyl, purinyl, pteridinyl, carbazolyl, xanthenyl,beiizoquiiiolyl, imidazopyrimidinyl,

imidazopyridazinyl, imidazothiazolyl or imidazothiadiazolyl.

"C2-6 alkenyl" refers to alkenyl groups preferably having from 2 to 6 carbon atoms and having at least 1 or 2 sites of alkenyl unsaturation. Preferable alkenyl groups include ethenyi (vinyl, -CH=CH2), n-2-propenyl (allyl, -CH2CH=CH2) and the like.

"C2-6 afkynyl" refers to alkynyl groups preferably having from 2 to 6 carbon atoms and having at least 1-2 sites of alkynyl unsaturation, preferred alkynyl groups include ethynyl (-C≡=CH), propargyl (-(. ' H2C * i l l ), and the like.

"C3.8 cycloaikyi" refers to a saturated carbocyelic group of from 3 to 8 carbon atoms having a single ring (e.g., cyclohexyl) or multiple condensed rings (e.g., norbomyl). Preferred cycloaikyi include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl and the like.

"Heterocycloalkyl" refers to a C3.8 cycloaikyi group according to die definition above, in which 1 to 3 carbon atoms are replaced by hetero atoms chosen from the group consisting of O, S, NR, R being defined as hydrogen or C-j.g alkyl.

"Alkoxy" refers to the group -O-R where R includes " C- g alky]", "C2-6 alkenyl", "C2-6 alkynyl", "C3.8 cycloaikyi", "heterocycloalkyl", "aiyl", "heteroar}'i".

"Amino" refers to the group -NRR' where each R, R' is independently hydrogen, "C-j.g alkyl", "C2-6 alkenyl", "C2-6 alkynyl", "C3-8 cycloaikyi",

"heterocycloalkyl", "aryl", "heteroaryl", and where R and R', together with the nitrogen atom to which they are attached, can optionally form a 3-8-membered heterocycloalkyl ring.

"Arnido" refers to the group -C(=0)NRR' where each R, R' is independently hydrogen, " C-|__5 alkyl", " C2-6 alkenyi", " C2-6 alkynyl", " C3.8 cycloalkyl", "heterocycloalkyl", "aryl",

"heteroaryl", and where R and R', together with the nitrogen atom to which they are attached, can optionally form a 3-8-membered heterocycloalkyl ring.

"AeySamino" refers to the group -NRC(0)R' wherein R and R' are as defined hereabove for the amino group.

"Ureido" refers to the group -NR"C(0)NRR' wherein R and R' are as defined hereabove for the amino group, and R" is as defined hereabove. "Su!fanyl" refers to the group -SR where R is "C-j.g alkyl", "C2-6 alkenyi", "C2-6 alkynyl", "C3.8 cycloalkyl", "heterocycloalkyl", "aryl" or "heteroaryl".

"Sulfinyl" refers to the group -S(=0)R where R is "C-|.g alkyl", "C2-6 alkenyi", "C2-6 alkynyl", "C3.8 cycloalkyl", "heterocycloalkyl", "aryl" or "heteroaryl".

"Suifonyi" refers to the group -S(=0)2R where R is "C-j.g alkyl", "C2-6 alkenyi", "C2-6 alkynyl", "C3.8 cycloalkyl", "heterocycloalkyl", "aryl" or "heteroaryl".

"Halogen" refers to fluoro, chloro, bromo and iodo atoms.

"Substituted or unsubstituted" : Unless otherwise constrained by the definition of the individual substituent, the above set out groups, like "alkyl", "alkenyi", "alkynyl", "aryl" and

"heteroaryl" etc. groups can optionally be substituted with from 1 to 5 substituents selected from the group consisting of "C-j.g alkyl", "C2-6 alkenyi", "C2-6 alkynyl",

"cycloalkyl", "heterocycloalkyl", "amino", "arnido", "acylamino", "ureido", "and", "heteroaryl", "alkoxy", "halogen", cyano, hydroxy, mercapto, nitro, "arnido", "suifanyl", "sulfinyl", "suifonyi" and the like.

The acid addition salt form of a compound of formula ( I) that occurs in its free form as a base can be obtained by treating the free base with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, trifluoroacetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesuifonic, ρ-toluenesulfonic, cyclamic, salicylic, p- aminosalicylic, pamoic and the like.

The compounds of formula (I) containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt forms, e.g. metal or amine salts, by treatment with appropriate organic and inorganic bases, Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e.g. lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.

Conversely said salt forms can be converted into the free forms by treatment with an appropriate base or acid.

Compounds of the formula (I) and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, aicoholates and the like.

Many of the compounds of formula ( ) and some of their intermediates have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem., 45 (1976) 11-30.

The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds of formula (I) or mixtures thereof (including all possible mixtures of stereoisomers). With respect to the present invention reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof, unless the particular isomeric form is referred to specifically.

Compounds according to the present invention may exist in different polymorphic forms. Although not explicitly indicated in the above formula, such forms are intended to be included within the scope of the present invention. Some of the compoimds of formula (I) may also exist in tautomeric forms. Such forms although not explicity indicated in the above formula are intended to be included within the scope of the present invention.

The invention also includes within its scope pro-drug forms of the compounds of formula (I) and its various sub-scopes and sub-groups.

In a specific embodiment, the present invention concerns a compound selected from the group consisting of: (2S)-2-[3~(4-nitrophenyl)-5-oxoirnida.zolidin-l- yljbutanamide; (2S)-2-[3-(2,4-dinitrophenyl)-5-oxoimidazolidm-l-yl]butanami de; (2S)-2-(5-oxo-3- phenylimidazoiidin- 1 -yl)butanamide; 2-[5-(iodomethyi)-2-oxo- 1 ,3-oxazolidin~3~ yijbutanamide; 2-(2-oxo-2,5-dihydro-l H-pyrrol- l-yi)butanamide; 2-(2-oxo-4-phenyl-2,5- dihyd.ro- 1 H-pyrrol- 1 -yl)butanamide; 2-(4-methyl-2-oxo- 2,5-dihydro-l H-pyrrol- 1- yl)butanamide; (+)-(2S)-2-(2-oxo-4-propyl-2,5-dihydro- 1 H-pyrrol- 1 -yl)butanamide; (2S)-2- (2-oxo-5-propyl-l ,3-thiazol-3(2H)- yi)butanamide; 2-( 2-oxo~5 -propyl- 1 ,3-thiazoi-3(2H)- yi)propanamide; 2~(5~butyl~ 2-oxo-l ,3-thiazolidin-3-yl)butanamide; 2-(5-butyl-2-oxo-l ,3- thiazolidin-3- yl)propanamide; 2-(2-oxo~5-phenyl-l ,3-thiazolidm-3-yl)propanamide; 2-(2- oxo- 5 -propyl- 1 ,3-thiazolidm-3-yl)butanamide; 2-(2-oxo-5 -phenyl- 1 ,3-thiazolidin-3- yljbutanamide; 2-(2-oxo-5-propyl- 1 ,3~thiazolidin~3~yl)propana.mide; (2S)-2-[2- oxo-5-f 2,2,2- trifluoroethyl)-! ,3-thiazolidin-3-yl]butanamide; 1.-{[6-chloro-2- (trifluoromethyl)imidazo[l ,2- b]pyridazin-3-yl]methyi}piperidin-2-one; 1 -(1 H- irnidazoi-4-ylmethyl)-5-propylpiperidm-2- one; l-( 1 H-imidazol-1 -ylmethyl)-5- propylpiperidin-2-one; 1 -(imidazo[ 1 ,2-a]pyridin-3- ylmethyl)-5-propylpiperidin- -one; 1-(1 H-imidazol-1 -ylmethyi)-5-phenylpiperidin-2-one; l - (imidazo[l ,2- a] pyridin-3-ylmethy[)-5-phenylpiperidm-2-one; 1 -(imidazo[l ,2-a]pyridin-3- ylmethyl)-4-phenylpiperidin-2-one; 1-(1 H-imidazol-1 -ylmethyl)-4- phenylpiperidin-2-one; 1 - (imidazof 1 ,2-a]pyridin-3-ylmethyl)-4-propylpiperidin- 2-one; 1-(1 H-imidazol-5-ylmethyl)-4- propylpiperidin-2-one; 1-(1 H-imidazol-1- ylmethyl)-4-propylpiperidin-2-one; 1 -{[6-chloro-2- (trifluoromethyl)iniidazo[] ,2- b] pyridazin-3-yl]methyl}azepan-2-one; 1 -(1 H-imidazol-5- yimethyl)-5- propylazepan-2-one; 5-propyl-l- {[2-(trifiuoromethyl)imidazo[l ,2-a]pyridin-3- yljmethyl} azepan-2-one; 1-(1 H-imidazol-5-ylmethyl)-5-phenyiazepan-2-one; 5- phenyl-1 - {[2-(trifluoromethyl)imidazo[l ,2-a]pyridin-3-yl]methyl}azepan-2-one; 1- (1 H-imidazol-5- ylmethyl)-6-propylazepan-2-one; 1-(1 H-imidazol-4-ylmethyl)- 4-propylazepan-2-one; 4- (1 H-imidazol-4-ylmethyi)-4,6-dihydro-5H-thieno[3,2- b]pyrrol-5 -one; 2-(5 -oxo-5 ,6-dihydro- 4H-thieno [3 ,2-b]pyrrol-4-yl)acetamide: 4- {[2-(trifluoromethyl)imidazo[l ,2-a]pyridin-3- yl]methyl}-4,6-dihydro-5H- thieno[3,2-b]pyrrol-5-one; 4-{[2-(trifluoromethy[)imidazo[l ,2- a]pyridin-3- yl]methyl}hexahydro-5H-thieno[3,2-b]pyrrol-5-one; I -(1 H-imidazol-4-ylrn.ethyl)- 1 H-thieno[3,4-b]pyrrol-2(3H)-one; 2-(6-bromo-2-oxo- 1 ,3-benzothiazol-3(2H)- yljacetamide; 2-(2-OXO- 1 ,3-berizothiazol-3(2H)-yl)acetamide; 2-(6-chloro-2- oxo-1 ,3-benzothiazol-3(2H)- yi)acetamide; 6~bromo-3-(] H-imidazol-1 -ylmethy!}- 1 ,3-benzomiazol-2(3H)-one; 6-bromo- 3-(2-oxopropyl)- 1 ,3-benzothiazol-2(3H)- one; 2-(6-nitro-2-oxo-l ,3-benzothiazol-3(2H)- yl)acetamide; 2-(6-bromo-2-oxo-l ,3-benzothiazol-3(2H)-yl)propanamide: 2-(6-bromo-2- oxo-1 ,3-benzothiazol- 3(2H)-yl)propanamide; 2-(6-fluoro-2-oxo- 1 ,3-benzothiazol-3(2H)- yl)acetamide;

2- (6-methyl-2-oxo-l ,3-benzothiazol-3(2H)-yl)acetamide; 6-fluoro-3-(l H- imidazol- 1 -ylmethyl)- ] ,3-benzoxazol-2(3H)-one; H-imidazol-4- ylmethyl)pyrazolo[l ,5- a]pyridin-2(l H)-one; 2-(6-chloro-3-oxo-3,4- dihydroisoquinolin-2(l H)-yl)propanamide; 5~ chloro-2-(l H-imidazol~4- ylmethyl)-! ,4-dihydroisoquinolin-3(2H)-one; 2-(6-chloro-2-oxo- 3,4- dihydroquinolin- 1 (2H)-yl)acetamide; 2-(6-bromo-2-oxo-3 ,4-dihydroquinolin- 1 (2H)~ yl)acetamide; 1-f l H-imidazol-4-ylmethyi)-3,4-dihydroquinolin-2(l H)-one; 2-(6-iodo-2-oxo- 3 ,4-dihydroquinolin- 1 (2H)-yl)acetamide; 2-(6-cya.no-2-oxo-3 ,4- dihydroquinolin- 1 (2HV yl)acetamide; 7-chloro-2-{[2-(trifluoroniethyl)imidazo[l ,2-a]pyridin-3-yl]methyl}-l ,2,4,5- teirahydro-3H-2-benzazepin-3-one; 7-chloro-2- (1 H-irmdazol-4-ylrnethyl)~l ,2,4,5- tetrahydro-3H-2-benzazepin-3-one; 7-chloro- 3-(l H-imidazol-4-ylmethy[)-l ,3,4,5- tetrahydro-2H-3-benzazepin-2-one; and 7- chloro-3-{[2-(trifluoromethyl)imidazo[l ,2- a]pyridm-3~yl]methyl}~l ,3,4,5- tetrahydro-2H-3-benzazepin-2-one.

xvi) UK Patent 1,039,1 13

The new compounds according to the present invention are N-substituted lactams of the general formula:

wherein N is a whole number of from 3 to 5 and R represents a

radical in which m is 0, 1 or 2 and R' is a hydrogen atom or an alkyl, cycloaikyi, alkenvl or alkynyl radical, which may contain 3 to 6 carbon atoms, or an aryl radical, and R" is a hydrogen atom or an alkyl radical, or both R' and R", together with the nitrogen atom to which they are attached, form a heterocyclic ring, such as 5 a pyrrolidine ring.

xvii) UK Patent 1,309,692

According to the present invention, there are provided new N-substituted lactams of the general formula:

wherein X is a hydrogen atom or an alkyl, alkenvl or alkynyl radical containing 1 to 6 carbon atoms, p is a whole number of from 1 to 6, Y is a hydrogen atom or an alkyl, alkenyl or alkynyl radical containing 1 to 6 carbon atoms or a cycloaikyi radical and R' and R", which may be the same or different, are hydrogen atoms or alkyl, alkenyl, alkynyl, cycloalkvl or aryl radicals or R' and R", together with the nitrogen atom to which they are attached, form a heterocyclic radical which may contain further heteroatoms. with the proviso that at least one of the symbols X and Y is other than a hydrogen atom.

V :e [0144] In some embodiments, an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered in combination with valproate or its analog, derivative or pharmaceutically acceptable salt.

[0145] Analogs and derivatives of valproate useful for the methods and compositions of this invention include compounds of the formula:

wherein, independently for each occurrence:

X is -OH, Ci.to alkoxy, -G-aikali metal, -NCR 1 )?., -SH, or -S-Ci.jo alkyl; R is a straight chain or branched C 1-3 o alkyl; and

R 1 is H, Ci-io alky, C 2 -J O alkenyl, C 2 -jo alkynyl, and, or aralkyl;

provided that R may be unsubstituted or substituted by one or more -OH, Ci-io alkoxy, -N(R 1 ) 2 , -SH, -S-C M O alkyl, or and.

[0146] In other embodiments, analogs and derivatives of valproate useful for the methods and compositions of this invention include compounds of the formula:

wherein, independently for each occurrence:

X is -OH, d-io alkoxy, -O-alkali metal, -N(R 1 ) 2 , -SH, or -S-Ci-io alkyl;

R is CH[(CH 2 ) 2 CH 3 ] 2 ; and

R 3 is H, CI-J O alky, C 2 -io alkenyl,€ 2 - 1 0 alkynyl, aryi, or aralkyl; provided that R may be unsubstituted or substituted by one or more -OH, Ci-io alkoxy, -N(R ] ) 2 , -SH, -S-Ci-io alkyl, or aryi. [0147] In other embodiments, analogs and derivati ves of valproate useful for the methods and compositions of this invention include compounds of the formula:

wherein, independently for each occurrence:

X is -OH, -O-alkali metal, -8H, or - Nl¾; and

R is CH[(CH 2 ) 2 CH 3 ] 2 .

[0148] Methods for making the compounds of formula may be found in, for example, U.S. Patent Nos.: 4,558,070; 4,595,695; 4,654,370; 4,895,873;

4,913,906; 5,017,613; 5,019,398; 5,049,586; 5,162,573; 5,440,023; 5,856,569;

6,131,106 and 6,610,326.

[0149] Other names and descriptions of valproate are also envisioned herein, such as Depakote, Valreiease, 2-propylpentanoate, valproic acid, VPA and sodium valproate.

Methods of Treating CNS Disorders with Cognitive Impairment with the

Administration of an SV2A Inhibitor

[0150] In one aspect, the invention provides methods and compositions for treating or improving cognitive function, delaying or slowing the progression of cognitive impairment, or reducing the rate of decline of cognitive function, in a subject suffering from a centra! nervous system (CNS) disorder with cognitive impairment (e.g., age- related cognitive impairment, MCI, amnestic MCI, dementia, AD, prodromal AD, PTSD, schizophrenia, ALS and cancer therapy-related cognitive impairment), or the risk thereof in a subject in need thereof by administering an SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. In some embodiments, the SV2A inhibitor is administered in combination with valproate or an analog, derivative or pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. In some embodiments, the SV2A inhibitor is selected from the group consisting of levetiracetam, seletracetam, and brivaracetam or derivatives or analogs or pharmaceutically acceptable salts, or solvates, or hydrates, or polymorphs, or prodrugs thereof. In other embodiments, the SV2A inhibitor is levetiracetam or a derivative or an analog or a pharmaceutically acceptable salt, or a solvate, or a hydrate, or a polymorph, or a prodrug thereof. In other embodiments, the SV2A inhibitor is brivaracetam or a derivative or an analog or a pharmaceutically acceptable salt, or a solvate, or a hydrate, or a polymorph, or a prodrug thereof. In other embodiments, the SV2A inhibitor is seletracetam or a derivative or an analog or a pharmaceutically acceptable salt, or a solvate, or a hydrate, or a polymorph, or a prodrug thereof. In some embodiments, the CNS disorder with cognitive impairment is age-related cognitive impairment, such as Mild Cognitive Impairment (MCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD). In one embodiment of the invention, the MCI is amnestic MCI. In some embodiments of the invention, the CNS disorder with cognitive impairment is dementia, post traumatic stress disorder (PTSD), schizophrenia, amyotrophic lateral sclerosis (ALS) or cancer- therapy-related cognitive impairment. In one embodiment, the subject that suffers such cognitive impairment is a human patient. The subject may be a human or other mammal such as a non-human primate, or rodent (e.g., rat). In some embodiments, the subject is a human patient.

[0151] The use of the SV2A inhibitors and its pharmaceutically acceptable salt, hydrate, sol vate or polymorph in combination with valproate or its analog,

derivative or pharmaceutically acceptable salt reduces the amount of valproate necessary for the treatment of CNS disorders involving cognitive dysfunction and other affective disorders, including MCI, amnestic MCI, AAMI, ARCE, dementia, AD, PTSD, schizophrenia, ALS or cancer-therapy-reiated cognitive impairment.

In one embodiment, the subject that suffers such cognitive impairment is a human patient, and thus reduce the side effects caused by valproate without diminishing efficacy. Further, the efficacy of a combination of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph and valproate or its analog, derivative or pharmaceutically acceptable salt exceeds the efficacy of either drug administered alone at its optimal dose and thus is an improved

treatment for CNS disorders with cognitive impairment. [0152] It will be appreciated that compounds and agents used in the compositions and methods of this in vention preferably should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier, however, can still be effectively administered directly into the central nervous system, e.g., by an intraventricular or other neuro-compatible route.

[0153] As used herein, administration of an SV2A inhibitor or its

pharmaceutically acceptable salt, hydrate, solvate or polymorph and valproate or its analog, derivative or pharmaceutically acceptable salt "in combination" includes simultaneous administration and/or administration at different times, such as sequential administration. Simultaneous administration of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph and valproate or its analog, derivative or pharmaceutically acceptable salt can optionally be combined with supplemental doses of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph and/or valproate or its analog, derivative or pharmaceutically acceptable salt. Simultaneous administration of dmgs encompasses administration as co-formulation or, alternatively, as separate compositions.

[0154] In accordance with this invention, the SV2A inhibitor or its

pharmaceutically acceptable salt, hydrate, solvate or polymorph, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt can be administered to a subject via any suitable route or routes. In some embodiments, the dmgs are administered orally; however, administration intravenously, subcutaneously, intra-arterially, intramuscularly, intraspinal ly, rectally, intrathoracically, intraperitoneally, intracentricularly, or transdermally, topically, or by inhalation is also contemplated. The agents can be administered orally, for example, in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, or the like, prepared by art recognized procedures. In certain embodiments, the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate and polymorps, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt, can be administered to a subject via different routes. For example, the SV2A inhibitor or its salt, solvate, hydrate, or polymorph is administered intravenously and the valproate or an aialog, derivative or pharmaceu tically acceptable salt, hydrate, solvate or polymorph thereof is administered orally. [0155] In some embodiments, the administration is a slow or extended release. The term "extended release" is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a controlled release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an

extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term "extended release" used herein includes the terms "controlled release," "prolonged release," "sustained release," "delayed release," or "slow release" as these terms are used in the

pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump,

[0156] Dosage schedules of the agents and compositions according to the methods of the invention will vary according to the particular compound or compositions selected, the route of adminis tration, the nature of the condition being treated, the age, and condition of the patient, the course, or stage of treatment, and will ultimately be at the discretion of the a ttending physician. I t will be understood that the amount of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph, alone or in combination with valproate or its analog, derivative or phaniiaceutically acceptable salt that is administered will be amounts effective to produce a desired biological effect, such as beneficial results, including clinical results, e.g., an amount that normalizes neural activity in areas of the brain that exhibit abberent activity (including, but not limited to DG, CA3 and/or entorhinal cortex) and/or results in an improvement in cognitive fimction). It will be understood that an effective amount can be administered in more than one dose and over a course of treatment. [0157 J If administered by an implant, a device or a slow or extended release formulation, the SV2A inhibitor or its phaniiaceutically acceptable salt, hydrate, solvate or polymorph, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt can be administered one time, or one or more times periodically throughout the lifetime of the patient as necessary. Other administration intervals intermediate to or shoxter than these dosage intervals for clinical applications may also be used and may be determined by one skilled in the art following the methods of this invention,

[0158] Desired duration of administration of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt can be determined by routine experimentation by one skilled in the art. For example, the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt may be administered for a period of 1 -4 weeks, 1-3 months, 3-6 months, 6-12 months, 1 -2 years, or more, up to the lifetime of the patient,

[0159] It is known in the art that normalization to body surface area is an appropriate method for extrapolating doses between species. The human equivalent dose (HED) for this dosage can be estimated using the following formula that accounts for differences in body surface area (see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologies Evaluation and Research):

HED = animal dose X (Km animal / Km human) where the Km factor is body weight divided by body surface area (Km rat has been determined as 6, and Km human is 37; see Reagan-Saw, Nihal, Ahmad, 2007). Thus, a dosage of 10 mg/kg in rats is equivalent to 1.6 mg kg in humans (10 mg/kg X (6 / 37) = 1.6 mg/kg), For human subjects, to calculate a dose in mg from the dose in mg/kg, the dose in mg/kg is multiplied bya typical adult weight of 70 kg. [0160] In certain embodiments of the invention, the dose of the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is 0.1 to 5 mg/kg/day (which, given a typical human subject of 70 kg, is 7 to 350 mg/day).

[0161] In certain embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof can be administered at doses according to, for example, U.S. Patent Application

12/580,464, International Patent Application PCT/US2009/005647, U.S. Patent Application 61/105,847, U.S. Patent Application 61/152,631, U.S. Patent

Application 61/175,536 and U.S. Patent Application 61/441,251. In certain embodiments of the in vention, the S V2A inhibitor or a pharmaceutical ly acceptable salt, hydra te, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.001 to 5 mg/kg, about 0.001 to 0.5 mg kg. about 0.01 to 0.5 mg/kg, about 0.1 to 5 mg/kg, or about 1 to 2 mg/kg, or about 2 to 4 mg/kg, or about 2 to 3 mg/kg, or about 3 to 4 mg/kg, or about 0.2 to 0.4 mg kg, or about 0.2 to 0.3 mg/kg, or about 0.3 to 0.4 mg/kg, or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg kg, or about 0.1 to 2.5 mg kg, or about 0.4 to 2.5 mg kg, or about 0.6 to 1 ,8 mg/kg, or about 0.5 to 2 mg/kg, or about 0.8 to 1.6, or about 0.8 to 3.6, or about 0.5 to 4 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1 ,8 mg/kg, or about 0.05 to 3 mg/kg or about 0.08 to about 1 .6 mg/kg, or about 0.08 to 3.6 or about 0.05 to 2 mg/kg, or about 0.01 to 1 mg/kg, or about 0.001 to 1 mg/kg, or about 0.5 to 5 mg/kg, or about 0.05 to 0.5 mg/kg, or about 0.8 mg/kg, or about 1.6 mg/kg, or about 3.6 mg/kg, or about 0.08 mg/kg, or about 0.16 mg/kg, or about 0.36 mg/kg. Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention, For repeated administrations over several days or weeks or longer, depending on the condition, the treatment is sustained until a sufficient level of cognitive function is achieved.

[0162] In certain embodiments of the invention, the dose of the SV2A inhibitor is

0.001 - 5 mg/kg/day (which, given a typical human subject of 70 kg, is about 0.07 - 350 mg day). Doses that may be used include, but are not limited to

0.001 mg/kg/day, 0.001 mg/kg/day, 0.002 mg/kg/day, 0.005 mg/kg/day, 0.0075 mg kg day, 0,01 mg/kg/day. 0.015 mg kg day, 0.02 mg/kg/day, 0.03 mg/kg/day, 0.04 mg/kg/day, 0.05 mg/kg/day, 0.1 mg/kg/day, 0.2 mg/kg/day, 0.3 mg/kg/day, 0.4 mg/kg/day, 0.5 mg/kg/day, 0.75 mg/kg/day, 1.0 mg/kg/day, 1.5 mg/kg/day, 2.0 mg/kg/day, 2.5 mg/kg/day, 3.0 mg/kg/day, 4.0 mg/kg/day,or 5.0 mg/kg/day. In some embodiments, the dose of the SV2A inhibitor is 0,001 - 0.5 mg/kg/day

(which, given a typical human subject of 70 kg, is about 0,07 - 35 mg/day), or 0.01

- 0.5 mg/kg/day (which is about 0.7 - 35 mg/day). Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art follo wing the methods of this in vention. [0163 J In certain embodiments of the in vention, the dose of the SV2A inhibitor is 0.1 to 5 mg/kg/day (which, given a typical human subject of 70 kg, is 7 to 350 mg/day). Doses that may be used include, but are not limited to 0.1 mg/kg/day, 0.5 mg/kg/day, 1 mg/kg/day, 1.5 mg/kg/day, 2 mg/kg/day, 2.5 mg/kg/day, 3 mg/kg/day, 4 mg/kg/day, or 5 mg/kg/day. In certain embodiments, the dose is 1-2 mg/kg/day (which, given a typical human subject of 70 kg, is 70-140 mg/day). in other embodiments of the invention, the dose of the SV2A inhibitor is 0.1 to 0.2 mg/kg/day. Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this in vention. [0164J in certain embodiments of the in vention, the dose of the SV2A inhibitor is 0.01 to 2.5 mg/kg/day (which, given a typical human subject of 70 kg, is about 0.7

- 1 80 mg/day). Doses that may be used include, but are not limited to 0.01 mg/kg/day, 0.02 mg/kg/day, 0.03 mg/kg/day, 0.04 mg/kg/day, 0.06 mg/kg/day, 0.08 mg/kg/day, 0, 12 mg/kg/day, 0.14 mg/kg/day, 0.16 mg/kg/day, 0, 1 8 mg/kg/day, 0,2 mg/kg/day, 0.4 mg/kg/day, 0.6 mg/kg/day, 0.8 mg/kg/day, 1.0 mg/kg/day, 1 ,2 mg/kg/day, 1.4 mg/kg/day, 1.6 mg/kg/day, 1 ,8 mg/kg/day, 2,0 mg/kg/day, 2.2 mg/kg/day, 2.4 mg/kg/day, or 2.5 mg/kg/day. In some

embodiments, the dose of the SV2A inhibitor is 0.1 - 2.5 mg/kg/day (which, given a typical human subject of 70 kg, is about 7 - 180 mg/day), 0.1 - 0.2 mg/kg/day (which is about 7 - 15 mg/day), 0.2 - 0.4 mg/kg/day (about 14 - 30 mg/day), 0.4 - 2.5 mg/kg/day (about 25 - 1 80 mg/day), 0.6 - 1 .8 mg/kg/day (about 40 - 130 nig/day), 0,04 - 2.5 mg kg day (about 2.5 - 180 mg/ ' day) or 0.06 - 1.8 mg kg/day (about 4 - 130 mg/day). In some embodiments of the invention, the dose of the SV2A inhibitor is 40 to 130 mg, 140 to 300 nig, 200 to 300 nig or 140 to 200 mg. Other doses higher than, intermediate to, or less than these doses may also be used and may be determined by one skilled in the art following the methods of this invention.

[0165] In certain embodiments of the invention, the interval of administration is 12 or 24 hours. Administration at less frequent intervals, such as once ever} ' 6 hours, may also be used. In some embodiments, the SV2A inhibitor is administered ever} ' 12 or 24 hours at a total daily dose of 0.1 to 5 mg/kg (e.g., in the case of administration every 12 hours of a daily dose of 2 mg/kg, each administration is 1 mg/kg). In some embodiments, the SV2A inhibitor is administered every 24 hours at a daily dose of 1 to 2 mg/kg. In another embodiment, the SV2A inhibitor is administered every 24 hours at a daily dose of 0.1 - 0.2 mg/kg. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0,01 to 2,5 mg/kg (e.g., in the case of administration every 12 hours of a daily dose of 0.8 mg/kg, each administration is 0.4 mg/kg). In some embodiments, the SV2A inhibitor is administered ever}' 12 or 24 hours at a daily dose of 0.1 to 2.5 mg/kg. In some embodiments, the SV2A inhibitor is administered ever}' 12 or 24 hours at a daily dose of 0.4 to 2.5 mg/kg. In some embodiments, the SV2A inhibitor is administered every 12 or 24 hours at a daily dose of 0.6 to 1.8 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.04 - 2.5 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.06 -1.8 mg/kg, In some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.001 -5 mg/kg. In some embodiments, the selective inhibitor of SV2A is administered ever}' 12 or 24 hours at a daily dose of 0.001 - 0.5 mg kg. in some embodiments, the selective inhibitor of SV2A is administered every 12 or 24 hours at a daily dose of 0.01 - 0.5 mg/kg. [0166] In certain embodiments of the invention, the SV2A inhibitor is

Ievetiracetam or a pharmaceutically acceptable salt, hydrate, sol vate or polymorph thereof. The Ievetiracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered ever}' 12 or 24 hours at a daily dose of about 1 to 2 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 2.0 to 3.0 mg/kg, or about 3.0 to 4,0 mg/kg, or about 2.0 to 4,0 mg/kg, or about 0, 1 to 5 mg/kg, or about 70 to 140 mg, or about 7 to 180 mg, or about 25 to 180 mg, or about 40 to 1 30 mg, or about 140 to 300 mg, or about 200 to 300 mg, or about 140 to 200 mg, or about 7 to 350 mg.

[0167 J in other embodiments, the ievetiracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours according to one of the daily dose ranges indicated as "+" listed in Table 1 or Table 2.

Table 1 - Dally Doses of Levetiracetam

Table 2 - Daily Doses of Leveiiraeetam in a Human Subject of 70 KG

[0168] In certain embodiments of the invention, the SV2A inhibitor is brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof. The brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of about 0.1 to 0,2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.2 to 0.4 mg/kg, or about 7 to 15 mg, or about 0.7 to 180 mg, or about 2.5 to 180 mg, or about 4.0 to 130 mg, or about 14 to 30 mg. [0169] In other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours at a daily dose of at least 0, 1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 mg, but no more than a daily dose of 2.5 mg, 5 mg, 10 mg, 1 mg, 20 mg, 25 mg, 30 mg, or 35 mg. In other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg, but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.15 mg/kg, 0.1 mg kg, 0.05 mg/kg, or 0.04 mg/kg. [0170 J in other embodiments, the brivaracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered every 12 or 24 hours according to one of the daily dose ranges indicated as "+" listed in Table 3 or Table 4. For example, the brivaracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be administered every 12 or 24 hours at a daily dose of 0.1 - 35 mg, 0.5 - 35 mg, 0.75 - 35 mg, 1 .0 - 35 mg, 1 ,5 - 35 mg, 2,0 - 35 mg, 0.1 - 30 mg, 0, 1 - 25 mg, 0, 1 - 20 mg, 0, 1 - 15 mg, 0, 1 - 1 0 mg, 0.1 - 5 mg, 0.1 - 2.5 mg, 0.0015 - 0.5 mg/kg, 0.0075 - 0,5 mg/kg, 0.01 - 0.5 mg/kg, 0,015 - 0.5 mg/kg, 0.02 - 0.5 mg/kg, 0,03 - 0.5 mg/kg, 0.001 - 0,4 mg/kg, 0.0015 - 0,3 mg/kg, 0.0015 - 0.2 mg/kg, 0.0015 - 0.1 5 mg/kg, 0.0015 - 0.1 mg/kg, 0.001 - 0.05 mg/kg, or 0.0015 - 0.04 mg kg.

- Daily Doses of Brivaracetam

of Brivaracetam

|0171] In certain embodiments of the invention, the SV2A inhibitor is seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof, in some embodiments, the seletracetam or its pharmaceutically acceptable salt, hydrate, solva te or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.1 mg, 0.5 mg, 0.75 mg, 1.0 mg, 1.5 mg, or 2.0 nig, but no more than a daily dose of 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg. or 35 mg. In other embodiments, the seletracetam or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is administered every 12 or 24 hours at a daily dose of at least 0.0015 mg/kg, 0.0075 mg/kg, 0.01 mg/kg, 0.015 mg/kg, 0.02 mg/kg, or 0.03 mg/kg, but no more than a daily dose of 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.15 mg/kg, 0.1 mg/kg, 0.05 mg/kg, or 0.04 mg/kg.

[0172] In certain embodiments of the invention, the seletracetam or its

pharmaceutically acceptable salt, hydrate, solvate or polymorph is administered according to one of the daily dose ranges indicated as "+" listed in Table 5 or Table 6. For example, the seletracetam or its pharmaceutically acceptable salt, hydrate, solvate or polymorph may be administered every 12 or 24 hours at a daily- dose of 0.1 - 35 mg, 0.5 - 35 mg, 0.75 - 35 mg, 1.0 - 35 mg, 1.5 - 35 mg, 2.0 - 35 mg, 0.1 - 30 mg, 0.1 - 25 mg, 0.1 - 20 mg, 0.1 - 15 mg, 0, 1 - 10 mg, 0.1 - 5 mg, 0.1 - 2.5 mg, 0.0015 - 0.5 mg/kg, 0,0075 - 0.5 mg/kg, 0,01 - 0,5 mg/kg, 0.015 - 0.5 mg/kg, 0.02 - 0,5 mg/kg, 0.03 - 0.5 mg/kg, 0,001 - 0.4 mg/kg, 0.0015 - 0.3 mg/kg, 0.001 - 0,2 mg/kg, 0.0015 - 0,1 mg/kg, 0.0015 - 0,1 mg/kg, 0.0015 - 0.05 mg/kg, or 0.0015 - 0.04 mg/kg.

" 7 Table 5 - Daily Doses of Seletracetam

Table 6 - Daily Doses of Seletracetam in a Human Subject of 70 KG

|0173] The SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph may be administered at a subtherapeutic dosage levels when provided in combination with valproate or its analog, derivative or pharmaceutically acceptable salt, due to valproate-dependent increase in the therapeutic index of the SV2A inhibitor. In some embodiments, the increase in the therapeutic index of the SV2A inhibitor, due to the comb illation with valproate, is greater than the therapeutic index of the SV2A inhibitor administered in the absence of the valproate by at least about 1 ,5x or 2. Ox or 2.5x or 3. Ox or 3.5x or 4, Ox or 4.5x or 5. Ox or 5.5x or 6, Ox or 6.5x or 7. Ox or 7.5x or 8. Ox or 8.5 or 9. Ox or 9,5x or lOx, or greater tha about lOx. In some embodiments, combinations of an SV2A inhibitor with valproate reduces the dosage of the SV2A inhibitor required for its therapeutic effect. In some embodiments of the invention, the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is administered in combination with valproate or its analog, derivative or pharmaceutically acceptable salt is administered ever} ' 12 or 24 hours at a daily dose of about 0,001 mg/kg to 5 mg/kg, or about 0.1 to 5 mg/kg, or about 1 to 2 mg kg. or about 0.1 to 0.2 mg/kg, or about 0.01 to 2.5 mg/kg, or about 0.1 to 2.5 mg/kg, or about 0.4 to 2.5 mg/kg, or about 0.6 to 1.8 mg/kg, or about 0.04 to 2.5 mg/kg, or about 0.06 to 1.8 mg/kg, or about 0.01 to 1 mg/kg, or about 0,001 to 1 mg/kg, or about 0.5 mg/kg to 5 mg/kg, or about 0,05 mg/kg to 0.5 mg/kg. In some embodiments, the amount of the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, sol vate or polymorph thereof that is administered in combination with valproate or its analog, derivative or pharmaceutically acceptable salt is a subtherapeutic amount. Such subtherapeutic amount, may be, for example, a daily dose, administered every 12 or 24 hours at a daily dose of less than 5 mg kg, less than 2.5 mg/kg, less than 2 mg/kg, less than 1.75 mg/kg, less than 1 .6 mg/kg, less than 1 .5 mg/kg, less than 1 mg/kg, less than 0.8 mg/kg, less than 0.6 mg kg, less than 0.5 mg/kg, less than 0.4 mg/kg, less than 0,3 mg/kg, less than 0.2 mg/kg, less than 0.1 mg/kg, less than 0,05 mg/kg, less tha 0,04 mg/kg, less than 0,03 mg/kg, less than 0.02 mg/kg, less than 0.01 mg/kg, less than 0.005 mg/kg, or less than 0.001 mg/kg,

[0174] Valproate or its analog, derivative or pharmaceutically acceptable salt may be administered at a dosage level up to conventional dosage levels. Valproate has been prescribed for treatment of epilepsy, bipolar disorder, migraine, and post- traumatic stress disorder. Valproate is also reported to be effective in treating cognitive impairment ( oh et al, 36th annual meeting of the Society for

Neuroscience, October 1 , 2006, No. 273.14, D.3). Chronic subcutaneous administration to memory-impaired aged ra ts of 100 mg kg/day sodium valproate treated their cognitive impairment and their performance in a memory test was significantly improved. This dosage results in a blood total valproate level of 10 .ug/mi plasma (10 ^tg/ml total valproate). Treatment with chronic subcutaneous administration of 50 mg/kg/day valproate, however, w r as not effective.

10175] The valproate or its analog, derivative or pharmaceutically acceptable salt may be administered at dosage levels distinct from conventional levels when provided in combination with an SV2A inhibitor, due to an SV2A inhibitor-dependent increase in the valproate's therapeutic index. In some embodiments, the increase in the valproate's therapeutic inde due to the combination with an S V2A inhibitor thereof is greater than the therapeutic index of the valproate administered in the absence of an S V2A inhibitor by at least about 1.5x or 2. Ox or 2.5x or 3. Ox or 3.5x or 4. Ox or 4.5x or 5. Ox or 5.5x or 6. Ox or 6.5x or 7. Ox or 7.5x or 8. Ox or 8.5x or 9. Ox or 9.5x or 1 Ox, or greater than about lOx. In some embodiments, combina tions of valproate with the SV2A inhibitor reduces the dosage of the valproate required for its therapeuti c effect, in some embodiments, the amount of the valproate or its analog, derivative or pharmaceutically acceptable salt administered in combination with the SV2A inhibitor is a subtherapeutic amount. In certain embodiments, the dose of valproate or its analog, derivative or pharmaceutically acceptable salt when administered in combination with an SV2A inhibitor is a dose that results in a total blood valproate of 0,5 to 5 ^ig/ml plasma. The doses useful for valproate or its analog, derivative or pharmaceutically acceptable salt are readily determined by those skill ed in the art, using the methods of this invention.

[0176] In certain embodiments, wherein a SV2A inhibitor or its

pharmaceutically acceptable salt, hydrate, solvate and polymorph is administered in combination with valproate or its analog, derivative or pharmaceutically

acceptabl e salt, the dosage of bot h the SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate and polymorph and the valproate or its analog, derivative or pharmaceutically acceptable salt, are each sub-therapeutic with respect to treating a CNS disorder with cognitive impairment when administered alone,

[0177] In some embodiments, a suitable amount of the SV2A inhibitor is administered so as to reduce the dose of the valproate (e.g., a dose required to effect a degree of cognitive function improvement or treat age-associated cognitive

impairment) by at least about 20%, at least about 30%, at least about 40%, or at least about 50%, at least about 60%, at least about 70%, at least about 80°/», at least about 90% or more from to the dose of valproate normally used when administered alone

(i.e., individually and not in combination with other therapeutic agents or compounds). The reduction may be reflected in terms of amount administered at a given

administration and/or amount administered over a given period of time (reduced frequency). [0178] In certain embodiments of the in vention, the combined administration of an

SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate and polymorph, and valproate or its analog, derivative or pharmaceutically acceptable salt, can attain a longer or improved therapeutic effect in the subject than that attained by administering only the valproate or only the SV2A inhibitor, by at least about 1.5x, or 2. Ox, or 2.5x, or 3. Ox, or 3.5x, or 4. Ox, or 4.5x, or 5. Ox, or 5.5x, or 6. Ox, or 6.5x, or 7. Ox, or 7.5x, or 8. Ox, or 8.5x, or 9. Ox, or 9.5x, or lOx, or greater than about lOx.

Compositions of the Isivesiiioii

[0179] In one aspect, the invention provides compositions comprising an SV2A inhibitor or the pharmaceutically acceptable salt, hydrate, solvate or polymorph alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt. In some embodiments, the SV2A inhibitor and the valproate may be present in a single dosage unit (e.g., combined together in one capsule, tablet, powder, or liquid, etc.). The composition described herein can contain more than one SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph and/or more than one valproate or its analog, derivative or pharmaceutically acceptable salt. In some embodiments, the SV2A inhibitor and the valproate are in separate

formulations packaged together.

[0180] The compositions described herein can further contain pharmaceutically acceptable excipient(s) and may contain other agents that serve to enhance and/or complement the effectiveness of the SV2A inhibitor and/or the valproate. The compositions may also contain additional agents known to be useful for treating cognitive function disorder.

10181] The composition in the present in vention may be in solid dosage forms such as capsules, tablets, dragrees, pills, lozenges, powders and granule. Where appropriate, they may be prepared with coatings such as enteric coatings or they may be formulated so as to provide controlled releases of one or more active ingredient such as sustained or prolonged release according to methods well known in the art, In certain

embodiments, the composition is in form of a slow, controlled, or extended release. The term "extended release" is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a control led release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term "extended release" used herein includes the terms "controlled release", "prolonged release", "sustained release", or "slow release", as these terms are used in the pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump. The composition may also be in liquid dosage forms including solutions, emulsions, suspensions, syrups, and elixirs. [0182J The compositions may be specifically formulated for administration by any suitable route as described herein and known in the art. Compositions for parental administration include sterile aqueous and nonaqueous injectable solutions, dispersions, suspensions or emulsions as well as sterile powders to be reconstituted in sterile injectable solutions or dispersions prior to use. Compositions for intraoral and oral delivery (including sublingual and buccal administration, e.g. Danckwerts et al, and oral) include but are not limited to bioadhesive polymers, tablets, patches, liquids and semisolids (see e.g., Smart et al). Compositions for respiratory deliver}' (pulmonary and nasal delivery) include but are not limited to a variety of pressurized metered dose inhalers, dry powder inhalers, nebulizers, aqueous mist inhalers, drops, solutions, suspensions, sprays, powders, gels, ointments, and specialized systems such as liposomes and microspheres (see e.g. Owens et al, "Alternative Routes of Insulin Deliveiy" and Martini et al). Compositions for transdermal delivery include but are not limited to colloids, patches, and microemulsions. Other suitable administration forms for the above and other include depot injectable formulations, suppositories, sprays, ointments, cremes, gels, inhalants, dermal patches, implants etc. [0183] The compositions may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions, in addition, prolonged absorption of the in jectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.

|0184] Therapeutic formulations can be prepared by methods well known in the art of pharmacy, see, e.g., Goodman et al., 2001; Ansel, et al, 2004; Stoklosa et al., 2001; and Bustamante, et al., 1993.

[0185] In certain embodiments of the invention, a composition containing an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph comprises the 8V2A inhibitor in an amount of 0.07 - 60 mg, 0.07 - 350 mg, 25 - 60 mg, 25 - 125 mg, 50 - 250 mg, 5 - 140 mg, 0.7 - 1 80 mg, 125 - 240 mg, 3 - 50 mg, or 3 - 60 mg. In some embodiments, a composition containing an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph comprises the SV2A inhibitor in an amount of 0.05 - 35 mg.

[0186] In certain embodiments of the invention, a composition containing an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph, in combination with valproate or its analog, derivative or pharmaceutically acceptable salt comprises an amount of the 8V2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate or polymorph of 0.05 - 35 mg, 0.07 - 60 mg, 0.07 - 350 mg, 25 - 60 mg, 25 - 125 mg, 50 - 250 mg, 5 - 15 mg, 5 - 30 mg, 5 - 140 mg, 0.7 - 180 mg, 125 - 240 mg, 3 -50 mg, or 0.07 - 50 mg, or 3 - 60 mg. In some embodiments, the amount of the SV2A inhibitor or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof is less than 350 mg, less than 250 mg, less than 200 mg, less than 150 mg, less than 100 mg, less than 50 mg, less than 35 mg, less than 10 mg, less than 5 mg, less than 1 mg, less than 0.5 mg, less than 0.1 mg, less than 0.07 mg, or less than 0.05 mg.

[0187] In addition to an SV2A inhibitor or its pharmaceutically acceptable salt, hydrate, solvate and polymorph, alone or in combination with valproate or its analog, derivative or pharmaceutically acceptable salt, the compositions and methods of this invention can also include other therapeutically useful agents.

These other therapeutically useful agents may be administered in a single

formulation, simultaneously or sequentially according to the methods of the invention.

[0188] It will be understood by one of ordinary skill in the art that the

compositions and methods described herein may be adapted and modified as is appropriate for the application being addressed and that the compositions and methods described herein may be employed in other suitable applications, and that such other additions and modifications will not depart from the scope hereof,

[0189] This invention will be better understood from the Experimental Details which follow. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the invention as described more fully in the embodiments which follow thereafter.

[0190] Introduction and Models of Cognitive Impairment

[0191] A variety of conditions characterized by cognitive impairment, e.g., Age- Associated Memory Impairment (AAMI), Mild Cognitive Impairment (MCI) and Age-related Cognitive Decline (ARCD) are believed to be related to aging. Others are related to disease, for example, AD. Animal models serve as an important resource for developing and evaluating treatments for such age-related cognitive impairments. Features tha characterize age-related cognitive impairment in animal models typically extend to age-related cognitive impairment in humans. Efficacy in such animal models is, thus, predictive of efficacy in humans.

[0192] Of available models, a Long-Evans rat model of cognitive impairment is particularly well suited for distinguishing the difference between cognitive impairment related to illness and that related to aging. Indeed, extensive behavioral characterization has identified a naturally occurring form of cognitive impairment in an outbred strain of aged Long-E vans rats (Charles River

Laboratories; Gallagher et al., Behav. Neurosci. 107:618-626, (1993)). In a behavioral assessment with the Morris Water Maze (MWM), rats learn and remember the location of an escape platform guided by a configuration of spatial cues surrounding the maze. The cognitive basis of performance is tested in probe trials using measures of the animal's spatial bias in searching for the location of the escape platform. Aged rats in the study population have no difficulty swimming to a visible platform, but an age-dependent impairment is detected when the platform is camouflaged, requiring the use of spatial information. Performance for individual aged rats in the outbred Long-Evans strain varies greatly. For example, a proportion of those rats perform on a par with young adults. However, approximately 40-50% fall outside the range of young performance, This variability among aged rats reflects reliable individual differences. Thus, within the aged population some animals are cognitively impaired and designated aged- impaired (AI) and other animals are not impaired and are designated aged- unimpaired (AU). See, e.g., Colombo et al, Proc. Natl. Acad. Set 94: 14195- 14199, (1997); Gallagher and Burweil, Neurobiol. Aging 10: 691 -708, (1989); Rapp and Gallagher, Proc. Natl Acad. Sci. 93: 9926-9930, (1996); Nicolle et al, Neuroscience 74: 741 -756, (1996); and Nicolle et al, J. Neurosci 19: 9604-9610, ( 1999).

[01 3] We used the above-described rat model to identify individual AI and AU rats. We then conducted behavioral assessment on AI rats while administering various pharmacological treatments. | 194] Example 1 : Increased Gene Expression of SV2A In Aged-Impaired Rats Behavioral Characterization of Young, Aged-Impaired and Aged- Unimpaired Rats in Morris Water Maze (MWM)

|01 5] Behavioral tests were performed on young (4 months old) and aged (24 months old) pathogen-free male Long-Evans rats. [0196] The MWM apparatus consists of a large, circular pool (diameter 1.83 m; height, 0.58 m) filled with water (27°C) that is made opaque throug the addition of non-toxic pigment or some other substance. In the typical "hidden platform" version of the test, rats are trained to find a camouflaged white escape platform (height, 34.5 cm) that is positioned in the center of one quadrant of the maze about 1.0 cm below the water surface. This platform can be retracted to the bottom of the tank or raised to its normal position from outside the maze during behavioral testing. The location of the pla tform remains constant from trial to trial. Because there are no local cues that mark the position of the platform, the rat's ability to locate it efficiently from any starting position at the perimeter of the pool depends on using information surrounding the maze. The maze is surrounded by black curtains to which white patterns are affixed to provide a configuration of spatial cues. A second platform (height 37.5 cm), with its surface painted black is elevated 2 cm above the water surface during cue training to control for factors unrelated to cognition. The behavior of a rat in the pool is recorded by a camera that is suspended 2.5 m abo ve the center of the pool . The camera is connected to a video tracking system (HVS Image Advanced Tracker VP200) and a PC computer running MVS software developed by Richard Baker of HVS Image, Hampton, UK.

|0197] The MWM protocol is optimized for sensitivity to the effects of aging on cognition and for measures of reliable individual differences within the aged population of out-bred Long-Evans rats (Gallagher et al. Behav. Neurosci,

107:618-626, (1993)). Rats receive three trials per day for 8 consecutive days, using a 60 sec inter-trial interval. On each training trial, the rat is released into the maze from one of four equally spaced starting positions around the perimeter of the pool. The starting position varies from trial to trial, thus preventing the use of a response strategy (e.g., always turning left from the start location to locate the escape platform). If a rat does not locate the escape platform within 90 sec on any trial, the experimenter guides the rat to the platform, where it remains for 30 sec. Every sixth trial consists of a probe trial to assess the development of spatial bias in the maze. During these trials, the rat swims with the platform retracted to the bottom of the pool for 30 sec, at which time the platform is raised to its normal position for completion of the escape trial . At the completion of the protocol using the hidden platform, rats are assessed for cue learning using the visible platform, The l ocation of this platform varies from trial to trial in a single session of 6 training trials,

[0198] The proximity of the anima s position with respect to the goal is used to analyze the training trial and probe trial performance. The proximity measure is obtained by sampling the position of the animal in the maze (lOtimes/sec) to provide a record of distance from the escape platform in 1 sec averages. For both probe trials and training trials, a correction procedure is implemented so that trial performance is relatively unbiased by differences in distance to the goal from the various start locations at the perimeter of the pool. In making this correction, the average swimming speed is calculated for each trial (path length/latency). Then, the amount of time required to swim to the goal at that speed from the start location used for the trial is removed from the record prior to computing trial performance, i.e., cumulative distance on training trials and average distance from the goal on probe trials. Thus, scores obtained using the proximity measure are designed to reflect search error, representing deviations from an optimal search, i.e. direct path to the goal and search in the immediate vicinity of that location during probe trials.

[0199] Computer records of video-tracking are compiled to provide data on each rat's performance in the maze. Measures on training trials and probe trials are analyzed by Analysis of Variance (ANOVA),

[0200] In one set of trials, the performance during training with the hidden, camouflaged platform differs between the groups of young and aged rats [F (1, 23) =12.69, p<0,002], In this set of trials, no difference between the groups is observed for the cue training trials with a visible platform. In this set of trials, latencies to escape during cue training averaged 9.36 seconds for young and 10,60 seconds for the aged rats.

10201] An average proximity measure on interpolated probe trials is used to calculate a spatial learning index for each individual subject as described in detail in Gallagher et a!,, Behav, Neurosci. 107:618-26, (1993). When a rat rapidly learns to search for the platform close to its position, its spatial learning index is low. Overall, in one set of trials aged rats differed from young rats [F (1, 23) ::: T5,18, p<0.001]. Aged rats are classified as either unimpaired or impaired relative to the learning index profile of the young study population. Aged rats that fail within the normative range of young rats (index scores <241 are designated aged-unimpaired (AU). The remaining aged subjects that have index scores outside the range of young performance are designated aged-impaired (AI).

Preparation ofRNA from Behavioraily Characterized Rats

10202] Twenty-four outbred Long-Evans rats, behavioraily characterized as is described above, are killed by live decapitation to obtain fresh brain tissue. The brain is removed, and the dentate gyms hippocampal region is microdissected from 500 micron sections taken through the transverse axis of the entire hippocampal formation (both left and right hippocampi) of 24 characterized rats. There are 8 animals in each group (AI, AU, and Y). 10203] Total RNA is isolated using Trizol reagent (Invitrogen, Carlsbad, CA) according to the standard protocol (homogenization in Trizol reagent followed by chloroform extraction and isopropanol precipitation). Total RNA is further purified using the RNeasy mini kit (Qiagen, Valencia, CA). cRNA probes are then generated from the RNA samples at the Johns Hopkins Microarray Core Facility, generally according to Affymetrix specifications,

10204] Briefly, 5 ug of total RNA is used to synthesize first strand cDNA using oligonucleotide probes with 24 oligo-dT plus T7 promoter as primer (Proligo LLC, Boulder, CA), and the Superscript Choice System (Invitrogen). Following the double stranded cDNA synthesis, the product is purified by phenol-chloroform extraction, and biotmilated anti-sense cRNA is generated through in vitro transcription using the BioArray RNA High Yield Transcript Labeling kit (ENZO Life Sciences Inc., Farniingdale, NY). 15 μ of the biotiniiated cRNA is fragmented at 94°C for 35 min (lOOmM Trix-acetate, pH 8.2, 500mM OAC, 150mM MgOAC). 10 ^ig of total fragmented cRNA is hybridized to the RAT genome 230-2 Affymetrix GeneChip array for 16 hours at 45°C with constant rotation (60 rpm).

[0205] Affymetrix Fiuidics Station 450 is then used to wash and stain the chips, removing the non-hybridized target and incubating with a streptavidin- phycoerythrin conjugate to stain the biotiniiated cRNA. The staining is then amplified using goat immunoglobulm-G (IgG) as blocking reagent and biotiniiated anti-streptavidin antibody (goat), followed by a second staining step with a streptavidin-ph coerythrin conjugate .

[0206] For quality control of the total RNA from the samples, the Agilent Bioarialyzer, Lab on a Chip technology, is used to confirm that all the samples had optimal rRNA ratios (1 :2, for 188 and 28S, respectively) and clean run patterns.

[0207] For quality control of the hybridization, chip image, and comparison between chips, the following parameters are considered: Scaling factor: related to the overall intensity of the chip, to confirm the similar signal intensity and staining through out the samples; Background: estimation of unspecific or cross- hybridization; Percentage of present calls: percentage of transcripts that are considered significantly hybridized to the chip (present) by the algorithm;

Glyseraldehyde-3-phosphate dehydrogenase (GAPDH) (375'): representation of the RNA integrity by measuring the ratio of 3 ' to 5 ' regions for the housekeeping gene GAPDH, its presence in the chip and a ratio close to 1 advocates for a good integrity of the target ( sampl e); Spikes (BioB/BioC) to confirm the detection level and sensitivity after hybridization.

Data Analysis of Microarray [0208] Fluorescence is detected using the Affymetrix G3000 GeneArray Scanner and image analysis of each GeneChip is done through the GeneChip Operating System 1.1.1 (GCOS) software from Affymetrix, using the standard default settings. All of the GeneChip arrays use short oligonucleotides for genes in an RNA sample.

[0209] For comparison between different chips, global scaling is used, scaling all probe sets to target intensity (TGT) of 150, Total number of present calls and scaling factors are similar across all chips. Further analysis for presence/ absence and statistical difference is performed on a region by region basis in the following manner. Probe sets are determined to be present in a region if it had a present call in four of eight animals in a single group,

[0210] Probe sets are annotated using the Affymetrix annotation of June 20, 2005, and all probe sets representing a specific gene are identified.

[0211] An ANOVA is conducted on the probe set signal values for all present probe sets by combining two groups of animals and comparing them to the third group. An "AI ANOVA" is performed, where AU group are combined with Young group and compared to AI group.

[0212] Pearsons 's correlations comparing probe set signal values to learning indices were calculated for the aged animals (excluding young) across all present probe sets. As shown in FIG. 1, expression of genes encoding SV2A was significantly increased in aged-impaired (AI) individuals relative to young individuals (Y) and aged-unimpaired individuals (AU) in a set of experiments performed as above. These results show that increased SV2A expression was correlated to the development of age-related cognitive impairment, [0213] Example 2: Effect of Levetiracetam in Aged-Impaired Rats

Morris Water Maze Results

[0214] Six Age-Impaired (AI) Long-Evans rats (as characterized above) were tested for their memory of new spatial information in the MWM, under different drug/control treatment conditions (vehicle control and two different dosage levels of levetiracetam). The MWM protocol was substantially the same as the one described in Example 1. Specifically for this study, a retention trial was performed after the training trials, as described below. [0215] AI rats were given six training trials per training day with a 60-sec inter- trial interval between each training trial for two consecutive days. On each training trial , the rat was released in the maze from one of four equally spaced starting positions around the perimeter of the pool. If the rat did not locate the escape platform within 90 sec on any trial, the experimenter guided the rat to the platform, where it remained for 30 sec. 30 minutes to 1 hour prior to all the training trials on each training day, AI rats were pretreated with one of three drug conditions: 1 ) vehicle control (0.9% saline solution); 2) levetiracetam (5m/kg/day); and 3) levetiracetam (lOmg/kg/ ' day); through intraperitoneal (i.p.) injection. The same six AI rats were used for the entire trials so that each treatment condition was tested on all six rats. Therefore, to counterbalance any potential bias, both the location of the escape platform and the spatial cues surrounding the water maze were different in the three treatment conditions. Therefore, using one set of locations and spatial cues, two rats were treated with saline control solution, two with levetiracetam (5m/kg/day) and two with levetiracetam ( l Omg/kg/'day). Using the second set of locations and spatial cues, the two rats treated with saline control solution in the first test were treated with either levetiracetam (Sm-'lig/day) or levetiracetam (lOmg/kg/day), and the two rats previously treated with

levetiracetam (5m/kg/day) were treated with either saline control solution or levetiracetam (lOmg/kg/day), and the two rats previously treated with

levetiracetam (lOmg/kg/day) were treated with either saline control solution or levetiracetam (5m kg day). Using the last set of locations and spatial cues, the rat groupings were again switched so that each group was treated with a different condition than they had been treated previously.

[0216J After the second training day and completion of the twel ve training trials (over the two days), the rat was returned to its home cage and placed in the animal housing room. After a delay of 24 hours from the last training trial, the rat was given one testing trial (the "retention trial"), which was the same MWM task as the training trials, but with the escape platform removed. f 0217] For the retention trial, the MWM circular pool was divided into 4 quadrants. The particular quadrant where the escape platform was placed in the training trials is referred as "target quadrant". The particular region where the platform was located in the training trials is referred as "target annulus". In the retention trial, the time the AI rats spent swimming in the target quadrant is measured and further plotted as a percentage of total swimming time. FIG. 2 displays the results of one such set of retention trials. The time the AI rats spend in the target annulus is also measured. FIG. 2 displays the results of one such set of retention trials. Time data are collected for all three drug treatment conditions.

[0218] In the retention trial, whose results are depicted in FIG. 2, the time the AI rats spent in the target quadrant was approximately 25%, which is a performance equivalent to them having no memory of the platfrom location. This performance did not significantly improve in the group treated with levetiractam at 5 mg/kg/day. However, the group treated with levetiractam at 10 mg/kg/day demonstrated significantly improved memory as compared to vehicle-treated controls, as indicated by a significant increase in the time spent in the target quadrant to approximately 35% of total swimming time (see FIG. 2). That level of performance is equivalent to young and age-unimpaired rats, indicating that treatment with 10 mg/kg/day levetiractam resulted in a significant recovery of the AI rats' ability to navigate this MWM. The effectivness of the 10 mg/kg/day levetiracetam treatment was also seen in the time spent in the target annulus (see FIG, 2), Radial Arm Maze Results

[0219] The effects of levetiracetam on the spatial memory retention of aged- impaired (AI) rats were assessed in a Radial Arm Maze (RAM) behavioral task using vehicle control and five different dosage levels of levetiracetam (1 ,25 mg/kg/day, 2.5 mg/kg/day, 5 mg/kg/day, 10 mg/kg/day and 20 mg/kg/day). RAM behavioral tasks were preformed on ten AI rats. All six treatment conditions were tested on all ten rats, as described above for the MWM test.

|0220] The RAM apparatus used consisted of eight equidistantly-spaced arms. An elevated maze arm (7 cm width x 75 cm length) projected from each facet of an octagonal center platform (30 cm diameter, 51.5 cm height). Clear side walls on the arms were 10 cm high and were angled at 65° to form a trough, A food well (4 cm diameter, 2 cm deep) was located at the distal end of each arm, Froot Loops 1M (Kellogg Company) were used as rewards. Blocks constructed of PlexigIas f (30 cm height x 12 cm width) could be positioned to prevent entry to any arm.

Numerous extra maze cues surrounding the apparatus were also provided.

[0221 ] The AI rats were initially subjected to a pre -training test (Chappell et al. Neuropharmacology 37: 481-487, 1998). The pre-training test consisted of a habituation phase (4 days), a training phase on the standard win-shift task (18 days) and another training phase (14 days) in which a brief delay was imposed between presentation of a subset of arms designated by the experimenter (e.g., 5 arms available and 3 arms blocked) and completion of the eight-arm win-shift task (i.e., with all eight arms available).

[0222] In the habituation phase, rats were familiarized to the maze for an 8- minute session on four consecutive days. In each of these sessions food rewards were scattered on the RAM, initially on the center platform and arms and then progressively confined to the arms. After this habituation phase, a standard training protocol was used, in which a food pellet was loca ted at the end of each arm. Rats received one trial each day for 18 days. Each daily trial terminated when ail eight food pellets had been obtained or when either 16 choices were made or 15 minutes had elapsed. After completion of this training phase, a second training phase was carried out in which the memory demand was increased by imposing a brief delay during the trial. At the beginning of each trial, three arms of the eight-arm maze were blocked. Rats were allowed to obtain food on the five arms to which access was permitted during this initial 'information phase' of the trial. Rats were then removed from the maze for 60 seconds, during which time the barriers on the maze were removed, thus allowing access to all eight arms. Rats were then placed back onto the center platform and allowed to obtain the remaining food rewards during this 'retention test' phase of the trial. The identity and configuration of the blocked arms varied across trials.

[0223] The number of "errors" the AI rats made during the retention test phase was tracked. An error occurred in the trial if the rats entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if it revisited an arm in the post-delay session that had already been visited.

[0224] After completion of the pre-training test, rats were subjected to trials with more extended delay intervals, i.e., a one-hour delay, between the information phase (presentation with some blocked arms) and the retention test (presentation of all arms). During the delay interval, rats remained off to the side of the maze in the testing room, on carts in their individual home cages. AI rats were pretreated 30 - 40 minutes before daily trials with a one-time shot of the following six conditions: 1) vehicle control (0.9% saline solution); 2) levetiracetam (1.25 mg/kg/day); 3) levetiracetam (2,5 mg/kg/day); 4) levetiracetam (5 mg/kg/day); 5) levetiracetam (10 mg kg/day); 6) levetiracetam (20 mg/kg/day); through intraperitoneal (i.p.) injection. Injections were given every other day with intervening washout days. Each AI rat was treated with all six conditions within 23 days of testing. To counterbalance any potential bias, drug effect was assessed using ascending- descending dose series, i.e., the dose series was given first in an ascending order and then repeated in a descending order. Therefore, each dose had two

determinations.

|0225] Parametric statistics (paired t-tests) was used to compare the retention test performance of the AI rats in the one-hour delay version of the RAM task in the context of different doses of levetiracetam and vehicle control (see FIG. 3). The average numbers of errors that occurred in the trials were also significantly fewer with levetiracetam treatment of 5 mg/kg/day (average no, of errors ± standard error of the mean (SEM) = 0.75 ± 0.32) and 10 mg/kg/day (average no. of errors ± SEM = 0.80 ± 0.27) than using vehicle control (average no. of errors ± SEM = 2.00 ± 0.42). Relative to vehicle control treatment, levetiracetam significantly improved memory performance at 5 mg/kg/day (t(9) = 2.18, p = 0.057) and 10 mg/kg/day (1( 9 ;· - 2.37, p = 0.042).

|0226] The radial ami maze task was also used to evaluate the effect of a combination therapy with Levetiracetam (i.p. administration) and valproate (subcutaneous administration). Levetiracetam, on its own, was effective in reducing the number of errors by Al rats in the radial arm maze at 5-10 mg/kg doses, but not at 1.25 mg/kg or 2,5 mg/kg. Valproate, on its own, was effective at 100 mg/kg but not at 25 mg/kg or 50 mg/kg. See FIG. 4. Combining the two drugs, however, had a synergistic effect, A combined administration of 50 mg/kg valproate with 2.5 mg/kg levetiracetam, neither being an effective dose when administered individually, resulted in a reduced number of errors in the radial arm maze task. This result was also obtained at an even lower dose of 1.25 mg/kg levetiracetam combined with 50 mg/kg valproate. See FIG. 5. An isobologram of levetiracetam and valproate dosages confirmed that the effect of the combined 50 mg/kg valproate and 1.25 mg/kg levetivacetam (VPA 50 + LEV 1.25; empty circle) had a synergistic (super- additive) effect. The combined 50 mg kg valproate and 2.5 mg kg levetivacetam (VPA 50 + LEV 2.5; dark circle), on the other hand, had a simple additive effect, as indicated by its placement on the line. See FIG. 6.

[0227] To calculate die human equivalent dose (HED) for levetiracetam dosage for treatment of age-dependent cogniti ve impairment in humans, we employed the formula HED (mg/kg) = rat dose (mg/kg) x 0.16 (see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologies Evaluation and Research), Therefore, the dosage of 5 mg/kg/day in rats is equivalent to 0.8 mg/kg/day in humans and the dosage of 10 mg/kg/day in rats is equivalent to 1.6 mg/kg/day in humans.

[0228] Example 3: Effect of Levetiracetam in human subjects with aMCI

[0229] A within-subjects trial of 8 weeks duration, involving 17 amnestic MCI (aMCI) subjects and 17 age-matched controls with a low dose treatment of levetiracetam is conducted. During the course of the study, each aMCI subject receives both drug and placebo treatments separately in two periods of two weeks each, with the order of treatments among different aMCI subjects counterbalanced (see FIG. 7). Age-matched control subjects treated with placebo serve as a farther control. Cognitive testing and fMRI imaging data are obtained from the subjects after each two week period of drug/placebo treatment. Participants and clinical characterization

[0230] 17 right-handed aMCI patients are recruited from the Alzheimer's Disease Research Center (ADRC) at the Johns Hopkins Hospital and other referrals. An additional 17 right-handed healthy volunteers are recruited from the pool of control participants in the ADRC and other referrals. Al l participants are administered the Telephone Interview of Cognitive Status to determine if they are likely to pass the entry criteria of the study (including criteri a for MRJ scanning). All participants further undergo neurological, psychiatric, and neuropsychological examination using standardized instruments and methods. The psychiatric evaluation includes administration of the Structured Clinical Interview for DSM- IV Axis I Disorders and the Clinical Dementia Rating (CDR) scale. All aMCI patients have CDR scores of 0.5. Diagnosis of aMCI is based on the criteria proposed by Petersen et al, (e.g., "Mild cognitive impairment: Aging to

Alzheimer's Disease," Oxford University Press, N.Y. (2003), which include a memory complaint (corroborated by an informant), impaired memory function on testing (1.5 standard deviations below norm), otherwise preserved cognitive functioning (within 1 standard deviation of norm), no decline in functional ability, and no dementia. Final aMCI diagnoses are reached by clinical consensus.

Exclusion criteria include major neurological or psychiatric disorders, head trauma with loss of consciousness, history of drug abuse or dependency, and general contraindications to an MRI examination (e.g. cardiac pacemaker, aneurysm coils, claustrophobia). Each aMCI subject is required to have a study partner (i.e., an informant) who can provide information about the subject's daily function and assure that medications are taken appropriately. See FIGS, 18A and 18B.

[0231] Study Visits: The study consists of 4 visits over the course of 8 weeks (see FIG. 7). The Baseline Visit is for the purpose of performing medical, neurological, psychiatric, and neurocognitive assessments. Visits 1 and 2 are identical to the Baseline Visit but include a fMRI session, The Washout Visit, at the end of a 4 week washout period, is for the purpose of a brief clinical assessment and initiation of the second drug/placebo phase.

[0232] Baseline Visit: At the screening visit, informed consent is obtained from the subject (and an informant in the case of MCI subjects). The subject and the informant participate in a standardized clinical interview that is used to determine the degree of the subject's functional impairment in daily life, based on the Clinical Dementia Rating (CDR) scale. The subject's medical, neurological, and psychiatric history is obtained (including a review of current medications), as well as the family history of dementia. Brief medical, neurological and psychiatric exams are conducted (including vital signs). Blood is drawn in order to perform standard laboratory tests needed to determine if the subject meets the entry criteria. The subject is re-screened for contraindications to MR] scanning, using the standard form employed at the Kirby Imaging Center. Brief cognitive testing is performed (described in section on neuropsychological assessment below). These assessments are used to determine if the subject meets the entry criteria. All of the foregoing are completed using standardized forms. If the subject meets entry criteria for the study, the subject is given the study medication (drug or placebo, randomly selected), and instructions about how it should be taken. The subject is advised about the potential for having suicidal thoughts and advised to stop taking the medication and immediately contact the study physician if this occurs.

|0233] Visit 1 : At the end of the first drag/placebo period 2 weeks after the Baseline Visit, the medical, neurological and psychiatric evaluations and cognitive testing are repeated, The subject is also clinically evaluated for suicidal ideation. Blood is drawn again to repeat the standard tests and to determine whether there are any changes related to drug treatment; the subject's blood levetiracetam level is also obtained. All medication dispensed at the Baseline Visit (drug or placebo) is collected and subject compliance with the medication regimen is assessed. The first fMRI session (with cognitive tests) is conducted on the same day, either immediately before or immediately after the clinical assessment. Subjects discontinue first period treatment at this visit. [0234] Washout Visit: At the end of a washout period (4 weeks) following Visit 1, the subject receives a brief medical screening, including a medical and psychiatric evaluation. Blood is drawn to obtain the blood levetiracetam level (to confirm washout). The subject is provided with new medication (drug or placebo, alternated from what was assigned in the previous treatment period) for the final phase of the study with instructions about how it should be taken.

[0235] Visit 2: At approximately 2 weeks after the Washout Visit (i.e., 2 weeks after starting the second treatment period), the medical, neurological and psychiatric evaluations and the cognitive testing are repeated. The subject is clinically evaluated for suicidal ideation. Blood is drawn again to repeat the standard tests and to determine whether there were any changes related to drug treatment; the subject's blood levetiracetam level is also obtained. All medication dispensed at the Washout Visit is collected and subject compliance with the medication regimen is assessed. The second fMRl session (with cognitive tests) is repeated on the same day, either immediately before or immediately after the clinical assessment.

Neuropsychological assessment

[0236] All participants undergo neuropsychological evaluation at the time of assessment for treatment efficacy (Visits I and 2), as well as at the Baseline Visit. The evaluation occurs outside of the scanner and includes the Buschke Selective Reminding Test (Buschke and Fuld, 1974) and the Verbal Paired Associates subtest, the Logical Memory subtest, the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997), and the Benton Visual Retention Test, as these tasks are particularly sensitive to medial temporal lobe function and early memory problems (Marquis et al., 2002 and Masur ei ai, 1994). Additionally, subjects are asked to complete tests of more general cognitive function such as tests to assess general menial status, executive function, attention and general naming ability. All neuropsychological tests are administered by a trained research assistant during a 60-minute session. As the three

neuropsychological assessments in this study occur within a time period of 8 weeks, different versions of the neuropsychological tests are used to minimize test specific practice effects. Breaks are provided to the subject as needed.

Drug administration

|0237] As described above, the drug treatment period is the two weeks preceding Visit 1 or 2 (with the two week period preceding the other Visit being the placebo phase). For the subjects receiving the drug treatment, half a scored 250 mg tablet of levetiracetam is used to achieve a dose of 125 mg/kg twice a day, which is approximately 3.6 mg/kg/day (assuming an average adult huma weight of 70 kg),

[0238] All drug and placebo preparations are performed on a 1 : 1 allocation. The pharmacy randomize patients as they enroll, and keep a list of drag assignment.

|0239] Levetiracetam is rapidly and almost completely absorbed after oral administration, and its bioavailability is not affected by food. Plasma half-life of levetiracetam is approximately 7 ± 1 hour (expected to be 9-10 hours in elderly due to decreased renal function). Absorption is rapid, with peak plasma concentrations occurring about 1 hour following oral administration. Steady state can be achieved after 2 days of multiple twice-daily dosing.

[0240] A typical starting dose of levetiracetam in treating epilepsy in humans is 500 mg twice a day, which is approximately 14,3 mg/kg/day. The dosage is then is increased until optimal efficacy, up to 50 mg/kg/day. Thus, the dose used in this experiment is a quarter of the lowest human dose used for treating epilepsy.

[0241] Even lower dosages, e.g., of 25-60 mg twice a day, are contemplated, based on the results of previous animal studies that indicated low-dose efficacy. The highest effective doses of levetiracetam used in die animal model are 5-10 mg/kg (given acutely). The huma equivalent dose (HED), calculated as described above, of this dosage for treatment of age-dependent cognitive impairment in humans is equivalent to 0.8-1.6 mg/kg/day (or 28-56 mg twice a day).

MRI data acquisition [0242] Imaging data are obtained through high-resolution methods developed in the Stark laboratory ' . Data are collected on a Phillips 3 Tesla scanner (Eindhoven, The Netherlands) equipped with an 8-channeI SENSE (Sensitivity Encoding) head coil, located at the F.M. Kirby Research Center for Functional Brain Imaging at the Kennedy Krieger institute (Baltimore, MD). High-resolution echo-planar images are collected using an acquisition matri of 64 x 64, a repetition time of 1500 milliseconds, an echo time of 30 milliseconds, a flip angle of 70 degrees, a SENSE factor of 2, and an isotropic resolution of 1 .5 mm x 1.5 mm x 1.5 mm with no gap. Nineteen oblique slices are acquired parallel to the principal longitudinal axis of the hippocampus and covered the entire medial temporal lobe region bilaterally. In addition to the functional runs, a whole-brain MPRAGE structural scan

(parameters: 150 oblique slices, lmm isotropic resolution) is acquired.

Image analysis

[0243] Data analysis is carried out using the Analysis for Functional

Neuroimages (AFNI, release 2008 07 18 1710) software. Images are first co- registered to correct for within- and across-scan head motion. Acquisitions in which a significant motion event occur (more than 3 degrees of rotation or 2 mm of translation in any direction relative to prior acquisition), plus and minus one time repetition for 1 ,5 seconds, are excluded from the analyses. Structural anatomical data are registered to standard stereotaxic space (Talairach & Tournoux, 1988), and the same parameters are subsequently applied to the functional data. Behavioral vectors are produced to model different trial types.

|0244] The ROI-LDDMM (large deformation diffeomorphic metric mapping of the region of interest) method, a technique for cross-subject alignment, increases the power of multisubjeci regional fMRI studies by focusing the alignment power specifically on the ROIs (regions of interest) and not elsewhere in the brain. First, all subjects' anatomical and functional scans are normalized to the Talairach atlas using AFNI. Sub-regions of the medial temporal lobe and the hippocampus (bilateral entorhinai cortex, perirhinal cortex, parahippocampal cortex,

CA3/dentate region, CA1 region, and subiculum) are segmented in three dimensions on the MPRAGE scans. The labels for the CA3 region and dentate gyrus (DG) are combined, The anatomically defined ROIs are then used to calculate the ROI-LDDMM 3D vector field transformation for each subject using a customized template based on the mean of the entire sample tested as the target. The ROI-LDDMM transformations for each individual subject's ROIs are then applied to the fit coefficient maps.

[0245] Group data are analyzed using a two-way Analysis of Variance

(ANOVA) with trial types and group as fixed factors, and subject as a random factor nested within group. A liberal peak threshold of p<0.05, along with a spatial extent threshold of 10 voxels are used to define functional ROIs on the overall F statistic. This approach, rather than using a direct pair-wise contrast, reduces voxel selection biases because any differences amongst the various conditions allowed for a voxel to be selected. This threshold is then combined with the anatomical segmentations to only include voxels inside the regions of interest. This serves to exclude voxels that does not change with any of the model's factors, effectively limiting the analysis to voxels showing any changes with task condition or group. Voxels within each functional ROI are collapsed for further analysis.

Cognitive tests during fMRI scans at Visits 1 and 2

[0246] The activity of the subject's medial temporal lobe is measured by functional MRI during the subject's participation in an explicit 3-alternative forced choice task, where participants view novel, repeated and similar ("lure") stimuli. The Psychophysics Toolbox extensions in Matlab 7.0 (The Math Works, Natick, MA) is used for stimulus presentation and behavioral data collection. Stimuli are color photographs of common objects. Each participant undergoes a series of testing runs duing the functional imaging sessions, each run consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. These image pairs are fully randomized throughout the run and presented individually as a series of images (see FIG. 1ΘΑ). Participants are instructed to make a judgment as to whether each object seen is new, old or similar. Of critical interest are the participants' responses when presented with the second of the pair of similar objects (the "lure"; see FIG. 10B). The correct identification by the subject of lure stimuli as "similar," provides behavioral evidence of pattern separation, i.e., the separation of similar experiences into distinct non-overlapping representations, Ho wever, an incorrect indemnification of lure stimuli as "old" or "new," indicates a failure of pattern separation. Identification of lure stimuli as "old' ' ' indicates that the subject focused on the similarities between the lure stimulus and the earlier- shown partner image. Identification of the lure stimulus as "new" indicates that the subject failed to recall the earlier-shown partner image altogether, Each run also contains a number of baseline trials that use a challenging perceptual

discrimination task known to provide a lower and more-stable estimate of basel ine activity in the medial temporal lobe (Stark & Squire, 2001 FNAS; Law et al, 2005).

[0247] A survery of the activity level of various subregions in the medial temporal lobe during the cogitive test, as measured by fMRI, shows that aMCI subjects have hyperactive DG/CA3 regions and a hypoactive entorhinal cortex during the performance of memory tasks, compared to age-matched control subjects.

[0248] W e assess the level of activity in DG/CA3 during successful memory judgments in control and aMCI subjects. The mean activity is calculated from the average activity, as measured by fMRI, during the presentation of lure stimuli correctly idenfied by subject as "similar" that is calibrated for baseline activity. FIG, 8A shows that aMCI patients exhibit DG/CA3 hyperactivity when making these judgments (p = 0.013). FIG. 8B, however, shows that treatment with levetiracetam reduces DG/CA3 hyper-activity in aMCI subjects (p :; = 0.037). The activity level in the aMCI subject treated with the drug, in fact, is normalized to the extent that that it is statistically indistinguishable from the activity of control subjects treated with placebo. See FIG. 8C for the mean activity values shown in FIGS. 8A and SB,

[0249] The activity level during successful memory judgments in EC is significantly lower in placebo-treated aMCI subjects compared to controls (p = 0.003). See FIG. 9A. However, levetiracetam treatment normalizes activity in aMCI subjects in EC as well. See FIG. 9B. Levetiracetam treatment increases EC activity during memory judgments in aMCI subjects, such that it is statistically indistinguishable from placebo-treated control subjects. See FIG. 9B. See FIG. 9C for the mean activity values shown in FIGS. 9A and 9B.

|Θ250] The normalization of DG/CA3 and EC activ ity during memory judgments by levetiracetam treatment is mirrored in the change seen in the aMCI subjects' performace in the cognitive task, With placebo treatment, aMCI patients perform worse that control subjects, correctly identify lure items as "similar" less often and incorrectly identifying them as "old" more often (p :::: 0.009). See FIG, 11, However, the performance of aMCI subjects improves significantly under levetiracetam treatment. See FIG. 12, The interaction of more correct "similar" indenficiations with less incorrect "old" identifications under drug treatment results in a significant improvement in the performance of this memory task (p = 0.039). See FIG. 13 for a table of the data represented in FIGS. 11 and 12.

[0251 ] The performance of control-placebo subjects and aMCI subjects with drug or placebo treatment is also compared in other common cognitive tests, such as the Buschke Selective Reminding Test - Delayed Recall (FIGS. 14A and MB), the Benton Visual Rentention Test (FIGS. ISA and 15B), Verbal Paired

Associates Test - Recognition (FIGS, 16.4 and 16B) and Verbal Paired Associates Test - Delayed Recall (FIGS. 17A and 17B). In all of these tests, aMCI subjects treated with placebo perform worse than placebo-treated control subjects, and levetiracetam treatment fail to rescue performance in aMCI subjects.

[0252 J There are a number of possible reasons why le vetiracetam treatment does not help aMCI subjects with performance in these other cognitive tests. The explicit 3-alternative forced choice task done in the fMRI study is a task that is especially sensitive to DG/C A3 function. As such, the performace of the subjects in this task may be particularly attuned to the changes in DG/CA3 activity resulting from levetiracetam treatment. Further, the aMCI subjects were treated with levetiracetam for only two weeks prior to the administration of the cognitive tests. It is contemplated that a treatment duration of longer than two weeks, e.g., 16 weeks or 8 months, for the drug treatment will result in improved efficacy.

Finally, comparative animal studies (see Example 1 ) indicate that an even lower dose would be more effective, The human dosage of 125 nig twice a day is equivalent to a rat dosage of 22.3 mg/kg/day. As is shown in Example 2 and FIG, 3, 20 mg kg levetiraeetam is too high a dose in rats, and it fails to improve the performace of AI rats in the radial maze task. The effective doses of levetiraeetam used in the animal model are 5-10 mg kg. The human equivalent dose (HED) of the optimal rat dose is 0.8-1.6 mg/kg/day. Such a dosage would result in the administration of 28-56 mg twice a day (which is substantially lower than the 125 mg twice a day used in this study). Thus, it is contemplated that aMCI subjects will exhibit a further normalization of DG/CA3 and EC activiiy, as well as further improved performance in cognitive tests, if they are treated with lower doses equivalent to the effective doses in rat, e.g., 25 - 60 mg twice a day of

levetiraeetam.