Compositions and Methods for Modulation of SMN2 Splicing in a Subject

RIGO; Frank ;   et al.

Patent Application Summary

U.S. patent application number 15/303829 was filed with the patent office on 2017-02-16 for compositions and methods for modulation of smn2 splicing in a subject. The applicant listed for this patent is Biogen MA Inc.. Invention is credited to C. Frank BENNETT, Frank RIGO.

Application Number20170044538 15/303829
Document ID /
Family ID54324712
Filed Date2017-02-16

United States Patent Application 20170044538
Kind Code A1
RIGO; Frank ;   et al. February 16, 2017

Compositions and Methods for Modulation of SMN2 Splicing in a Subject

Abstract

Disclosed herein are compounds, compositions and methods for modulating splicing of SMN2 mRNA in a subject.


Inventors: RIGO; Frank; (Carlsbad, CA) ; BENNETT; C. Frank; (Carlsbad, CA)
Applicant:
Name City State Country Type

Biogen MA Inc.

Cambridge

MA

US
Family ID: 54324712
Appl. No.: 15/303829
Filed: April 17, 2015
PCT Filed: April 17, 2015
PCT NO: PCT/US2015/026326
371 Date: October 13, 2016

Related U.S. Patent Documents

Application Number Filing Date Patent Number
61981139 Apr 17, 2014

Current U.S. Class: 1/1
Current CPC Class: C12N 2310/11 20130101; C12N 2320/33 20130101; C12N 15/113 20130101; C12N 2310/315 20130101; C12N 2310/346 20130101; C12N 2310/343 20130101; C12N 2310/321 20130101; C12N 2310/322 20130101; C12N 2310/3525 20130101; C12N 2310/3341 20130101
International Class: C12N 15/113 20060101 C12N015/113

Claims



1. A compound comprising a modified oligonucleotide consisting of 16-20 linked nucleosides, wherein the modified oligonucleotide is complementary to SMN2; and wherein each internucleoside linkage is either phosphorothioate or phosphodiester and at least one internucleoside linkage is phosphorothioate and at least one internucleoside linkage is phosphodiester.

2. A compound comprising a modified oligonucleotide consisting of 16-20 linked nucleosides, wherein the modified oligonucleotide has a nucleobase sequence selected from any one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 and having 6 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

3. A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

4. A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 7, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

5. A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

6. The compound of any of claims 1 to 5, having 7 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

7. The compound of any of claims 1 to 5, having 8 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

8. The compound of any of claims 1 to 5, having 9 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

9. The compound of any of claims 1 to 5, having 10 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

10. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNoNoNsNsNsNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

11. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

12. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

13. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

14. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNoNsNoNsNoNsNoNsNoNsNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

15. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

16. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

17. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

18. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNsNoNoNsNoNoNsNoNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

19. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

20. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

21. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

22. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

23. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNoNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

24. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

25. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

26. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

27. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

28. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

29. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNsNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

30. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

31. The compound of claim 3, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

32. The compound of claim 3, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

33. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

34. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNoNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

35. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

36. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

37. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

38. The compound of claim 4, wherein the modified oligonucleotide has an NsNoNsNsNsNsNoNsNsNsNsNoNsNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

39. The compound of claim 5, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

40. The compound of claim 5, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

41. The compound of claim 5, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

42. The compound of claim 5, wherein the modified oligonucleotide has an NsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

43. The compound of claim 5, wherein the modified oligonucleotide has an NsNsNsNsNoNsNsNsNsNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

44. The compound of any of claims 1 to 43, wherein each nucleoside of the modified oligonucleotide comprises a modified sugar moiety, and wherein each modified sugar moiety comprises a 2'-methoxyethyl modification.

45. A pharmaceutical composition comprising the compound of any of claims 1 to 44.

46. A method of promoting inclusion of exon 7 in SMN2 transcripts in a cell, tissue or organ, comprising contacting said cell, tissue or organ with the compound of any of claims 1 to 44 or the composition of claim 45.

47. A method of treating Spinal Muscular Atrophy Type I, comprising administering the compound of any of claims 1 to 44 or the composition of claim 45 to a patient in need thereof.

48. A method of treating Spinal Muscular Atrophy Type II, comprising administering the compound of any of claims 1 to 44 or the composition of claim 45 to a patient in need thereof.

49. A method of treating Spinal Muscular Atrophy Type III, comprising administering the compound of any of claims 1 to 44 or the composition of claim 45 to a patient in need thereof.

50. A method of treating Spinal Muscular Atrophy Type IV, comprising administering the compound of any of claims 1 to 44 or the composition of claim 45 to a patient in need thereof.

51. An antisense oligonucleotide of any of claims 1 to 44 or the composition of claim 45 for use in therapy.

52. Use of an antisense oligonucleotide of any of claims 1 to 44 or the composition of claim 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type I.

53. Use of an antisense oligonucleotide of any of claims 1 to 44 or the composition of claim 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type II

54. Use of an antisense oligonucleotide of any of claims 1 to 44 or the composition of claim 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type III.

55. Use of an antisense oligonucleotide of any of claims 1 to 44 or the composition of claim 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type IV.

56. Use of an antisense oligonucleotide of any of claims 1 to 45 for the preparation of a medicament for the treatment of spinal muscular atrophy.

57. A compound comprising a modified oligonucleotide consisting of 16-20 linked nucleosides, wherein the modified oligonucleotide has a nucleobase sequence selected from any one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, or 27 and having 6 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

58. The compound of claim 57, having 7 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

59. The compound of claim 57, having 8 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

60. The compound of claim 57, having 9 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

61. The compound of claim 57, having 10 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

62. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNoNoNsNsNsNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

63. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

64. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

65. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

66. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNoNsNoNsNoNsNoNsNoNsNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

67. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

68. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

69. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

70. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNsNoNoNsNoNoNsNoNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

71. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

72. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

73. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

74. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

75. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNoNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

76. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

77. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

78. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

79. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

80. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

81. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNsNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

82. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

83. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

84. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

85. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

86. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNoNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

87. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

88. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

89. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

90. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNsNsNsNoNsNsNsNsNoNsNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

91. The compound of claim 57, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

92. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

93. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

94. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

95. The compound of claim 57, wherein the modified oligonucleotide has an NsNsNsNsNoNsNsNsNsNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

96. The compound of any of claims 57 to 95, wherein each nucleoside of the modified oligonucleotide comprises a modified sugar moiety, and wherein each modified sugar moiety comprises a 2'-methoxyethyl modification.

97. A pharmaceutical composition comprising the compound of any of claims 57 to 96.

98. A method of promoting inclusion of exon 7 in SMN2 transcripts in a cell, tissue or organ, comprising contacting said cell, tissue or organ with the compound of any of claims 57 to 95 or the composition of claim 96.

99. A method of treating Spinal Muscular Atrophy Type I, comprising administering the compound of any of claims 57 to 95 or the composition of claim 96 to a patient in need thereof.

100. A method of treating Spinal Muscular Atrophy Type II, comprising administering the compound of any of claims 57 to 95 or the composition of claim 96 to a patient in need thereof.

101. A method of treating Spinal Muscular Atrophy Type III, comprising administering the compound of any of claims 57 to 95 or the composition of claim 96 to a patient in need thereof.

102. A method of treating Spinal Muscular Atrophy Type IV, comprising administering the compound of any of claims 57 to 95 or the composition of claim 96 to a patient in need thereof.

103. An antisense oligonucleotide of any of claims 57 to 95 or the composition of claim 96 for use in therapy.

104. Use of an antisense oligonucleotide of any of claims 57 to 95 or the composition of claim 96 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type I.

105. Use of an antisense oligonucleotide of any of claims 57 to 95 or the composition of claim 96 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type II

106. Use of an antisense oligonucleotide of any of claims 57 to 95 or the composition of claim 96 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type III.

107. Use of an antisense oligonucleotide of any of claims 57 to 95 or the composition of claim 96 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type IV.

108. Use of an antisense oligonucleotide of any of claims 57 to 96 for the preparation of a medicament for the treatment of spinal muscular atrophy.
Description



SEQUENCE LISTING

[0001] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0245WOSEQ_ST25.txt, created Apr. 3, 2015, which is 44 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

FIELD

[0002] Provided herein are methods, compounds, and compositions for modulation of SMN2 splicing in a subject.

BACKGROUND

[0003] Newly synthesized eukaryotic mRNA molecules, known as primary transcripts or pre-mRNA are processed before translation. Processing of the pre-mRNAs includes addition of a 5' methylated cap and an approximately 200-250 base poly(A) tail to the 3' end of the transcript. Processing of mRNA from pre-mRNA also frequently involves splicing of the pre-mRNA, which occurs in the maturation of 90-95% of mammalian mRNAs. Introns (or intervening sequences) are regions of a pre-mRNA (or the DNA encoding it) that are not included in the coding sequence of the mature mRNA. Exons are regions of a primary transcript that remain in the mature mRNA. The exons are spliced together to form the mature mRNA sequence. Splice junctions are also referred to as splice sites with the 5' side of the junction often called the "5' splice site," or "splice donor site" and the 3' side the "3' splice site" or "splice acceptor site." In splicing, the 3' end of an upstream exon is joined to the 5' end of the downstream exon. Thus the unspliced pre-mRNA has an exon/intron junction at the 5' end of an intron and an intron/exon junction at the 3' end of an intron. After the intron is removed, the exons are contiguous at what is sometimes referred to as the exon/exon junction or boundary in the mature mRNA. Cryptic splice sites are those which are less often used but may be used when the usual splice site is blocked or unavailable. Alternative splicing, defined as the splicing together of different combinations of exons, often results in multiple mRNA transcripts from a single gene.

[0004] Up to 50% of human genetic diseases resulting from a point mutation result in aberrant pre-mRNA processing. Such point mutations can either disrupt a current splice site or create a new splice site, resulting in mRNA transcripts comprised of a different combination of exons or with deletions in exons. Point mutations also can result in activation of a cryptic splice site or disrupt regulatory cis elements (i.e. splicing enhancers or silencers) (Cartegni et al., Nat. Rev. Genet., 2002, 3, 285-298; Drawczak et al., Hum. Genet., 1992, 90, 41-54). Antisense oligonucleotides have been used to target mutations that lead to aberrant splicing in several genetic diseases in order to redirect splicing to give a desired splice product (Kole, Acta Biochimica Polonica, 1997, 44, 231-238).

[0005] Antisense compounds have also been used to alter the ratio of naturally occurring alternate splice variants such as the long and short forms of Bcl-x pre-mRNA (U.S. Pat. No. 6,172,216; U.S. Pat. No. 6,214,986; Taylor et al., Nat. Biotechnol. 1999, 17, 1097-1100) or to force skipping of specific exons containing premature termination codons (Wilton et al., Neuromuscul. Disord., 1999, 9, 330-338). U.S. Pat. No. 5,627,274 and WO 94/26887 disclose compositions and methods for combating aberrant splicing in a pre-mRNA molecule containing a mutation using antisense oligonucleotides which do not activate RNAse H.

[0006] Proximal spinal muscular atrophy (SMA) is a genetic, neurodegenerative disorder characterized by the loss of spinal motor neurons. SMA is an autosomal recessive disease of early onset and is currently the leading cause of death among infants. The severity of SMA varies among patients and has thus been classified into three types. Type I SMA is the most severe form with onset at birth or within 6 months and typically results in death within 2 years. Children with type I SMA are unable to sit or walk. Type II SMA is the intermediate form and patients are able to sit, but cannot stand or walk. Patients with type III SMA, a chronic form of the disease, typically develop SMA after 18 months of age (Lefebvre et al., Hum. Mol. Genet., 1998, 7, 1531-1536).

[0007] The molecular basis of SMA is caused by the loss of both copies of survival motor neuron gene 1 (SMN1), which may also be known as SMN Telomeric, a protein that is part of a multi-protein complex thought to be involved in snRNP biogenesis and recycling. A nearly identical gene, SMN2, which may also be known as SMN Centromeric, exists in a duplicated region on chromosome 5q13 and modulates disease severity. Expression of the normal SMN1 gene results solely in expression of survival motor neuron (SMN) protein. Although SMN1 and SMN2 have the potential to code for the same protein, SMN2 contains a translationally silent mutation at position +6 of exon 7, which results in inefficient inclusion of exon 7 in SMN2 transcripts. Thus, the predominant form of SMN2 is a truncated version, lacking exon 7, which is unstable and inactive (Cartegni and Krainer, Nat. Genet., 2002, 30, 377-384). Expression of the SMN2 gene results in approximately 10-20% of the SMN protein and 80-90% of the unstable/non-functional SMNdelta7 protein. SMN protein plays a well-established role in assembly of the spliceosome and may also mediate mRNA trafficking in the axon and nerve terminus of neurons.

[0008] Antisense technology is an effective means for modulating the expression of one or more specific gene products, including alternative splice products, and is uniquely useful in a number of therapeutic, diagnostic, and research applications. The principle behind antisense technology is that an antisense compound, which hybridizes to a target nucleic acid, modulates gene expression activities such as transcription, splicing or translation through one of a number of antisense mechanisms. The sequence specificity of antisense compounds makes them extremely attractive as tools for target validation and gene functionalization, as well as therapeutics to selectively modulate the expression of genes involved in disease.

[0009] Certain antisense compounds complementary to SMN2 are known in the art. See for example, WO 2007/002390; U.S. 61/168,885; Hua et al., American J. of Human Genetics (April 2008) 82, 1-15; Singh et al., RNA Bio. 6:3, 1-10 (2009). Certain antisense compounds and methods disclosed herein posses desirable characteristics compared to such compounds and methods known in the art. Chimeric peptide nucleic acid molecules designed to modulate splicing of SMN2 have been described (WO 02/38738; Cartegni and Krainer, Nat. Struct. Biol., 2003, 10, 120-125).

SUMMARY

[0010] In certain embodiments, the present invention provides methods comprising administering to a subject an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2 pre-mRNA, wherein the antisense compound is administered into the cerebrospinal fluid. In certain embodiments, the administration is into the intrathecal space. In certain embodiments, the administration is into the cerebrospinal fluid in the brain. In certain embodiments, the administration comprises a bolus injection. In certain embodiments, the administration comprises infusion with a delivery pump.

[0011] In certain embodiments, the antisense compound is administered at a dose from 0.01 to 10 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the dose is from 0.01 to 10 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the dose is from 0.01 to 5 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the dose is from 0.05 to 1 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the dose is from 0.01 to 0.5 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the dose is from 0.05 to 0.5 milligrams of antisense compound per kilogram of body weight of the subject.

[0012] In certain embodiments, the dose is administered daily. In certain embodiments, the dose is administered weekly. In certain embodiments, the antisense compound is administered continuously and wherein the dose is the amount administered per day. In certain embodiments, the method comprises administering at least one induction dose during an induction phase and administering at least one maintenance dose during a maintenance phase. In certain embodiments, the induction dose is from 0.05 to 5.0 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the maintenance dose is from 0.01 to 1.0 milligrams of antisense compound per kilogram of body weight of the subject. In certain embodiments, the duration of the induction phase is at least 1 week. In certain embodiments, the duration of the maintenance phase is at least 1 week. In certain embodiments, each induction dose and each maintenance dose comprises a single injection. In certain embodiments, each induction dose and each maintenance dose independently comprise two or more injections. In certain embodiments, antisense compound is administered at least 2 times over a treatment period of at least 1 week. In certain embodiments, the treatment period is at least one month. In certain embodiments, the treatment period is at least 2 months. In certain embodiments, the treatment period is at least 4 months. In certain embodiments, the induction dose is administered by one or more bolus injections and the maintenance dose is administered by an infusion pump.

[0013] In certain embodiments, the method comprises assessing the tolerability and/or effectiveness of the antisense compound. In certain embodiments, dose amount or frequency of antisense compound is reduced following an indication that administration of the antisense compound is not tolerated. In certain embodiments, the dose amount or frequency of antisense compound is maintained or reduced following an indication that administration of the antisense compound is effective. In certain embodiments, the dose of antisense compound is increased following an indication that administration of the antisense compound is not effective. In certain embodiments, frequency of administration of antisense compound is reduced following an indication that administration of the antisense compound is effective. In certain embodiments, frequency of administration of antisense compound is increased following an indication that administration of the antisense compound is not effective.

[0014] In certain embodiments, the methods comprise co-administration of the antisense compound and at least one other therapy. In certain embodiments, an antisense compound and at least one other therapy are co-administered at the same time. In certain embodiments, an antisense compound is administered prior to administration of the at least one other therapy. In certain embodiments, an antisense compound is administered after administration of the at least one other therapy. In certain embodiments, the at least one other therapy comprises administration of one or more of valproic acid, riluzole, hydroxyurea, and a butyrate. In certain embodiments, at least one other therapy comprises administration of trichostatin-A. In certain embodiments, the at least one other therapy comprises administration of stem cells. In certain embodiments, at least one other therapy is gene therapy. In certain embodiments, gene therapy is administered to the CSF and an antisense compound is administered systemically. In certain embodiments, gene therapy is administered to the CSF and an antisense compound is administered systemically and to the CSF. In certain embodiments, the invention provides treatment regimens where initially, an antisense compound is administered to the CSF and systemically, followed by gene therapy administration to the CSF and systemic administration of antisense compound. In certain such embodiments, the subject is an infant at the time of initial treatment. In certain such embodiments, the subject is less that 2 years old. In certain embodiments, antisense compound is administered to the CNS of a subject until the subject is old enough for gene therapy. In certain such embodiments, antisense compound is administered systemically throughout.

[0015] In certain embodiments, the antisense compound is administered at a concentration of about 0.01 mg/ml, about 0.05 mg/ml, about 0.1 mg/ml, about 0.5 mg/ml, about 1 mg/ml, about 5 mg/ml, about 10 mg/ml, about 50 mg/ml, or about 100 mg/ml.

[0016] In certain embodiments, inclusion of exon 7 of SMN2 mRNA in a motoneuron in the subject is increased. In certain embodiments, inclusion of exon 7 amino acids in SMN2 polypeptide in a motoneuron in the subject is increased.

[0017] In certain embodiments, the invention provides methods of increasing inclusion of exon 7 of SMN2 mRNA in a motoneuron in a subject comprising administering to the subject an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2 and thereby increasing inclusion of exon 7 of SMN2 mRNA in the motoneuron in the subject.

[0018] In certain embodiments, the invention provides methods of increasing inclusion of exon 7 amino acids in SMN2 polypeptide in a motoneuron in a subject comprising administering to the subject an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2 and thereby increasing inclusion of exon 7 amino acids in SMN2 polypeptide in the motoneuron in the subject.

[0019] In certain embodiments, the subject has SMA. In certain embodiments, the subject has type I SMA. In certain embodiments, the subject has type II SMA. In certain embodiments, the subject has type III SMA.

[0020] In certain embodiments, a first dose is administered in utero. In certain embodiments, the first dose is administered prior to complete formation of the blood-brain-barrier. In certain embodiments, a first dose is administered within 1 week of birth of the subject. In certain embodiments, a first dose is administered within 1 month of birth of the subject. In certain embodiments, a first dose is administered within 3 months of birth of the subject. In certain embodiments, a first dose is administered within 6 months of birth of the subject. In certain embodiments, a first dose is administered when the subject is from 1 to 2 years of age. In certain embodiments, a first dose is administered when the subject is from 1 to 15 years of age. In certain embodiments, a first dose is administered when the subject is older than 15 years of age.

[0021] In certain embodiments, the subject is a mammal. In certain embodiments, the subject is a human.

[0022] In certain embodiments, the methods comprise identifying a subject having SMA. In certain embodiments, the subject is identified by measuring electrical activity of one or more muscles of the subject. In certain embodiments, the subject is identified by a genetic test to determine whether the subject has a mutation in the subject's SMN1 gene. In certain embodiments, the subject is identified by muscle biopsy.

[0023] In certain embodiments, administering the antisense compound results in an increase in the amount of SMN2 mRNA having exon 7 of at least 10%. In certain embodiments, the increase in the amount of SMN2 mRNA having exon 7 is at least 20%. In certain embodiments, the increase in the amount of SMN2 mRNA having exon 7 is at least 50%. In certain embodiments, the amount of SMN2 mRNA having exon 7 is at least 70%.

[0024] In certain embodiments, administering of the antisense compound results in an increase in the amount of SMN2 polypeptide having exon 7 amino acids of at least 10%. In certain embodiments, wherein the increase in the amount of SMN2 polypeptide having exon 7 amino acids is at least 20%. In certain embodiments, the increase in the amount of SMN2 polypeptide having exon 7 amino acids is at least 50%. In certain embodiments, the increase in the amount of SMN2 polypeptide having exon 7 amino acids is at least 70%.

[0025] In certain embodiments, the administering of the antisense compound ameliorates at least one symptom of SMA in the subject. In certain embodiments, the administering of the antisense compound results in improved motor function in the subject. In certain embodiments, the administering of the antisense compound results in delayed or reduced loss of motor function in the subject. In certain embodiments, administering of the antisense compound results in improved respiratory function. In certain embodiments, the administering of the antisense compound results in improved survival.

[0026] In certain embodiments, at least one nucleoside of the antisense oligonucleotide comprises a modified sugar moiety. In certain embodiments, at least one modified sugar moiety comprises a 2'-methoxyethyl sugar moiety. In certain embodiments, essentially each nucleoside of the antisense oligonucleotide comprises a modified sugar moiety. In certain embodiments, the nucleosides comprising a modified sugar moiety all comprise the same sugar modification. In certain embodiments, wherein each modified sugar moiety comprises a 2'-methoxyethyl sugar moiety. In certain embodiments, each nucleoside of the antisense oligonucleotide comprises a modified sugar moiety. In certain embodiments, the nucleosides all comprise the same sugar modification. In certain embodiments, each modified sugar moiety comprises a 2'-methoxyethyl sugar moiety. In certain embodiments, at least one internucleoside linkage is a phosphorothioate internucleoside linkage. In certain embodiments, each internucleoside linkage is a phosphorothioate internucleoside linkage.

[0027] In certain embodiments, the antisense oligonucleotide consists of 10 to 25 linked nucleosides. In certain embodiments, the antisense oligonucleotide consists of 12 to 22 linked nucleosides. In certain embodiments, the antisense oligonucleotide consists of 15 to 20 linked nucleosides. In certain embodiments, the antisense oligonucleotide consists of 18 linked nucleosides.

[0028] In certain embodiments, the antisense oligonucleotide is at least 90% complementary to the nucleic acid encoding human SMN2. In certain embodiments, the antisense oligonucleotide is fully complementary to the nucleic acid encoding human SMN2. In certain embodiments, the oligonucleotide has a nucleobase sequence comprising at least 10 contiguous nucleobases of the nucleobase sequence SEQ ID NO: 1. In certain embodiments, the oligonucleotide has a nucleobase sequence comprising at least 15 contiguous nucleobases of the nucleobase sequence SEQ ID NO: 1. In certain embodiments, the oligonucleotide has a nucleobase sequence comprising the nucleobase sequence SEQ ID NO: 1. In certain embodiments, the oligonucleotide has a nucleobase sequence consisting of the nucleobase sequence SEQ ID NO: 1.

[0029] In certain embodiments, the antisense compound comprises a conjugate group or terminal group.

[0030] In certain embodiments, the antisense compound consists of the antisense oligonucleotide.

[0031] In certain embodiments, the antisense compound is also administered systemically. In certain embodiments, the systemic administration is by intravenous or intraperitoneal injection. In certain embodiments, systemic administration and the administration into the central nervous system are performed at the same time. In certain embodiments, systemic administration and the administration into the central nervous system are performed at different times.

[0032] In certain embodiments, the invention provides systemic administration of antisense compounds, either alone or in combination with delivery into the CSF. In certain embodiments, pharmaceutical compositions are administered systemically. In certain embodiments, pharmaceutical compositions are administered subcutaneously. In certain embodiments, pharmaceutical compositions are administered intravenously. In certain embodiments, pharmaceutical compositions are administered by intramuscular injection.

[0033] In certain embodiments, pharmaceutical compositions are administered both directly to the CSF (e.g., IT and/or ICV injection and/or infusion) and systemically.

[0034] In certain embodiments, the invention provides methods of administering to a subject having at least one symptom associated with SMA, at least one dose of an antisense compound comprising an oligonucleotide consisting of 15 to 20 linked nucleosides and having a nucleobase sequence which is 100% complementary to SEQ ID NO. 7 over its entire length, and wherein each nucleoside is a 2'-MOE modified nucleoside; and wherein at least one dose is between 0.1 mg/kg and 5 mg/kg administered to the CSF. In certain such embodiments, the dose is between 0.5 mg/kg and 2 mg/kg. In certain embodiments, at least one dose is administered by bolus injection. In certain such embodiments, the dose is administered by bolus intrathecal injection. In certain embodiments, at least one second dose is administered. In certain such embodiments, the second dose is administered at least 2 weeks after the first dose. In certain embodiments, the second dose is administered at least 4 weeks after the first dose. In certain embodiments, the second dose is administered at least 8 weeks after the first dose. In certain embodiments, the second dose is administered at least 12 weeks after the first dose. In certain embodiments, the second dose is administered at least 16 weeks after the first dose. In certain embodiments, the second dose is administered at least 20 weeks after the first dose. In certain embodiments, the subject is under 2 years old at the time of the first dose. In certain embodiments, the subject is between 2 and 15 years old. In certain embodiments, the subject is between 15 and 30 years old. In certain embodiments, the subject is older than 30 years old. In certain embodiments, at least one symptom associated with SMA is reduced its progression has slowed. In certain embodiments, the oligonucleotide is ISIS396443.

[0035] In certain embodiments, the invention provides methods of administering to a subject having at least one symptom associated with SMA, at least one dose of an antisense compound comprising an oligonucleotide consisting of 15 to 20 linked nucleosides and having a nucleobase sequence comprising which is 100% complementary to SEQ ID NO. 7 over its entire length, and wherein each nucleoside is a 2'-MOE modified nucleoside; and wherein at least one dose is administered systemically. In certain such embodiments, at least one dose is administered by bolus injection. In certain such embodiments, the dose is administered by bolus subcutaneous injection. In certain embodiments, the dose administered is between 0.5 mg/kg and 50 mg/kg. In certain embodiments, the dose is between 1 mg/kg and 10 mg/kg. In certain embodiments, the dose is between 1 mg/kg and 5 mg/kg. In certain embodiments, the dose is between 0.5 mg/kg and 1 mg/kg. In certain embodiments, at least one second dose is administered. In certain such embodiments, the second dose is administered at least 2 weeks after the first dose. In certain embodiments, the second dose is administered at least 4 weeks after the first dose. In certain embodiments, the second dose is administered at least 8 weeks after the first dose. In certain embodiments, the second dose is administered at least 12 weeks after the first dose. In certain embodiments, the second dose is administered at least 16 weeks after the first dose. In certain embodiments, the second dose is administered at least 20 weeks after the first dose. In certain embodiments, the subject is under 2 years old at the time of the first dose. In certain embodiments, the subject is between 2 and 15 years old. In certain embodiments, the subject is between 15 and 30 years old. In certain embodiments, the subject is older than 30 years old. In certain embodiments, at least one symptom associated with SMA is reduced its progression has slowed. In certain embodiments, the oligonucleotide is ISIS396443.

[0036] In certain embodiments, the invention provides methods of administering to a subject having at least one symptom associated with SMA, at least one dose to the CSF and at least one systemic dose of an antisense compound comprising an oligonucleotide consisting of 15 to 20 linked nucleosides and having a nucleobase sequence which is 100% complementary to SEQ ID NO. 7 over its entire length, and wherein each nucleoside is a 2'-MOE modified nucleoside. In certain such embodiments, the CSF dose is between 0.1 mg/kg and 5 mg/kg. In certain embodiments, the systemic dose is between 0.5 mg/kg and 50 mg/kg. In certain embodiments, at least one CSF dose is administered by bolus injection. In certain such embodiments, at least one CSF dose is administered by bolus intrathecal injection. In certain embodiments, at least one systemic dose is administered by bolus injection. In certain such embodiments, at least one systemic dose is administered by subcutaneous injection. In certain embodiments, the CSF dose and the systemic dose are administered at the same time. In certain embodiments, the CSF dose and the systemic dose are administered at different times. In certain embodiments, the subject is under 2 years old at the time of the first dose. In certain embodiments, the subject is between 2 and 15 years old. In certain embodiments, the subject is between 15 and 30 years old. In certain embodiments, the subject is older than 30 years old. In certain embodiments, at least one symptom associated with SMA is reduced its progression has slowed. In certain embodiments, the oligonucleotide is ISIS396443.

[0037] In certain embodiments, the invention provides methods of administering to a subject having at least one symptom associated with SMA, at least one systemic dose of an antisense compound comprising an oligonucleotide consisting of 15 to 20 linked nucleosides and having a nucleobase sequence which is 100% complementary to SEQ ID NO. 7 over its entire length, and wherein each nucleoside is a 2'-MOE modified nucleoside; and at least one dose of a gene therapy agent. In certain embodiments, the systemic dose is between 0.5 mg/kg and 50 mg/kg. In certain embodiments, at least one systemic dose is administered by bolus injection. In certain such embodiments, at least one systemic dose is administered by subcutaneous injection. In certain embodiments, the systemic dose and the gene therapy agent are administered at the same time. In certain embodiments, the systemic dose and the gene therapy agent are administered at different times. In certain embodiments, the gene therapy agent is administered to the CSF. In certain such embodiments, the gene therapy agent is administered by intrathecal injection and/or infusion. In certain such embodiments, the gene therapy agent is administered by intracerebroventricular injection and/or infusion. In certain embodiments, the subject is under 2 years old at the time of the first dose. In certain embodiments, the subject is between 2 and 15 years old. In certain embodiments, the subject is between 15 and 30 years old. In certain embodiments, the subject is older than 30 years old. In certain embodiments, at least one symptom associated with SMA is reduced or its progression has slowed. In certain embodiments, the oligonucleotide is ISIS396443.

[0038] In certain embodiments, the invention provides methods of selecting a subject having at least one symptom associated with SMA and administering an antisense compound according to any of the methods above. In certain such embodiments, at least one symptom of SMA is assessed after administration. In certain such embodiments, at least one symptom of SMA is improved. In certain such embodiments, at least one symptom of SMA does not progress or progresses more slowly compared to a subject who has not received administration of antisense compound.

[0039] In certain embodiments, the invention provides an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2, for use in any of the above methods. In certain embodiments, the invention provides such a compound for use in treating a disease or condition associated with survival motor neuron 1 (SMN1).

[0040] In certain embodiments, the invention provides use of an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2 in the manufacture of a medicament for use in any of the above methods. In certain embodiments, the medicament is for treating a disease or condition associated with survival motor neuron 1 (SMN1).

[0041] In certain embodiments, the present invention provides methods comprising administering to a subject an antisense compound comprising an antisense oligonucleotide complementary to intron 7 of a nucleic acid encoding human SMN2 pre-mRNA, wherein the antisense compound is administered into the cerebrospinal fluid. In certain embodiments, the administration is into the intrathecal space. In certain embodiments, the administration is into the cerebrospinal fluid in the brain. In certain embodiments, the administration comprises a bolus injection. In certain embodiments, the administration comprises infusion with a delivery pump.

[0042] The present disclosure provides the following non-limiting numbered embodiments:

Embodiment 1

[0043] A compound comprising a modified oligonucleotide consisting of 16-20 linked nucleosides, wherein the modified oligonucleotide is complementary to SMN2; and wherein each internucleoside linkage is either phosphorothioate or phosphodiester and at least one internucleoside linkage is phosphorothioate and at least one internucleoside linkage is phosphodiester.

Embodiment 2

[0044] A compound comprising a modified oligonucleotide consisting of 16-20 linked nucleosides, wherein the modified oligonucleotide has a nucleobase sequence selected from any one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 and having 6 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 3

[0045] A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 4

[0046] A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 7, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 5

[0047] A compound comprising a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2, wherein the modified oligonucleotide has 6 or more phosphodiester internucleoside linkages, and wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 6

[0048] The compound of any of embodiments 1 to 5, having 7 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 7

[0049] The compound of any of embodiments 1 to 5, having 8 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 8

[0050] The compound of any of embodiments 1 to 5, having 9 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 9

[0051] The compound of any of embodiments 1 to 5, having 10 or more phosphodiester internucleoside linkages, wherein each internucleoside linkage that is not a phosphodiester internucleoside linkage is a phosphorothioate internucleoside linkage.

Embodiment 10

[0052] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNoNoNsNsNsNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 11

[0053] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 12

[0054] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 13

[0055] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 14

[0056] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNoNsNoNsNoNsNoNsNoNsNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 15

[0057] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 16

[0058] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 17

[0059] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNsNoNsNoNsNoNsNoNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 18

[0060] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNsNoNoNsNoNoNsNoNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 19

[0061] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNsNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 20

[0062] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNoNsNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 21

[0063] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNsNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 22

[0064] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNoNoNsNsNsNsNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 23

[0065] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNoNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 24

[0066] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNoNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 25

[0067] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNoNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 26

[0068] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 27

[0069] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNsNoNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 28

[0070] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNoNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 29

[0071] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNsNsNsNsNoNoNoNoNoNsNsNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 30

[0072] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNoNoNsNsNsNsNsNsNsNsNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 31

[0073] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNsNoNoNoNoNoNoNoNoNoNoNoNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 32

[0074] The compound of embodiment 3, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 33

[0075] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 34

[0076] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNoNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 35

[0077] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 36

[0078] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 37

[0079] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 38

[0080] The compound of embodiment 4, wherein the modified oligonucleotide has an NsNoNsNsNsNsNoNsNsNsNsNoNsNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 39

[0081] The compound of embodiment 5, wherein the modified oligonucleotide has an NsNoNsNoNsNoNsNoNsNoNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 40

[0082] The compound of embodiment 5, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNoNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 41

[0083] The compound of embodiment 5, wherein the modified oligonucleotide has an NsNsNoNsNsNoNsNsNoNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 42

[0084] The compound of embodiment 5, wherein the modified oligonucleotide has an NsNsNsNoNsNsNsNoNsNsNsNoNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 43

[0085] The compound of embodiment 5, wherein the modified oligonucleotide has an NsNsNsNsNoNsNsNsNsNoNsNsNsNsN internucleoside motif, wherein each "N" represents a nucleobase, each "s" represents a phosphorothioate internucleoside linkage, and each "o" represents a phosphodiester internucleoside linkage.

Embodiment 44

[0086] The compound of any of embodiments 1 to 43, wherein each nucleoside of the modified oligonucleotide comprises a modified sugar moiety, and wherein each modified sugar moiety comprises a 2'-methoxyethyl modification.

Embodiment 45

[0087] A pharmaceutical composition comprising the compound of any of embodiments 1 to 44.

Embodiment 46

[0088] A method of promoting inclusion of exon 7 in SMN2 transcripts in a cell, tissue or organ, comprising contacting said cell, tissue or organ with the compound of any of embodiments 1 to 44 or the composition of embodiment 45.

Embodiment 47

[0089] A method of treating Spinal Muscular Atrophy Type I, comprising administering the compound of any of embodiments 1 to 44 or the composition of embodiment 45 to a patient in need thereof.

Embodiment 48

[0090] A method of treating Spinal Muscular Atrophy Type II, comprising administering the compound of any of embodiments 1 to 44 or the composition of embodiment 45 to a patient in need thereof.

Embodiment 49

[0091] A method of treating Spinal Muscular Atrophy Type III, comprising administering the compound of any of embodiments 1 to 44 or the composition of embodiment 45 to a patient in need thereof.

Embodiment 50

[0092] A method of treating Spinal Muscular Atrophy Type IV, comprising administering the compound of any of embodiments 1 to 44 or the composition of embodiment 45 to a patient in need thereof.

Embodiment 51

[0093] An antisense oligonucleotide of any of embodiments 1 to 44 or the composition of embodiment 45 for use in therapy.

Embodiment 52

[0094] Use of an antisense oligonucleotide of any of embodiments 1 to 44 or the composition of embodiment 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type I.

Embodiment 53

[0095] Use of an antisense oligonucleotide of any of embodiments 1 to 44 or the composition of embodiment 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type II

Embodiment 54

[0096] Use of an antisense oligonucleotide of any of embodiments 1 to 44 or the composition of embodiment 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type III.

Embodiment 55

[0097] Use of an antisense oligonucleotide of any of embodiments 1 to 44 or the composition of embodiment 45 for the preparation of a medicament for the treatment of Spinal Muscular Atrophy Type IV.

Embodiment 56

[0098] Use of an antisense oligonucleotide of any of embodiments 1 to 45 for the preparation of a medicament for the treatment of spinal muscular atrophy.

DETAILED DESCRIPTION

[0099] It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed. Herein, the use of the singular includes the plural unless specifically stated otherwise. As used herein, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including" as well as other forms, such as "includes" and "included", is not limiting. Also, terms such as "element" or "component" encompass both elements and components comprising one unit and elements and components that comprise more than one subunit, unless specifically stated otherwise.

[0100] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference in their entirety for any purpose.

I. DEFINITIONS

[0101] Unless specific definitions are provided, the nomenclature utilized in connection with, and the procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques may be used for chemical synthesis, and chemical analysis. Certain such techniques and procedures may be found for example in "Carbohydrate Modifications in Antisense Research" Edited by Sangvi and Cook, American Chemical Society, Washington D.C., 1994; "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., 18th edition, 1990; and "Antisense Drug Technology, Principles, Strategies, and Applications" Edited by Stanley T. Crooke, CRC Press, Boca Raton, Fla.; and Sambrook et al., "Molecular Cloning, A laboratory Manual," 2.sup.nd Edition, Cold Spring Harbor Laboratory Press, 1989, which are hereby incorporated by reference for any purpose. Where permitted, all patents, applications, published applications and other publications and other data referred to throughout in the disclosure herein are incorporated by reference in their entirety.

[0102] Unless otherwise indicated, the following terms have the following meanings:

[0103] "Nucleoside" means a compound comprising a heterocyclic base moiety and a sugar moiety. Nucleosides include, but are not limited to, naturally occurring nucleosides, modified nucleosides, and nucleosides having mimetic bases and/or sugar groups. Nucleosides may be modified with any of a variety of substituents.

[0104] "Sugar moiety" means a natural or modified sugar or sugar surrogate.

[0105] "Natural sugar" means a ribofuranose moiety of DNA (2'-H) or RNA (2'-OH).

[0106] "Modified sugar" means a ribofuranose moiety comprising at least one substituent other than that of a natural sugar.

[0107] "Sugar surrogate" means a structure other than a ribofuranose ring which is capable of substituting for the sugar of a nucleoside. Examples of sugar surrogates include, but are not limited to, open ring systems, 6-membered rings, sugars in which the oxygen is replace with, for example, sulfur or nitrogen. For example, sugar surrogates include, but are not limited to morpholinos and 4'-thio-containing sugars.

[0108] "Nucleobase" means the heterocyclic base portion of a nucleoside. Nucleobases may be naturally occurring or may be modified. In certain embodiments, a nucleobase may comprise any atom or group of atoms capable of hydrogen bonding to a nucleobase of another nucleic acid.

[0109] "Nucleotide" means a nucleoside comprising a phosphate linking group. As used herein, nucleosides include nucleotides.

[0110] "Modified nucleoside" a nucleoside comprising at least one modification compared to naturally occurring RNA or DNA nucleosides. Such modification may be at the sugar moiety and/or at the nucleobase.

[0111] "Bicyclic nucleoside" or "BNA" means a nucleoside wherein the sugar moiety of the nucleoside comprises a bridge connecting two carbon atoms of the sugar ring, thereby forming a bicyclic sugar moiety.

[0112] "4'-2' bicyclic nucleoside" means a bicyclic nucleoside comprising a furanose ring comprising a bridge connecting two carbon atoms of the furanose ring connects the 2' carbon atom and the 4' carbon atom of the sugar ring.

[0113] "2'-modified" or "2'-substituted" means a nucleoside comprising a sugar comprising a substituent at the 2' position other than H or OH.

[0114] "2'-OMe" or "2'-OCH.sub.3" or "2'-O-methyl" each means a nucleoside comprising a sugar comprising an --OCH.sub.3 group at the 2' position of the sugar ring.

[0115] "MOE" or "2'-MOE" or "2'-OCH.sub.2CH.sub.2OCH.sub.3" or "2'-O-methoxyethyl" each means a nucleoside comprising a sugar comprising a --OCH.sub.2CH.sub.2OCH.sub.3 group at the 2' position of the sugar ring.

[0116] "Oligonucleotide" means a compound comprising a plurality of linked nucleosides. In certain embodiments, one or more of the plurality of nucleosides is modified. In certain embodiments, an oligonucleotide comprises one or more ribonucleosides (RNA) and/or deoxyribonucleosides (DNA).

[0117] "Oligonucleoside" means an oligonucleotide in which none of the internucleoside linkages contains a phosphorus atom. As used herein, oligonucleotides include oligonucleosides.

[0118] "Modified oligonucleotide" means an oligonucleotide comprising at least one modified nucleoside and/or at least one modified internucleoside linkage.

[0119] "Internucleoside linkage" means a covalent linkage between adjacent nucleosides of an oligonucleotide.

[0120] "Naturally occurring internucleoside linkage" means a 3' to 5' phosphodiester linkage.

[0121] "Modified internucleoside linkage" means any internucleoside linkage other than a naturally occurring internucleoside linkage.

[0122] "Oligomeric compound" means a compound comprising an oligonucleotide. In certain embodiments, an oligomeric compound consists of an oligonucleotide. In certain embodiments, an oligomeric compound further comprises one or more conjugate and/or terminal groups.

[0123] "Antisense compound" means an oligomeric compound, at least a portion of which is at least partially complementary to a target nucleic acid to which it hybridizes, wherein such hybridization results at least one antisense activity.

[0124] "Antisense oligonucleotide" means an antisense compound wherein the oligomeric compound consists of an oligonucleotide.

[0125] "Antisense activity" refers to any detectable and/or measurable effect attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, such antisense activity is an increase or decrease in an amount of a nucleic acid or protein. In certain embodiments, such antisense activity is a change in the ratio of splice variants of a nucleic acid or protein. In certain embodiments, such antisense activity is a phenotypic change in a cell and/or subject.

[0126] "Detecting" or "measuring" of antisense activity may be direct or indirect. For example, in certain embodiments, antisense activity is assessed by detecting and/or measuring the amount of target nucleic acid or protein or the relative amounts of splice variants of a target nucleic acid or protein. In certain embodiments, antisense activity is detected by observing a phenotypic change in a cell or animal. In connection with any activity, response, or effect, the terms "detecting" and "measuring," indicate that a test for detecting or measuring is performed. Such detection and/or measuring may include values of zero. Thus, if a test for detection or measuring results in a finding of no activity (activity of zero), the step of detecting or measuring the activity has nevertheless been performed.

[0127] "Target nucleic acid" refers to any nucleic acid molecule the expression, amount, or activity of which is capable of being modulated by an antisense compound.

[0128] "Target mRNA" means a pre-selected RNA molecule that encodes a protein.

[0129] "Target pre-mRNA" means a pre-selected RNA transcript that has not been fully processed into mRNA. Notably, pre-mRNA includes one or more intron.

[0130] "Target protein" means a protein encoded by a target nucleic acid.

[0131] "Modulation" means to a perturbation of function or activity. In certain embodiments, modulation means an increase in gene expression. In certain embodiments, modulation means a decrease in gene expression.

[0132] "Expression" means any functions and steps by which a gene's coded information is converted into structures present and operating in a cell.

[0133] "Nucleobase sequence" means the order of contiguous nucleobases, in a 5' to 3' orientation, independent of any sugar, linkage, and/or nucleobase modification.

[0134] "Contiguous nucleobases" means nucleobases immediately adjacent to each other in a nucleic acid.

[0135] "Nucleobase complementarity" means the ability of two nucleobases to pair non-covalently via hydrogen bonding.

[0136] "Complementary" means that a first nucleic acid is capable of hybridizing to a second nucleic acid under stringent hybridization conditions. For example, an antisense compound is complementary to its target nucleic acid if it is capable of hybridizing to the target nucleic acid under stringent hybridization conditions.

[0137] "Fully complementary" means each nucleobase of a first nucleic acid is capable of pairing with a nucleobase at each corresponding contiguous position in a second nucleic acid.

[0138] "Percent complementarity" of an antisense compound means the percentage of nucleobases of the antisense compound that are complementary to an equal-length portion of a target nucleic acid. Percent complementarity is calculated by dividing the number of nucleobases of the antisense oligonucleotide that are complementary to nucleobases at corresponding contiguous positions in the target nucleic acid by the total length of the antisense compound.

[0139] "Percent identity" means the number of nucleobases in first nucleic acid that are identical to nucleobases at corresponding positions in a second nucleic acid, divided by the total number of nucleobases in the first nucleic acid.

[0140] "Hybridize" means the annealing of complementary nucleic acids that occurs through nucleobase complementarity.

[0141] "Mismatch" means a nucleobase of a first nucleic acid that is not capable of pairing with a nucleobase at a corresponding position of a second nucleic acid.

[0142] "Identical nucleobase sequence" means having the same nucleobase sequence, independent of any chemical modifications to the nucleosides.

[0143] "Different modifications" or "differently modified" refer to nucleosides or internucleoside linkages that have different nucleoside modifications or internucleoside linkages than one another, including absence of modifications. Thus, for example, a MOE nucleoside and an unmodified DNA nucleoside are "differently modified," even though the DNA nucleoside is unmodified. Likewise, DNA and RNA are "differently modified," even though both are naturally-occurring unmodified nucleosides. Nucleosides that are the same but for comprising different nucleobases are not differently modified, unless otherwise indicated. For example, a nucleoside comprising a 2'-OMe modified sugar and an adenine nucleobase and a nucleoside comprising a 2'-OMe modified sugar and a thymine nucleobase are not differently modified.

[0144] "The same modifications" refer to nucleosides and internucleoside linkages (including unmodified nucleosides and internucleoside linkages) that are the same as one another. Thus, for example, two unmodified DNA nucleoside have "the same modification," even though the DNA nucleoside is unmodified.

[0145] "Type of modification" or nucleoside of a "type" means the modification of a nucleoside and includes modified and unmodified nucleosides. Accordingly, unless otherwise indicated, a "nucleoside having a modification of a first type" may be an unmodified nucleoside.

[0146] "Separate regions" of an oligonucleotide means a portion of an oligonucleotide wherein the nucleosides and internucleoside linkages within the region all comprise the same modifications; and the nucleosides and/or the internucleoside linkages of any neighboring portions include at least one different modification.

[0147] "Motif" means a pattern of modified and/or unmodified nucleobases, sugars, and/or internucleoside linkages in an oligonucleotide.

[0148] "Fully modified oligonucleotide" means each nucleobase, each sugar, and/or each internucleoside linkage is modified.

[0149] "Uniformly modified oligonucleotide" means each nucleobase, each sugar, and/or each internucleoside linkage has the same modification throughout the modified oligonucleotide.

[0150] "Alternating motif" means an oligonucleotide or a portion thereof, having at least four separate regions of modified nucleosides in a pattern (AB).sub.nA.sub.m where A represents a region of nucleosides having a first type of modification; B represent a region of nucleosides having a different type of modification; n is 2-15; and m is 0 or 1. Thus, in certain embodiments, alternating motifs include 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 or more alternating regions. In certain embodiments, each A region and each B region independently comprises 1-4 nucleosides.

[0151] "Subject" means a human or non-human animal selected for treatment or therapy.

[0152] "Subject in need thereof" means a subject identified as in need of a therapy or treatment. In such embodiments, a subject has one or more indications of having or developing SMA.

[0153] "Administering" means providing a pharmaceutical agent or composition to a subject, and includes, but is not limited to, administering by a medical professional and self-administering.

[0154] "Parenteral administration," means administration through injection or infusion. Parenteral administration includes, but is not limited to, subcutaneous administration, intravenous administration, or intramuscular administration.

[0155] "Systemic administration" means administration to an area other than the intended locus of activity. Examples or systemic administration are subcutaneous administration and intravenous administration, and intraperitoneal administration.

[0156] "Subcutaneous administration" means administration just below the skin.

[0157] "Intravenous administration" means administration into a vein.

[0158] "Cerebrospinal fluid" or "CSF" means the fluid filling the space around the brain and spinal cord.

[0159] "Administration into the cerebrospinal fluid" means any administration that delivers a substance directly into the CSF.

[0160] "Intracerebroventricular" or "ICV" mean administration into the ventricular system of the brain.

[0161] "Intrathecal" or "IT" means administration into the CSF under the arachnoid membrane which covers the brain and spinal cord. IT injection is performed through the theca of the spinal cord into the subarachnoid space, where a pharmaceutical agent is injected into the sheath surrounding the spinal cord.

[0162] "Induction phase" means a dosing phase during which administration is initiated and steady state concentrations of active pharmaceutical agent are achieved in a target tissue. For example, an induction phase is a dosing phase during which steady state concentrations of antisense oligonucleotide are achieved in liver.

[0163] "Maintenance phase" means a dosing phase after target tissue steady state concentrations of drug have been achieved.

[0164] "Duration" means the period of time during which an activity or event continues. For example, the duration of an induction phase is the period of time during which induction doses are administered.

[0165] "Maintenance dose" means a dose administered at a single administration during the maintenance phase. As used herein, "induction dose" means a dose administered at a single administration during the induction phase.

[0166] "Co-administration" means administration of two or more pharmaceutical agents to a subject. The two or more pharmaceutical agents may be in a single pharmaceutical composition, or may be in separate pharmaceutical compositions. Each of the two or more pharmaceutical agents may be administered through the same or different routes of administration. Co-administration encompasses administration in parallel or sequentially.

[0167] "Therapy" means a disease treatment method. In certain embodiments, therapy includes, but is not limited to surgical therapies, chemical therapies, and physical interventions, such as assisted respiration, feeding tubes, and physical therapy for the purpose of increasing strength.

[0168] "Treatment" means the application of one or more specific procedures used for the cure or amelioration of a disease. In certain embodiments, the specific procedure is the administration of one or more pharmaceutical agents.

[0169] "Amelioration" means a lessening of severity of at least one indicator of a condition or disease. In certain embodiments, amelioration includes a delay or slowing in the progression of one or more indicators of a condition or disease. The severity of indicators may be determined by subjective or objective measures which are known to those skilled in the art.

[0170] "Prevent the onset of" means to prevent the development a condition or disease in a subject who is at risk for developing the disease or condition. In certain embodiments, a subject at risk for developing the disease or condition receives treatment similar to the treatment received by a subject who already has the disease or condition.

[0171] "Delay the onset of" means to delay the development of a condition or disease in a subject who is at risk for developing the disease or condition.

[0172] "Slow the progression of" means that the severity of at least one symptom associated with a disease or condition worsens less quickly.

[0173] "Exon 7 amino acids" means the portion of an SMN protein that correspond to exon 7 of the SMN RNA. Exon 7 amino acids are present in SMN protein expressed from SMN RNA where exon 7 was not excluded during splicing.

[0174] "SMN protein" means normal full length survival motor neuron protein. SMN may be expressed from either an SMN1 gene or from an SMN2 gene, provided that exon 7 is present in the mature mRNA and the exon 7 amino acids are present in the SMN protein.

[0175] "Dose" means a specified quantity of a pharmaceutical agent provided in a single administration or over a specified amount of time. In certain embodiments, a dose may be administered in two or more boluses, tablets, or injections. For example, in certain embodiments, where subcutaneous or inrathecal or ICV administration is desired, the desired dose requires a volume not easily accommodated by a single injection. In such embodiments, two or more injections may be used to achieve the desired dose. In the setting of continuous infusion, dose may be expressed as the quantity of a pharmaceutical agent delivered per unit of time.

[0176] "Dosage unit" means a form in which a pharmaceutical agent is provided. In certain embodiments, a dosage unit is a vial containing lyophilized oligonucleotide. In certain embodiments, a dosage unit is a vial containing reconstituted oligonucleotide.

[0177] "Therapeutically effective amount" means an amount of a pharmaceutical agent that provides a therapeutic benefit to an animal.

[0178] "Pharmaceutical composition" means a mixture of substances suitable for administering to an individual that includes a pharmaceutical agent. For example, a pharmaceutical composition may comprise a modified oligonucleotide and a sterile aqueous solution.

[0179] "Acceptable safety profile" means a pattern of side effects that is within clinically acceptable limits.

[0180] "Side effect" means a physiological response attributable to a treatment other than desired effects.

1. Certain Modified Oligonucleotides

[0181] In certain embodiments, the present invention provides methods and compositions involving antisense oligonucleotides comprising one or more modification compared to oligonucleotides of naturally occurring oligomers, such as DNA or RNA. Such modified antisense oligonucleotides may possess one or more desirable properties. Certain such modifications alter the antisense activity of the antisense oligonucleotide, for example by increasing affinity of the antisense oligonucleotide for its target nucleic acid, increasing its resistance to one or more nucleases, and/or altering the pharmacokinetics or tissue distribution of the oligonucleotide. In certain embodiments, such modified antisense oligonucleotides comprise one or more modified nucleosides and/or one or more modified nucleoside linkages and/or one or more conjugate groups.

[0182] a. Certain Modified Nucleosides

[0183] In certain embodiments, antisense oligonucleotides comprise one or more modified nucleosides. Such modified nucleosides may include a modified sugar and/or a modified nucleobase. In certain embodiments, incorporation of such modified nucleosides in an oligonucleotide results in increased affinity for a target nucleic acid and/or increased stability, including but not limited to, increased resistance to nuclease degradation, and or improved toxicity and/or uptake properties of the modified oligonucleotide.

[0184] i. Certain Nucleobases

[0185] The naturally occurring base portion of nucleosides are heterocyclic base, typically purines and pyrimidines. In addition to "unmodified" or "natural" nucleobases such as the purine nucleobases adenine (A) and guanine (G), and the pyrimidine nucleobases thymine (T), cytosine (C) and uracil (U), many modified nucleobases or nucleobase mimetics known to those skilled in the art are amenable to incorporation into the compounds described herein. In certain embodiments, a modified nucleobase is a nucleobase that is fairly similar in structure to the parent nucleobase, such as for example a 7-deaza purine, a 5-methyl cytosine, or a G-clamp. In certain embodiments, nucleobase mimetic include more complicated structures, such as for example a tricyclic phenoxazine nucleobase mimetic. Methods for preparation of the above noted modified nucleobases are well known to those skilled in the art.

[0186] ii. Certain Modified Sugars and Sugar Surrogates

[0187] Antisense oligonucleotides of the present invention can optionally contain one or more nucleosides wherein the sugar moiety is modified, compared to a natural sugar. Oligonucleotides comprising such sugar modified nucleosides may have enhanced nuclease stability, increased binding affinity or some other beneficial biological property. Such modifications include without limitation, addition of substituent groups, bridging of non-geminal ring atoms to form a bicyclic nucleic acid (BNA), replacement of the ribosyl ring oxygen atom with S, N(R), or C(R.sub.1)(R).sub.2 (R.dbd.H, C.sub.1-C.sub.12 alkyl or a protecting group) and combinations of these such as for example a 2'-F-5'-methyl substituted nucleoside (see PCT International Application WO 2008/101157 Published on Aug. 21, 2008 for other disclosed 5',2'-bis substituted nucleosides) or replacement of the ribosyl ring oxygen atom with S with further substitution at the 2'-position (see published U.S. Patent Application US2005-0130923, published on Jun. 16, 2005) or alternatively 5'-substitution of a BNA (see PCT International Application WO 2007/134181 Published on Nov. 22, 2007 wherein LNA is substituted with for example a 5'-methyl or a 5'-vinyl group).

[0188] Examples of nucleosides having modified sugar moieties include without limitation nucleosides comprising 5'-vinyl, 5'-methyl (R or S), 4'-S, 2'-F, 2'-OCH.sub.3 and 2'-O(CH.sub.2).sub.2OCH.sub.3 substituent groups. The substituent at the 2' position can also be selected from allyl, amino, azido, thio, O-allyl, O--C.sub.1-C.sub.10 alkyl, OCF.sub.3, O(CH.sub.2).sub.2SCH.sub.3, O(CH.sub.2).sub.2--O--N(R.sub.m)(R.sub.n), and O--CH.sub.2--C(.dbd.O)--N(R.sub.m)(R.sub.n), where each R.sub.m and R.sub.n is, independently, H or substituted or unsubstituted C.sub.1-C.sub.10 alkyl.

[0189] Examples of bicyclic nucleic acids (BNAs) include without limitation nucleosides comprising a bridge between the 4' and the 2' ribosyl ring atoms. In certain embodiments, antisense compounds provided herein include one or more BNA nucleosides wherein the bridge comprises one of the formulas: 4'-.beta.-D-(CH.sub.2)--O-2' (.beta.-D-LNA); 4'-(CH.sub.2)--S-2'; 4'-.alpha.-L-(CH.sub.2)--O-2' (.alpha.-L-LNA); 4'-(CH.sub.2).sub.2--O-2' (ENA); 4'-C(CH.sub.3).sub.2--O-2' (see PCT/US2008/068922); 4'-CH(CH.sub.3)--O-2' and 4'-C--H(CH.sub.2OCH.sub.3)--O-2' (see U.S. Pat. No. 7,399,845, issued on Jul. 15, 2008); 4'-CH.sub.2--N(OCH.sub.3)-2' (see PCT/US2008/064591); 4'-CH.sub.2--O--N(CH.sub.3)-2' (see published U.S. Patent Application US2004-0171570, published Sep. 2, 2004); 4'-CH.sub.2--N(R)--O-2' (see U.S. Pat. No. 7,427,672, issued on Sep. 23, 2008); 4'-CH.sub.2--C(CH.sub.3)-2' and 4'-CH.sub.2--C(.dbd.CH.sub.2)-2' (see PCT/US2008/066154); and wherein R is, independently, H, C.sub.1-C.sub.12 alkyl, or a protecting group.

[0190] In certain embodiments, the present invention provides modified nucleosides comprising modified sugar moieties that are not bicyclic sugar moieties. Certain such modified nucleosides are known. In certain embodiments, the sugar ring of a nucleoside may be modified at any position. Examples of sugar modifications useful in this invention include, but are not limited to compounds comprising a sugar substituent group selected from: OH, F, O-alkyl, S-alkyl, N-alkyl, or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C.sub.1 to C.sub.10 alkyl or C.sub.2 to C.sub.10 alkenyl and alkynyl. In certain such embodiments, such substituents are at the 2' position of the sugar.

[0191] In certain embodiments, modified nucleosides comprise a substituent at the 2' position of the sugar. In certain embodiments, such substituents are selected from among: a halide (including, but not limited to F), allyl, amino, azido, thio, O-allyl, O--C.sub.1-C.sub.10 alkyl, --OCF.sub.3, O--(CH.sub.2).sub.2--O--CH.sub.3, 2'-O(CH.sub.2).sub.2SCH.sub.3, O--(CH.sub.2).sub.2--O--N(R.sub.m)(R.sub.n), or O--CH2-C(.dbd.O)--N(R.sub.m)(R.sub.n), where each R.sub.m and R.sub.n is, independently, H or substituted or unsubstituted C.sub.1-C.sub.10 alkyl.

[0192] In certain embodiments, modified nucleosides suitable for use in the present invention are: 2-methoxyethoxy, 2'-O-methyl (2'-O--CH.sub.3), 2'-fluoro (2'-F).

[0193] In certain embodiments, modified nucleosides having a substituent group at the 2'-position selected from: O[(CH.sub.2).sub.nO].sub.mCH.sub.3, O(CH.sub.2).sub.nNH.sub.2, O(CH.sub.2).sub.nCH.sub.3, O(CH.sub.2).sub.nONH.sub.2, OCH.sub.2C(.dbd.O)N(H)CH.sub.3, and O(CH.sub.2).sub.nON[(CH.sub.2)CH.sub.3].sub.2, where n and m are from 1 to about 10. Other 2'-sugar substituent groups include: C.sub.1 to C.sub.10 alkyl, substituted alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH.sub.3, OCN, Cl, Br, CN, CF.sub.3, OCF.sub.3, SOCH.sub.3, SO.sub.2CH.sub.3, ONO.sub.2, NO.sub.2, N.sub.3, NH.sub.2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving pharmacokinetic properties, or a group for improving the pharmacodynamic properties of an oligomeric compound, and other substituents having similar properties.

[0194] In certain embodiments, modified nucleosides comprise a 2'-MOE side chain (Baker et al., J. Biol. Chem., 1997, 272, 11944-12000). Such 2'-MOE substitution have been described as having improved binding affinity compared to unmodified nucleosides and to other modified nucleosides, such as 2'-O-methyl, O-propyl, and O-aminopropyl. Oligonucleotides having the 2'-MOE substituent also have been shown to be antisense inhibitors of gene expression with promising features for in vivo use (Martin, P., Helv. Chim. Acta, 1995, 78, 486-504; Altmann et al., Chimia, 1996, 50, 168-176; Altmann et al., Biochem. Soc. Trans., 1996, 24, 630-637; and Altmann et al., Nucleosides Nucleotides, 1997, 16, 917-926).

[0195] In certain embodiments, 2'-sugar substituent groups are in either the arabino (up) position or ribo (down) position. In certain such embodiments, a 2'-arabino modification is 2'-F arabino (FANA). Similar modifications can also be made at other positions on the sugar, particularly the 3' position of the sugar on a 3' terminal nucleoside or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide.

[0196] In certain embodiments, nucleosides suitable for use in the present invention have sugar surrogates such as cyclobutyl in place of the ribofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920, each of which is herein incorporated by reference in its entirety.

[0197] In certain embodiments, the present invention provides nucleosides comprising a modification at the 2'-position of the sugar. In certain embodiments, the invention provides nucleosides comprising a modification at the 5'-position of the sugar. In certain embodiments, the invention provides nucleosides comprising modifications at the 2'-position and the 5'-position of the sugar. In certain embodiments, modified nucleosides may be useful for incorporation into oligonucleotides. In certain embodiment, modified nucleosides are incorporated into oligonucleosides at the 5'-end of the oligonucleotide.

[0198] b. Certain Internucleoside Linkages

[0199] Antisense oligonucleotides of the present invention can optionally contain one or more modified internucleoside linkages. The two main classes of linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus containing linkages include, but are not limited to, phosphodiesters (P.dbd.O), phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates (P.dbd.S). Representative non-phosphorus containing linking groups include, but are not limited to, methylenemethylimino (--CH2-N(CH3)-O--CH2-), thiodiester (--O--C(O)--S--), thionocarbamate (--O--C(O)(NH)--S--); siloxane (--O--Si(H)2-O--); and N,N'-dimethylhydrazine (--CH2-N(CH3)-N(CH3)-). Oligonucleotides having non-phosphorus linking groups are referred to as oligonucleosides. Modified linkages, compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotides. In certain embodiments, linkages having a chiral atom can be prepared as racemic mixtures, as separate enantiomers. Representative chiral linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing linkages are well known to those skilled in the art.

[0200] The antisense oligonucleotides described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), such as for sugar anomers, or as (D) or (L) such as for amino acids et al. Included in the antisense compounds provided herein are all such possible isomers, as well as their racemic and optically pure forms.

[0201] In certain embodiments, antisense oligonucleotides have at least one modified internucleoside linkage. In certain embodiments, antisense oligonucleotides have at least 2 modified internucleoside linkages. In certain embodiments, antisense oligonucleotides have at least 3 modified internucleoside linkages. In certain embodiments, antisense oligonucleotides have at least 10 modified internucleoside linkages. In certain embodiments, each internucleoside linkage of an antisense oligonucleotide is a modified internucleoside linkage. In certain embodiments, such modified internucleoside linkages are phosphorothioate linkages.

[0202] c. Certain Internucleoside Linkage Motifs

[0203] In certain embodiments, oligonucleotides comprise modified internucleoside linkages arranged along the oligonucleotide or region thereof in a defined pattern or modified internucleoside linkage motif.

[0204] In certain embodiments, oligonucleotides comprise a region having an alternating internucleoside linkage motif. In certain embodiments, oligonucleotides of the present invention comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligonucleotide comprises a region that is uniformly linked by phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide is uniformly linked by phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate.

[0205] In certain embodiments, the oligonucleotide comprises at least 5 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 7 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 9 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 11 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 12 phosphorothioate internucleoside linkages.

[0206] In certain embodiments, the oligonucleotide comprises at least one block of at least 2 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 3 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 4 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 5 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.

[0207] In certain embodiments, the oligonucleotide comprises at least one block of at least 2 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 3 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 4 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 5 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.

[0208] In certain embodiments, the oligonucleotide comprises at least one block of at least 2 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 3 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 4 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 5 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphodiester internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphodiester internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.

[0209] In certain embodiments, the oligonucleotide comprises at least one block of at least 2 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 3 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 4 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 5 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphodiester internucleoside linkages, wherein the remainder of the internucleoside linkages in the oligonucleotide comprise phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide. In certain such embodiments, at least one such block is located at the center of the oligonucleotide.

[0210] In certain embodiments, it is desirable to arrange the number of phosphorothioate internucleoside linkages and phosphodiester internucleoside linkages to maintain nuclease resistance. In certain embodiments, it is desirable to arrange the number and position of phosphorothioate internucleoside linkages and the number and position of phosphodiester internucleoside linkages to maintain nuclease resistance. In certain embodiments, the number of phosphorothioate internucleoside linkages may be decreased and the number of phosphodiester internucleoside linkages may be increased. In certain embodiments, the number of phosphorothioate internucleoside linkages may be decreased and the number of phosphodiester internucleoside linkages may be increased while still maintaining nuclease resistance. In certain embodiments it is desirable to decrease the number of phosphorothioate internucleoside linkages while retaining nuclease resistance. In certain embodiments it is desirable to increase the number of phosphodiester internucleoside linkages while retaining nuclease resistance.

[0211] In certain embodiments, an oligomeric compound has an internucleoside linkage motif selected from the table below, wherein each "N" represents a nucleoside, each subscript "s" represents a phosphorothioate internucleoside linkage, and each subscript "o" represents a phosphodiester internucleoside linkage:

TABLE-US-00001 Internucleoside Linkage Motifs NsNsNsNsNsNsNsNsNsNsNsNsNsNsN NsNoNsNoNsNoNsNoNsNoNsNoNsNsN NsNsNoNsNsNoNsNsNoNsNoNoNsNsN NsNsNoNsNsNoNsNsNoNsNsNoNsNsN NsNsNsNoNsNsNsNoNsNsNsNoNsNsN NsNsNsNsNoNsNsNsNsNoNsNsNsNsN NsNsNsNsNsNsNsNsNsNsNsNsNsNsNsNsNsNsNsN NsNsNoNoNoNsNsNsNoNoNoNoNoNoNoNsNsN NsNsNoNoNoNoNoNoNoNsNsNsNoNoNoNsNsN NsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN NsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNoNsNsN NsNoNoNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNoNoNsNsN NsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsNoNsNsN NsNoNsNsNoNsNsNsNoNsNsNsNoNsNsNsNoNsNsN NsNoNsNsNsNsNoNsNsNsNsNoNsNsNsNsNoNsNsN NsNsNoNoNoNoNoNoNoNoNoNoNoNoNoNsNsN NsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsN NsNsNsNoNsNoNsNoNsNoNsNoNsNoNsNsNsN NsNoNoNoNsNsNsNsNsNsNsNsNoNoNoNsNsN NsNsNsNoNsNoNsNoNsNoNsNoNsNoNsNsNsN NsNoNoNoNsNsNsNsNsNsNsNsNoNoNoNsNsN NsNsNoNsNoNsNoNsNoNsNoNsNoNsNoNsNsN NsNsNoNsNoNsNoNsNoNsNoNsNoNoNoNsNsN NsNoNoNsNoNoNsNoNoNsNoNoNsNoNoNsNsN NsNoNoNoNoNsNsNsNsNsNsNsNoNoNoNsNsN NsNoNoNoNoNsNsNsNsNsNsNoNoNoNoNsNsN NsNoNoNoNoNoNsNsNsNsNsNoNoNoNoNsNsN NsNoNoNoNoNoNsNsNsNsNoNoNoNoNoNsNsN NsNsNsNoNoNoNoNoNoNoNoNoNoNoNsNsNsN NsNsNsNsNoNoNoNoNoNoNoNoNoNoNsNsNsN NsNsNsNsNoNoNoNoNoNoNoNoNoNsNsNsNsN NsNsNsNsNsNoNoNoNoNoNoNoNoNsNsNsNsN NsNsNsNsNsNoNoNoNoNoNoNoNsNsNsNsNsN NsNsNsNsNsNsNoNoNoNoNoNoNsNsNsNsNsN NsNsNsNsNsNsNoNoNoNoNoNsNsNsNsNsNsN NsNoNoNoNsNsNsNsNsNsNsNsNsNoNoNsNsN

[0212] In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide improves therapeutic index. In certain embodiments, pharmaceutical compositions having 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages have improved therapeutic indices. In certain embodiments, pharmaceutical compositions having 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages and having improved therapeutic indices allows one having skill in the art to administer the pharmaceutical compositions of the present disclosure more infrequently. In certain embodiments, pharmaceutical compositions of the present disclosure having improved therapeutic indices allows one having skill in the art to allow longer intervals between the first dose and the second dose.

[0213] In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0214] In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 2 or 7, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0215] In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 6 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0216] In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 7 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0217] In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 8 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0218] In certain embodiments, the inclusion of 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at infrequent intervals. In certain embodiments, the inclusion of 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 3 month intervals. In certain embodiments, the inclusion of 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 4 month intervals. In certain embodiments, the inclusion of 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 5 month intervals. In certain embodiments, the inclusion of 3, 4, 5, 6, 7, 8, or 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 6 month intervals. In certain embodiments, the inclusion of 9 phosphodiester internucleoside linkages into a modified oligonucleotide, wherein the modified oligonucleotide has the nucleobase sequence of SEQ ID NO.: 1, allows one having skill in the art to administer the modified oligonucleotide or a pharmaceutical composition thereof at greater than 7 month intervals.

[0219] d. Lengths

[0220] In certain embodiments, the present invention provides antisense oligonucleotides of any of a variety of ranges of lengths. In certain embodiments, the invention provides antisense compounds or antisense oligonucleotides comprising or consisting of X-Y linked nucleosides, where X and Y are each independently selected from 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, and 50; provided that X<Y. For example, in certain embodiments, the invention provides antisense compounds or antisense oligonucleotides comprising or consisting of: 8-9, 8-10, 8-11, 8-12, 8-13, 8-14, 8-15, 8-16, 8-17, 8-18, 8-19, 8-20, 8-21, 8-22, 8-23, 8-24, 8-25, 8-26, 8-27, 8-28, 8-29, 8-30, 9-10, 9-11, 9-12, 9-13, 9-14, 9-15, 9-16, 9-17, 9-18, 9-19, 9-20, 9-21, 9-22, 9-23, 9-24, 9-25, 9-26, 9-27, 9-28, 9-29, 9-30, 10-11, 10-12, 10-13, 10-14, 10-15, 10-16, 10-17, 10-18, 10-19, 10-20, 10-21, 10-22, 10-23, 10-24, 10-25, 10-26, 10-27, 10-28, 10-29, 10-30, 11-12, 11-13, 11-14, 11-15, 11-16, 11-17, 11-18, 11-19, 11-20, 11-21, 11-22, 11-23, 11-24, 11-25, 11-26, 11-27, 11-28, 11-29, 11-30, 12-13, 12-14, 12-15, 12-16, 12-17, 12-18, 12-19, 12-20, 12-21, 12-22, 12-23, 12-24, 12-25, 12-26, 12-27, 12-28, 12-29, 12-30, 13-14, 13-15, 13-16, 13-17, 13-18, 13-19, 13-20, 13-21, 13-22, 13-23, 13-24, 13-25, 13-26, 13-27, 13-28, 13-29, 13-30, 14-15, 14-16, 14-17, 14-18, 14-19, 14-20, 14-21, 14-22, 14-23, 14-24, 14-25, 14-26, 14-27, 14-28, 14-29, 14-30, 15-16, 15-17, 15-18, 15-19, 15-20, 15-21, 15-22, 15-23, 15-24, 15-25, 15-26, 15-27, 15-28, 15-29, 15-30, 16-17, 16-18, 16-19, 16-20, 16-21, 16-22, 16-23, 16-24, 16-25, 16-26, 16-27, 16-28, 16-29, 16-30, 17-18, 17-19, 17-20, 17-21, 17-22, 17-23, 17-24, 17-25, 17-26, 17-27, 17-28, 17-29, 17-30, 18-19, 18-20, 18-21, 18-22, 18-23, 18-24, 18-25, 18-26, 18-27, 18-28, 18-29, 18-30, 19-20, 19-21, 19-22, 19-23, 19-24, 19-25, 19-26, 19-29, 19-28, 19-29, 19-30, 20-21, 20-22, 20-23, 20-24, 20-25, 20-26, 20-27, 20-28, 20-29, 20-30, 21-22, 21-23, 21-24, 21-25, 21-26, 21-27, 21-28, 21-29, 21-30, 22-23, 22-24, 22-25, 22-26, 22-27, 22-28, 22-29, 22-30, 23-24, 23-25, 23-26, 23-27, 23-28, 23-29, 23-30, 24-25, 24-26, 24-27, 24-28, 24-29, 24-30, 25-26, 25-27, 25-28, 25-29, 25-30, 26-27, 26-28, 26-29, 26-30, 27-28, 27-29, 27-30, 28-29, 28-30, or 29-30 linked nucleosides.

[0221] In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 15 nucleosides in length. In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 16 nucleosides in length. In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 17 nucleosides in length. In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 18 nucleosides in length. In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 19 nucleosides in length. In certain embodiments, antisense compounds or antisense oligonucleotides of the present invention are 20 nucleosides in length.

[0222] e. Certain Oligonucleotide Motifs

[0223] In certain embodiments, antisense oligonucleotides have chemically modified subunits arranged in specific orientations along their length. In certain embodiments, antisense oligonucleotides of the invention are fully modified. In certain embodiments, antisense oligonucleotides of the invention are uniformly modified. In certain embodiments, antisense oligonucleotides of the invention are uniformly modified and each nucleoside comprises a 2'-MOE sugar moiety. In certain embodiments, antisense oligonucleotides of the invention are uniformly modified and each nucleoside comprises a 2'-OMe sugar moiety. In certain embodiments, antisense oligonucleotides of the invention are uniformly modified and each nucleoside comprises a morpholino sugar moiety.

[0224] In certain embodiments, oligonucleotides of the invention comprise an alternating motif. In certain such embodiments, the alternating modification types are selected from among 2'-MOE, 2'-F, a bicyclic sugar-modified nucleoside, and DNA (unmodified 2'-deoxy). In certain such embodiments, each alternating region comprises a single nucleoside.

[0225] In certain embodiments, oligonucleotides of the invention comprise one or more block of nucleosides of a first type and one or more block of nucleosides of a second type.

[0226] In certain embodiments, one or more alternating regions in an alternating motif include more than a single nucleoside of a type. For example, oligomeric compounds of the present invention may include one or more regions of any of the following nucleoside motifs:

[0227] Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.1;

[0228] Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.2;

[0229] Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1 Nu.sub.2;

[0230] Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.2;

[0231] Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1;

[0232] Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2;

[0233] Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1;

[0234] Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1;

[0235] Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1Nu.sub.2 Nu.sub.1 Nu.sub.1; or

[0236] Nu.sub.1 Nu.sub.2Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.1 Nu.sub.2 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.2 Nu.sub.1 Nu.sub.1;

wherein Nu.sub.1 is a nucleoside of a first type and Nu.sub.2 is a nucleoside of a second type. In certain embodiments, one of Nu.sub.1 and Nu.sub.2 is a 2'-MOE nucleoside and the other of Nu.sub.1 and Nu.sub.2 is a selected from: a 2'-OMe modified nucleoside, BNA, and an unmodified DNA or RNA nucleoside.

2. Oligomeric Compounds

[0237] In certain embodiments, the present invention provides oligomeric compounds. In certain embodiments, oligomeric compounds are comprised only of an oligonucleotide. In certain embodiments, an oligomeric compound comprises an oligonucleotide and one or more conjugate and/or terminal group. Such conjugate and/or terminal groups may be added to oligonucleotides having any of the chemical motifs discussed above. Thus, for example, an oligomeric compound comprising an oligonucleotide having region of alternating nucleosides may comprise a terminal group.

[0238] a. Certain Conjugate Groups

[0239] In certain embodiments, oligonucleotides of the present invention are modified by attachment of one or more conjugate groups. In general, conjugate groups modify one or more properties of the attached oligomeric compound including but not limited to, pharmacodynamics, pharmacokinetics, stability, binding, absorption, cellular distribution, cellular uptake, charge and clearance. Conjugate groups are routinely used in the chemical arts and are linked directly or via an optional conjugate linking moiety or conjugate linking group to a parent compound such as an oligomeric compound, such as an oligonucleotide. Conjugate groups includes without limitation, intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, thioethers, polyethers, cholesterols, thiocholesterols, cholic acid moieties, folate, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins and dyes. Certain conjugate groups have been described previously, for example: cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., do-decan-diol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10, 1111-1118; Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimie, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937).

[0240] In certain embodiments, a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic. Oligonucleotide-drug conjugates and their preparation are described in U.S. patent application Ser. No. 09/334,130.

[0241] Representative U.S. patents that teach the preparation of oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941.

[0242] Conjugate groups may be attached to either or both ends of an oligonucleotide (terminal conjugate groups) and/or at any internal position.

[0243] b. Terminal Groups

[0244] In certain embodiments, oligomeric compounds comprise terminal groups at one or both ends. In certain embodiments, a terminal group may comprise any of the conjugate groups discussed above. In certain embodiments, terminal groups may comprise additional nucleosides and/or inverted abasic nucleosides. In certain embodiments, a terminal group is a stabilizing group.

[0245] In certain embodiments, oligomeric compounds comprise one or more terminal stabilizing group that enhances properties such as for example nuclease stability. Included in stabilizing groups are cap structures. The terms "cap structure" or "terminal cap moiety," as used herein, refer to chemical modifications, which can be attached to one or both of the termini of an oligomeric compound. Certain such terminal modifications protect the oligomeric compounds having terminal nucleic acid moieties from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5'-terminus (5'-cap) or at the 3'-terminus (3'-cap) or can be present on both termini. (for more details see Wincott et al., International PCT publication No. WO 97/26270; Beaucage and Tyer, 1993, Tetrahedron 49, 1925; U.S. Patent Application Publication No. US 2005/0020525; and WO 03/004602.

[0246] In certain embodiments, one or more additional nucleosides is added to one or both terminal ends of an oligonucleotide of an oligomeric compound. Such additional terminal nucleosides are referred to herein as terminal-group nucleosides. In a double-stranded compound, such terminal-group nucleosides are terminal (3' and/or 5') overhangs. In the setting of double-stranded antisense compounds, such terminal-group nucleosides may or may not be complementary to a target nucleic acid. In certain embodiments, the terminal group is a non-nucleoside terminal group. Such non-terminal groups may be any terminal group other than a nucleoside.

[0247] c. Oligomeric Compound Motifs

In certain embodiments, oligomeric compounds of the present invention comprise a motif: T.sub.1-(Nu.sub.1).sub.n1-(Nu.sub.2).sub.n2-(Nu.sub.1).sub.n3-(Nu.sub.2).- sub.n4-(Nu.sub.1).sub.n5-T.sub.2, wherein: [0248] Nu.sub.1, is a nucleoside of a first type; [0249] Nu.sub.2, is a nucleoside of a second type; [0250] each of n1 and n5 is, independently from 0 to 3; [0251] the sum of n2 plus n4 is between 10 and 25; [0252] n3 is from 0 and 5; and [0253] each T.sub.1 and T.sub.2 is, independently, H, a hydroxyl protecting group, an optionally linked conjugate group or a capping group. In certain such embodiments, the sum of n2 and n4 is 13 or 14; n1 is 2; n3 is 2 or 3; and n5 is 2. In certain such embodiments, oligomeric compounds of the present invention comprise a motif selected from Table A.

TABLE-US-00002 [0253] TABLE A n1 n2 n3 n4 n5 2 16 0 0 2 2 2 3 11 2 2 5 3 8 2 2 8 3 5 2 2 11 3 2 2 2 9 3 4 2 2 10 3 3 2 2 3 3 10 2 2 4 3 9 2 2 6 3 7 2 2 7 3 6 2 2 8 6 2 2 2 2 2 12 2 2 3 2 11 2 2 4 2 10 2 2 5 2 9 2 2 6 2 8 2 2 7 2 7 2 2 8 2 6 2 2 9 2 5 2 2 10 2 4 2 2 11 2 3 2 2 12 2 2 2

[0254] Table A is intended to illustrate, but not to limit the present invention. The oligomeric compounds depicted in Table A each comprise 20 nucleosides. Oligomeric compounds comprising more or fewer nucleosides can easily by designed by selecting different numbers of nucleosides for one or more of n1-n5. In certain embodiments, Nu.sub.1 and Nu.sub.2 are each selected from among: 2'-MOE, 2'-OMe, DNA, and a bicyclic nucleoside.

3. Antisense

[0255] In certain embodiments, oligomeric compounds of the present invention are antisense compounds. Accordingly, in such embodiments, oligomeric compounds hybridize with a target nucleic acid, resulting in an antisense activity.

[0256] a. Hybridization

[0257] In certain embodiments, the invention provides antisense compounds that specifically hybridize to a target nucleic acid when there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.

[0258] Thus, "stringent hybridization conditions" or "stringent conditions" means conditions under which an antisense compounds hybridize to a target sequence, but to a minimal number of other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances, and "stringent conditions" under which antisense oligonucleotides hybridize to a target sequence are determined by the nature and composition of the antisense oligonucleotides and the assays in which they are being investigated.

[0259] It is understood in the art that incorporation of nucleotide affinity modifications may allow for a greater number of mismatches compared to an unmodified compound. Similarly, certain nucleobase sequences may be more tolerant to mismatches than other nucleobase sequences. One of ordinary skill in the art is capable of determining an appropriate number of mismatches between oligonucleotides, or between an antisense oligonucleotide and a target nucleic acid, such as by determining melting temperature (Tm). Tm or .DELTA.Tm can be calculated by techniques that are familiar to one of ordinary skill in the art. For example, techniques described in Freier et al. (Nucleic Acids Research, 1997, 25, 22: 4429-4443) allow one of ordinary skill in the art to evaluate nucleotide modifications for their ability to increase the melting temperature of an RNA:DNA duplex.

[0260] b. pre-mRNA Processing

[0261] In certain embodiments, antisense compounds provided herein are complementary to a pre-mRNA. In certain embodiments, such antisense compounds alter splicing of the pre-mRNA. In certain such embodiments, the ratio of one variant of a mature mRNA corresponding to a target pre-mRNA to another variant of that mature mRNA is altered. In certain such embodiments, the ratio of one variant of a protein expressed from the target pre-mRNA to another variant of the protein is altered. Certain oligomeric compounds and nucleobase sequences that may be used to alter splicing of a pre-mRNA may be found for example in U.S. Pat. No. 6,210,892; U.S. Pat. No. 5,627,274; U.S. Pat. Nos. 5,665,593; 5,916,808; U.S. Pat. No. 5,976,879; US2006/0172962; US2007/002390; US2005/0074801; US2007/0105807; US2005/0054836; WO 2007/090073; WO2007/047913, Hua et al., PLoS Biol 5(4):e73; Vickers et al., J. Immunol. 2006 Mar. 15; 176(6):3652-61; and Hua et al., American J. of Human Genetics (April 2008) 82, 1-15, each of which is hereby incorporated by reference in its entirety for any purpose. In certain embodiments antisense sequences that alter splicing are modified according to motifs of the present invention.

[0262] Antisense is an effective means for modulating the expression of one or more specific gene products and is uniquely useful in a number of therapeutic, diagnostic, and research applications. Provided herein are antisense compounds useful for modulating gene expression via antisense mechanisms of action, including antisense mechanisms based on target occupancy. In one aspect, the antisense compounds provided herein modulate splicing of a target gene. Such modulation includes promoting or inhibiting exon inclusion. Further provided herein are antisense compounds targeted to cis splicing regulatory elements present in pre-mRNA molecules, including exonic splicing enhancers, exonic splicing silencers, intronic splicing enhancers and intronic splicing silencers. Disruption of cis splicing regulatory elements is thought to alter splice site selection, which may lead to an alteration in the composition of splice products.

[0263] Processing of eukaryotic pre-mRNAs is a complex process that requires a multitude of signals and protein factors to achieve appropriate mRNA splicing. Exon definition by the spliceosome requires more than the canonical splicing signals which define intron-exon boundaries. One such additional signal is provided by cis-acting regulatory enhancer and silencer sequences. Exonic splicing enhancers (ESE), exonic splicing silencers (ESS), intronic splicing enhancers (ISE) and intron splicing silencers (ISS) have been identified which either repress or enhance usage of splice donor sites or splice acceptor sites, depending on their site and mode of action (Yeo et al. 2004, Proc. Natl. Acad. Sci. U.S.A. 101(44):15700-15705). Binding of specific proteins (trans factors) to these regulatory sequences directs the splicing process, either promoting or inhibiting usage of particular splice sites and thus modulating the ratio of splicing products (Scamborova et al. 2004, Mol. Cell. Biol. 24(5):1855-1869; Hovhannisyan and Carstens, 2005, Mol. Cell. Biol. 25(1):250-263; Minovitsky et al. 2005, Nucleic Acids Res. 33(2):714-724).

4. Pharmaceutical Compositions

[0264] In certain embodiments, the present invention provides pharmaceutical compositions comprising one or more antisense compound. In certain embodiments, such pharmaceutical composition comprises a sterile saline solution and one or more antisense compound. In certain embodiments, such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound.

[0265] In certain embodiments, antisense compounds may be admixed with pharmaceutically acceptable active and/or inert substances for the preparation of pharmaceutical compositions or formulations. Compositions and methods for the formulation of pharmaceutical compositions depend on a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.

[0266] In certain embodiments antisense compounds, can be utilized in pharmaceutical compositions by combining such oligomeric compounds with a suitable pharmaceutically acceptable diluent or carrier. A pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS). PBS is a diluent suitable for use in compositions to be delivered parenterally. Accordingly, in certain embodiments, employed in the methods described herein is a pharmaceutical composition comprising an antisense compound and a pharmaceutically acceptable diluent. In certain embodiments, the pharmaceutically acceptable diluent is PBS.

[0267] Pharmaceutical compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters. In certain embodiments, pharmaceutical compositions comprising antisense compounds comprise one or more oligonucleotide which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents. Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.

[0268] A prodrug can include the incorporation of additional nucleosides at one or both ends of an oligomeric compound which are cleaved by endogenous nucleases within the body, to form the active antisense oligomeric compound.

[0269] Lipid-based vectors have been used in nucleic acid therapies in a variety of methods. For example, in one method, the nucleic acid is introduced into preformed liposomes or lipoplexes made of mixtures of cationic lipids and neutral lipids. In another method, DNA complexes with mono- or poly-cationic lipids are formed without the presence of a neutral lipid.

[0270] Certain preparations are described in Akinc et al., Nature Biotechnology 26, 561-569 (1 May 2008), which is herein incorporated by reference in its entirety.

5. Administration to a Subject

[0271] In certain embodiments, pharmaceutical compositions comprising one or more antisense compound are administered to a subject. In certain embodiments, such pharmaceutical compositions are administered by injection. In certain embodiments, such pharmaceutical compositions are administered by infusion.

[0272] In certain embodiments, pharmaceutical compositions are administered by injection or infusion into the CSF. In certain such embodiments, pharmaceutical compositions are administered by direct injection or infusion into the spine. In certain embodiments, pharmaceutical compositions are administered by injection or infusion into the brain. In certain embodiments, pharmaceutical compositions are administered by intrathecal injection or infusion rather than into the spinal cord tissue itself. Without being limited as to theory, in certain embodiments, the antisense compound released into the surrounding CSF and may penetrate into the spinal cord parenchyma. An additional advantage of intrathecal delivery is that the intrathecal route mimics lumbar puncture administration (i.e., spinal tap) already in routine use in humans.

[0273] In certain embodiments, pharmaceutical compositions are administered by intracerebroventricular (ICV) injection or infusion. Intracerebroventricular, or intraventricular, delivery of a pharmaceutical composition comprising one or more antisense compounds may be performed in any one or more of the brain's ventricles, which are filled with cerebrospinal fluid (CSF). CSF is a clear fluid that fills the ventricles, is present in the subarachnoid space, and surrounds the brain and spinal cord. CSF is produced by the choroid plexuses and via the weeping or transmission of tissue fluid by the brain into the ventricles. The choroid plexus is a structure lining the floor of the lateral ventricle and the roof of the third and fourth ventricles. Certain studies have indicated that these structures are capable of producing 400-600 ccs of fluid per day consistent with an amount to fill the central nervous system spaces four times in a day. In adult humans, the volume of this fluid has been calculated to be from 125 to 150 ml (4-5 oz). The CSF is in continuous formation, circulation and absorption. Certain studies have indicated that approximately 430 to 450 ml (nearly 2 cups) of CSF may be produced every day. Certain calculations estimate that production equals approximately 0.35 ml per minute in adults and 0.15 per minute in infant humans. The choroid plexuses of the lateral ventricles produce the majority of CSF. It flows through the foramina of Monro into the third ventricle where it is added to by production from the third ventricle and continues down through the aqueduct of Sylvius to the fourth ventricle. The fourth ventricle adds more CSF; the fluid then travels into the subarachnoid space through the foramina of Magendie and Luschka. It then circulates throughout the base of the brain, down around the spinal cord and upward over the cerebral hemispheres. The CSF empties into the blood via the arachnoid villi and intracranial vascular sinuses.

[0274] In certain embodiments, such pharmaceutical compositions are administered systemically. In certain embodiments, pharmaceutical compositions are administered subcutaneously. In certain embodiments, pharmaceutical compositions are administered intravenously. In certain embodiments, pharmaceutical compositions are administered by intramuscular injection.

[0275] In certain embodiments, pharmaceutical compositions are administered both directly to the CSF (e.g., IT and/or ICV injection and/or infusion) and systemically.

[0276] In certain embodiments, an antisense compound administered systemically enters neurons. In certain embodiments, systemically administered antisense compounds may penetrate the blood-brain barrier, particularly in young subjects where the blood-brain barrier is not fully formed (e.g., in subjects in eutero and/or in newborn subjects). In certain embodiments, some amount of systemically administered antisense compound may be taken up by nerve cells, even in subjects in which the blood-brain barrier is fully formed. For example, antisense compounds may enter a neuron at or near the neuromuscular junction (retrograde uptake). In certain embodiments, such retrograde uptake results in antisense activity inside the neuron, including, but not limited to, a motor neuron, and provides a therapeutic benefit by antisense activity inside the neuron.

[0277] In certain embodiments, systemic administration provides therapeutic benefit by antisense activity occurring in cells and/or tissues other than neurons. While evidence suggests that functional SMN inside neurons is required for normal neuron function, the consequence of reduced functional SMN in other cells and tissues is not well characterized. In certain embodiments, antisense activity in non-neuronal cells results in restoration of SMN function in those non-neuronal cells, which in turn results in therapeutic benefit.

[0278] In certain embodiments, improved SMN function in non-neuronal cells provides improved neuronal cell function, whether or not SMN function inside neurons is improved. For example, in certain embodiments, systemic administration of pharmaceutical compositions of the present invention results in antisense activity in muscle cells. Such antisense activity in muscle cells may provide a benefit to the motor-neurons associated with that muscle cell or to neurons generally. In such embodiments, the muscle cell having restored SMN function may provide a factor that improves neuronal viability and/or function. In certain embodiments, such antisense activity is independent of benefit from antisense activity occurring from antisense compounds inside neurons. In certain embodiments, systemic administration of pharmaceutical compositions of the present invention results in antisense activity in other non-neuronal cells, including cells not in immediate association with neurons. Such antisense activity in non-neuronal cells may improve function of neurons. For example, antisense activity in a non-neuronal cell (e.g., liver cell) may result in that cell producing a factor that improves function of neurons. Note: since the term "antisense activity" includes direct and indirect activities, a benefit to neuronal function is an "antisense activity" even if no antisense compound enters the neuron.

[0279] In certain embodiments, systemic administration of a pharmaceutical composition results in therapeutic benefit independent of direct or indirect antisense activities in neurons. Typically, in the setting of SMA, neuronal function is diminished, resulting in significant symptoms. Additional symptoms may result from diminished SMN activity in other cells. Certain such symptoms may be masked by the relative severity of symptoms from diminished neuronal function. In certain embodiments, systemic administration results in restored or improved SMN function in non-neuronal cells. In certain such embodiments, such restored or improved SMN function in non-neuronal cells has therapeutic benefit. For example, in certain instances, subjects having SMA have reduced growth. Such reduced growth may not result from diminished function in neuronal cells. Indeed, reduced growth may be related to impaired function of cells in another organ, such as the pituitary gland, and/or may be the result of SMN deficiencies throughout the cells of the body. In such embodiments, systemic administration may result in improved SMN activity in pituitary cells and/or other cells, resulting in improved growth. In certain instances, administration to the CSF restores sufficient neuronal function to allow a subject to live longer, however one or more symptoms previously unknown because subjects typically died before such symptoms appeared emerges, because the subject lives longer. Certain such emergent symptoms may be lethal. In certain embodiments, emergent symptoms are treated by systemic administration. Regardless of mechanism, in certain embodiments, a variety of symptoms of SMA, including, but not limited to symptoms previously masked by more severe symptoms associated with impaired neuronal function, may be treated by systemic administration.

[0280] In certain embodiments, systemic administration of pharmaceutical compositions of the present invention result in increased SMN activity in muscle cells. In certain embodiments, such improved SMN activity in muscle cells provides therapeutic benefit. Improved SMN activity in muscle alone has been reported to be insufficient to provide therapeutic benefit (e.g., Gravrilina, et al., Hum Mol Genet 2008 17(8):1063-1075). In certain embodiments, the present invention provides methods that result improve SMN function in muscle and do provide therapeutic benefit. In certain instances, therapeutic benefit may be attributable to improved SMN function in other cells (alone or in combination with muscle cells). In certain embodiments, improved SMN function in muscle alone may provide benefit.

[0281] In certain embodiments, systemic administration results in improved survival.

6. Spinal Muscular Atrophy (SMA)

[0282] SMA is a genetic disorder characterized by degeneration of spinal motor neurons. SMA is caused by the homozygous loss of both functional copies of the SMN1 gene. However, the SMN2 gene has the potential to code for the same protein as SMN1 and thus overcome the genetic defect of SMA patients. SMN2 contains a translationally silent mutation (C.fwdarw.T) at position+6 of exon 7, which results in inefficient inclusion of exon 7 in SMN2 transcripts. Therefore, the predominant form of SMN2, one which lacks exon 7, is unstable and inactive. Thus, therapeutic compounds capable of modulating SMN2 splicing such that the percentage of SMN2 transcripts containing exon 7 is increased, would be useful for the treatment of SMA.

[0283] In certain embodiments, the present invention provides antisense compounds complementary to a pre-mRNA encoding SMN2. In certain such embodiments, the antisense compound alters splicing of SMN2. Certain sequences and regions useful for altering splicing of SMN2 may be found in PCT/US06/024469, which is hereby incorporated by reference in its entirety for any purpose. In certain embodiments, oligomeric compounds having any motif described herein have a nucleobase sequence complementary to intron 7 of SMN2. Certain such nucleobase sequences are exemplified in the non-limiting table below.

TABLE-US-00003 Sequence Length SEQ ID TGCTGGCAGACTTAC 15 3 CATAATGCTGGCAGA 15 4 TCATAATGCTGGCAG 15 5 TTCATAATGCTGGCA 15 6 TTTCATAATGCTGGC 15 2 ATTCACTTTCATAATGCTGG 20 7 TCACTTTCATAATGCTGG 18 1 CTTTCATAATGCTGG 15 8 TCATAATGCTGG 12 9 ACTTTCATAATGCTG 15 10 TTCATAATGCTG 12 11 CACTTTCATAATGCT 15 12 TTTCATAATGCT 12 13 TCACTTTCATAATGC 15 14 CTTTCATAATGC 12 15 TTCACTTTCATAATG 15 16 ACTTTCATAATG 12 17 ATTCACTTTCATAAT 15 18 CACTTTCATAAT 12 19 GATTCACTTTCATAA 15 20 TCACTTTCATAA 12 21 TTCACTTTCATA 12 22 ATTCACTTTCAT 12 23 AGTAAGATTCACTTT 15 24

[0284] Antisense compounds of the present invention can be used to modulate the expression of SMN2 in a subject, such as a human. In certain embodiments, the subject has spinal muscular atrophy. In certain such subjects, the SMN1 gene is absent or otherwise fails to produce sufficient amounts of functional SMN protein. In certain embodiments, the antisense compounds of the present invention effectively modulate splicing of SMN2, resulting in an increase in exon 7 inclusion in SMN2 mRNA and ultimately in SMN2 protein that includes the amino acids corresponding to exon 7. Such alternate SMN2 protein resembles wild-type SMN protein. Antisense compounds of the present invention that effectively modulate expression of SMN2 mRNA or protein products of expression are considered active antisense compounds.

[0285] Modulation of expression of SMN2 can be measured in a bodily fluid, which may or may not contain cells; tissue; or organ of the animal. Methods of obtaining samples for analysis, such as body fluids (e.g., sputum, serum, CSF), tissues (e.g., biopsy), or organs, and methods of preparation of the samples to allow for analysis are well known to those skilled in the art. Methods for analysis of RNA and protein levels are discussed above and are well known to those skilled in the art. The effects of treatment can be assessed by measuring biomarkers associated with the target gene expression in the aforementioned fluids, tissues or organs, collected from an animal contacted with one or more compounds of the invention, by routine clinical methods known in the art.

[0286] Methods whereby bodily fluids, organs or tissues are contacted with an effective amount of one or more of the antisense compounds or compositions of the invention are also contemplated. Bodily fluids, organs or tissues can be contacted with one or more of the compounds of the invention resulting in modulation of SMN2 expression in the cells of bodily fluids, organs or tissues. An effective amount can be determined by monitoring the modulatory effect of the antisense compound or compounds or compositions on target nucleic acids or their products by methods routine to the skilled artisan.

[0287] The invention also provides an antisense compound as described herein, for use in any of the methods as described herein. For example, the invention provides an antisense compound comprising an antisense oligonucleotide complementary to a nucleic acid encoding human SMN2, for use in treating a disease or condition associated with survival motor neuron protein (SMN), such as spinal muscular atrophy (SMA). As a further example, the invention provides an antisense compound comprising an antisense oligonucleotide complementary to a nucleic acid encoding human SMN2, for use in treating a disease or condition associated with survival motor neuron protein (SMN) by administering the antisense compound directly into the central nervous system (CNS) or CSF.

[0288] The invention also provides the use of an antisense compound as described herein in the manufacture of a medicament for use in any of the methods as described herein. For example, the invention provides the use of an antisense compound comprising an antisense oligonucleotide complementary to a nucleic acid encoding human SMN2 in the manufacture of a medicament for treating a disease or condition associated with survival motor neuron protein (SMN), such as spinal muscular atrophy (SMA). As a further example, the invention provides the use of an antisense compound comprising an antisense oligonucleotide complementary to a nucleic acid encoding human SMN2 in the manufacture of a medicament for treating a disease or condition associated with survival motor neuron protein (SMN) by administration of the medicament directly into the central nervous system (CNS) or CSF.

[0289] In certain embodiments, oligomeric compounds having any motif described herein have a nucleobase sequence complementary to exon 7 of SMN2.

[0290] In certain embodiments, oligomeric compounds having any motif described herein have a nucleobase sequence complementary to intron 6 of SMN2.

[0291] In certain embodiments, an antisense compound comprises an antisense oligonucleotide having a nucleobase sequence comprising at least 10 nucleobases of the sequence: TCACTTTCATAATGCTGG (SEQ ID NO: 1). In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 11 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 12 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 13 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 14 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 15 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 16 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising at least 17 nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence comprising the nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide has a nucleobase sequence consisting of the nucleobases of such sequence. In certain embodiments, an antisense oligonucleotide consists of 10-18 linked nucleosides and has a nucleobase sequence 100% identical to an equal-length portion of the sequence: TCACTTTCATAATGCTGG (SEQ ID NO: 1).

[0292] In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 1. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 2. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 3. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 4. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 5. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 6. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 7. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 8. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 9. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 10. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 11. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 12. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 13. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 14. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 15. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 16. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 17. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 18. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 19. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 20. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 21. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 22. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 23. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 24. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 26. In certain embodiments, a compound comprises a modified oligonucleotide having a nucleobase sequence that consists of SEQ ID NO: 27.

[0293] A compound comprising Isis 449320. A compound comprising Isis 449323. A compound comprising Isis 605918. A compound comprising Isis 605919. A compound comprising Isis 663006. A compound comprising Isis 663007. A compound comprising Isis 663008. A compound comprising Isis 663009. A compound comprising Isis 663010. A compound comprising Isis 663011. A compound comprising Isis 663012. A compound comprising Isis 663013. A compound comprising Isis 663014. A compound comprising Isis 663015. A compound comprising Isis 663016. A compound comprising Isis 663017. A compound comprising Isis 663018. A compound comprising Isis 663019. A compound comprising Isis 669541. A compound comprising Isis 710554. A compound comprising Isis 710555. A compound comprising Isis 710556. A compound comprising Isis 710557. A compound comprising Isis 710558. A compound comprising Isis 449321. A compound comprising Isis 449322. A compound comprising Isis 710543. A compound comprising Isis 710544. A compound comprising Isis 710548. A compound comprising Isis 710549. A compound comprising Isis 710550. A compound comprising Isis 710551. A compound comprising Isis 710552. A compound comprising Isis 710553. A compound comprising Isis 758133. A compound comprising Isis 758134. A compound comprising Isis 758135. A compound comprising Isis 758136. A compound comprising Isis 758137. A compound comprising Isis 758138. A compound comprising Isis 758139. A compound comprising Isis 758140. A compound comprising Isis 494323.

[0294] A compound consisting of Isis 449320. A compound consisting of Isis 449323. A compound consisting of Isis 605918. A compound consisting of Isis 605919. A compound consisting of Isis 663006. A compound consisting of Isis 663007. A compound consisting of Isis 663008. A compound consisting of Isis 663009. A compound consisting of Isis 663010. A compound consisting of Isis 663011. A compound consisting of Isis 663012. A compound consisting of Isis 663013. A compound consisting of Isis 663014. A compound consisting of Isis 663015. A compound consisting of Isis 663016. A compound consisting of Isis 663017. A compound consisting of Isis 663018. A compound consisting of Isis 663019. A compound consisting of Isis 669541. A compound consisting of Isis 710554. A compound consisting of Isis 710555. A compound consisting of Isis 710556. A compound consisting of Isis 710557. A compound consisting of Isis 710558. A compound consisting of Isis 449321. A compound consisting of Isis 449322. A compound consisting of Isis 710543. A compound consisting of Isis 710544. A compound consisting of Isis 710548. A compound consisting of Isis 710549. A compound consisting of Isis 710550. A compound consisting of Isis 710551. A compound consisting of Isis 710552. A compound consisting of Isis 710553. A compound consisting of Isis 758133. A compound consisting of Isis 758134. A compound consisting of Isis 758135. A compound consisting of Isis 758136. A compound consisting of Isis 758137. A compound consisting of Isis 758138. A compound consisting of Isis 758139. A compound consisting of Isis 758140. A compound consisting of Isis 494323.

7. Certain Subjects

[0295] In certain embodiments, a subject has one or more indicator of SMA. In certain embodiments, the subject has reduced electrical activity of one or more muscles. In certain embodiments, the subject has a mutant SMN1 gene. In certain embodiment, the subject's SMN1 gene is absent or incapable of producing functional SMN protein. In certain embodiments, the subject is diagnosed by a genetic test. In certain embodiments, the subject is identified by muscle biopsy. In certain embodiments, a subject is unable to sit upright. In certain embodiments, a subject is unable to stand or walk. In certain embodiments, a subject requires assistance to breathe and/or eat. In certain embodiment, a subject is identified by electrophysiological measurement of muscle and/or muscle biopsy.

[0296] In certain embodiments, the subject has SMA type I. In certain embodiments, the subject has SMA type II. In certain embodiments, the subject has SMA type III. In certain embodiments, the subject is diagnosed as having SMA in utero. In certain embodiments, the subject is diagnosed as having SMA within one week after birth. In certain embodiments, the subject is diagnosed as having SMA within one month of birth. In certain embodiments, the subject is diagnosed as having SMA by 3 months of age. In certain embodiments, the subject is diagnosed as having SMA by 6 months of age. In certain embodiments, the subject is diagnosed as having SMA by 1 year of age. In certain embodiments, the subject is diagnosed as having SMA between 1 and 2 years of age. In certain embodiments, the subject is diagnosed as having SMA between 1 and 15 years of age. In certain embodiments, the subject is diagnosed as having SMA when the subject is older than 15 years of age.

[0297] In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered in utero. In certain such embodiments, the first dose is administered before complete development of the blood-brain-barrier. In certain embodiments, the first dose is administered to the subject in utero systemically. In certain embodiments, the first dose is administered in utero after formation of the blood-brain-barrier. In certain embodiments, the first dose is administered to the CSF.

[0298] In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than one week old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than one month old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than 3 months old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than 6 months old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than one year old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than 2 years old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is less than 15 years old. In certain embodiments, the first dose of a pharmaceutical composition according to the present invention is administered when the subject is older than 15 years old.

8. Certain Doses

[0299] In certain embodiments, the present invention provides dose amounts and frequencies. In certain embodiments, pharmaceutical compositions are administered as a bolus injection. In certain such embodiments, the dose of the bolus injection is from 0.01 to 25 milligrams of antisense compound per kilogram body weight of the subject. In certain such embodiments, the dose of the bolus injection is from 0.01 to 10 milligrams of antisense compound per kilogram body weight of the subject. In certain embodiments, the dose is from 0.05 to 5 milligrams of antisense compound per kilogram body weight of the subject. In certain embodiments, the dose is from 0.1 to 2 milligrams of antisense compound per kilogram body weight of the subject. In certain embodiments, the dose is from 0.5 to 1 milligrams of antisense compound per kilogram body weight of the subject. In certain embodiments, such doses are administered twice monthly. In certain embodiments, such doses are administered every month. In certain embodiments, such doses are administered every 2 months. In certain embodiments, such doses are administered every 6 months. In certain embodiments, such doses are administered by bolus injection into the CSF. In certain embodiments, such doses are administered by intrathecal bolus injection. In certain embodiments, such doses are administered by bolus systemic injection (e.g., subcutaneous, intramuscular, or intravenous injection). In certain embodiments, subjects receive bolus injections into the CSF and bolus systemic injections. In such embodiments, the doses of the CSF bolus and the systemic bolus may be the same or different from one another. In certain embodiments, the CSF and systemic doses are administered at different frequencies. In certain embodiments, the invention provides a dosing regimen comprising at least one bolus intrathecal injection and at least one bolus subcutaneous injection.

[0300] In certain embodiments, pharmaceutical compositions are administered by continuous infusion. Such continuous infusion may be accomplished by an infusion pump that delivers pharmaceutical compositions to the CSF. In certain embodiments, such infusion pump delivers pharmaceutical composition IT or ICV. In certain such embodiments, the dose administered is between 0.05 and 25 milligrams of antisense compound per kilogram body weight of the subject per day. In certain embodiments, the dose administered is from 0.1 to 10 milligrams of antisense compound per kilogram body weight of the subject per day. In certain embodiments, the dose administered is from 0.5 to 10 milligrams of antisense compound per kilogram body weight of the subject per day. In certain embodiments, the dose administered is from 0.5 to 5 milligrams of antisense compound per kilogram body weight of the subject per day. In certain embodiments, the dose administered is from 1 to 5 milligrams of antisense compound per kilogram body weight of the subject per day. In certain embodiments, the invention provides a dosing regimen comprising infusion into the CNS and at least one bolus systemic injection. In certain embodiments, the invention provides a dosing regimen comprising infusion into the CNS and at least one bolus subcutaneous injection. In certain embodiments, the dose, whether by bolus or infusion, is adjusted to achieve or maintain a concentration of antisense compound from 0.1 to 100 microgram per gram of CNS tissue. In certain embodiments, the dose, whether by bolus or infusion, is adjusted to achieve or maintain a concentration of antisense compound from 1 to 10 microgram per gram of CNS tissue. In certain embodiments, the dose, whether by bolus or infusion, is adjusted to achieve or maintain a concentration of antisense compound from 0.1 to 1 microgram per gram of CNS tissue.

[0301] In certain embodiments, dosing a subject is divided into an induction phase and a maintenance phase. In certain such embodiments, the dose administered during the induction phase is greater than the dose administered during the maintenance phase. In certain embodiments, the dose administered during the induction phase is less than the dose administered during the maintenance phase. In certain embodiments, the induction phase is achieved by bolus injection and the maintenance phase is achieved by continuous infusion.

[0302] In certain embodiments, the invention provides systemic administration of antisense compounds, either alone or in combination with delivery into the CSF. In certain embodiments, the dose for systemic administration is from 0.1 mg/kg to 200 mg/kg. In certain embodiments, the dose for systemic administration is from 0.1 mg/kg to 100 mg/kg. In certain embodiments, the dose for systemic administration is from 0.5 mg/kg to 100 mg/kg. In certain embodiments, the dose for systemic administration is from 1 mg/kg to 100 mg/kg. In certain embodiments, the dose for systemic administration is from 1 mg/kg to 50 mg/kg. In certain embodiments, the dose for systemic administration is from 1 mg/kg to 25 mg/kg. In certain embodiments, the dose for systemic administration is from 0.1 mg/kg to 25 mg/kg. In certain embodiments, the dose for systemic administration is from 0.1 mg/kg to 10 mg/kg. In certain embodiments, the dose for systemic administration is from 1 mg/kg to 10 mg/kg. In certain embodiments, the dose for systemic administration is from 1 mg/kg to 5 mg/kg. In certain embodiments comprising both systemic and CSF delivery, the doses for those two routes are independently determined.

[0303] a. Calculation of Appropriate Human Doses

[0304] In certain embodiments, the subject is a human. In certain embodiments, a human dose is calculated or estimated from data from animal experiments, such as those described herein. In certain embodiments, a human dose is calculated or estimated from data from monkey and/or mouse experiments, such as those described herein. In certain embodiments, a human dose is calculated or estimated from data from mouse experiments, such as those described herein. In certain embodiments, appropriate human doses can be calculated using pharmacokinetic data from mouse along with knowledge of brain weight and/or cerebrospinal fluid (CSF) turnover rates. For example, the mouse brain weight is approximately 0.4 g, which is approximately 2% of its body weight. In humans, the average brain weight is 1.5 kg which is approximately 2.5% of body weight. In certain embodiments, administration into the CSF results in elimination of a portion of the compound through uptake in brain tissue and subsequent metabolism. By using the ratio of human to mouse brain weight as a scaling factor an estimate of the elimination and clearance through the brain tissue can be calculated. Additionally, the CSF turnover rate can be used to estimate elimination of compound from the CSF to blood. Mouse CSF turnover rate is approximately 10-12 times per day (0.04 mL produced at 0.325 .mu.l/min). Human CSF turnover rate is approximately 4 times per day (100-160 mL produced at 350-400 .mu.l/min). Clearance, and therefore dosing requirements, can be based on brain weight elimination scaling, and/or the CSF turnover scaling. The extrapolated human CSF clearance can be used to estimate equivalent doses in humans that approximate doses in mice. In this way, human doses can be estimated that account for differences in tissue metabolism based on brain weight and CSF turnover rates. Such methods of calculation and estimate are known to those skilled in the art.

[0305] By way of non-limiting example, in certain embodiments, an equivalent human dose can be estimated from a desired mouse dose by multiplying the mg/kg mouse dose by a factor from about 0.25 to about 1.25 depending on the determined clearance and elimination of a particular compound. Thus, for example, in certain embodiments, a human dose equivalent of a 0.01 mg dose for a 20 g mouse will range from about 8.75 mg to about 43.75 mg total dose for a 70 kg human. Likewise, in certain embodiments, a human dose equivalent of a 0.01 mg dose for a 4 g newborn mouse will range from about 1.9 mg to about 9.4 mg total dose for a 3 kg newborn human. These example doses are merely to illustrate how one of skill may determine an appropriate human dose and are not intended to limit the present invention.

[0306] In certain embodiments, a human dose for systemic delivery (whether administered alone or in combination with CSF delivery) is calculated or estimated from data from animal experiments, such as those described herein. Typically, an appropriate human dose (in mg/kg) for systemic dose is between 0.1 and 10 times an effective dose in animals. Thus, solely for example, a subcutaneous dose of 50 .mu.g in a 2 g newborn mouse is a dose of 25 mg/kg. The corresponding dose for a human is predicted to be between 2.5 mg/kg and 250 mg/kg. For a 3 kilogram infant, the corresponding dose is between 7.5 mg and 750 mg. For a 25 kg child, the corresponding dose is from 62.5 mg to 6250 mg.

9. Treatment Regimens

[0307] In certain embodiments, the above dose amounts, dose frequencies, routes of administration, induction and maintenance phases, and timing of first dose are combined to provide dosing regimens for subjects having SMA. Such dosing regimens may be selected and adjusted to provide amelioration of one or more symptom of SMA and/or to reduce or avoid toxicity or side effects attributable to the administration of the pharmaceutical composition. In certain embodiments, subjects are in utero or newborn. In such embodiments, administration of pharmaceutical compositions, particularly by continuous infusion, presents particular challenges. Accordingly, in certain embodiments, the present invention provides for administration of pharmaceutical compositions by bolus administration while the subject is in utero or very young, followed by continuous infusion via an implanted infusion pump when the subject is older and placement of such pump is more practical. Further, in certain embodiments, as a subject grows, the absolute dose is increased to achieve the same or similar dose:body-weight ratio.

[0308] In certain embodiments, pharmaceutical compositions of the present disclosure have improved therapeutic indices. In certain embodiments, pharmaceutical compositions of the present disclosure having improved therapeutic indices allows one having skill in the art to administer the pharmaceutical compositions of the present disclosure more infrequently. In certain embodiments, pharmaceutical compositions of the present disclosure having improved therapeutic indices allows one having skill in the art to allow longer intervals between the first dose and the second dose.

[0309] The following table is intended to exemplify treatment regimens and is not intended to limit the possible combinations of treatments which will be easily accomplished by one of skill in the art.

TABLE-US-00004 Dosing period First Second Third Fourth Fifth Regimen 1 Subject Age In utero, prior In utero, after >1 week 6 months 1.5 years to formation formation of of blood- blood-brain- brain-barrier barrier Dose Amount 50 .mu.g 50 .mu.g 100 .mu.g 10 .mu.g/day 50 .mu.g/day Frequency Single admin Single admin Monthly Continuous Continuous Route of Systemic IT injection IT injections IT infusion IT infusion Administration injection Duration N/A N/A 6 months 1 year Ongoing Regimen 2 Subject Age In utero, after >1 week 6 months 1.5 years N/A formation of blood-brain- barrier Dose Amount 50 .mu.g 100 .mu.g 5 mg/day 10 mg/day N/A Frequency Single admin Monthly Continuous Continuous N/A Route of ICV injection ICV injection ICV infusion ICV infusion N/A Administration Duration N/A 6 months 1 year Ongoing N/A Regimen 3 Subject Age >1 week 6 months 1.5 years 2.5 years* Dose Amount 100 .mu.g 500 .mu.g/day 20 mg/day 20 mg/day 100 mg Frequency 2xMonthly Continuous Continuous Continuous 2xMonthly Route of ICV injection ICV infusion ICV infusion ICV infusion IP Administration Duration 6 months 1 year 1 year Ongoing Ongoing *Note: the 4.sup.th dosing period in regimen 3 exemplifies continuous CSF infusion combined with periodic systemic administration. These treatment regimens are intended to exemplify and not to limit the present invention.

[0310] In certain embodiments, the dosing regimen comprises a systemic administration, either alone or in combination with administration into the CSF (for example regimen 3, above). The table, below further exemplifies such regimens.

TABLE-US-00005 Systemic administration CSF administration Dose Route Frequency Dose Route Frequency 1-5 mg/kg subcutaneous weekly 5-10 mg/kg bolus IT monthly 1-5 mg/kg subcutaneous monthly 1-5 mg/kg bolus ICV 2 months 10-50 mg/kg subcutaneous monthly 0.5-1 mg/kg bolus IT 6 months 0.5-25 mg/kg subcutaneous monthly 10 mg/kg/day IT infusion continuous for 7 days every 6 months 0.1-10 mg/kg subcutaneous monthly none none 0.5-1 mg/kg bolus IT 6 months

These treatment regimens are intended to exemplify and not to limit the present invention. One of skill in the art will be able to select an appropriate combination of the doses and deliveries in view of the present disclosure and based on a variety of factors, such as the severity of the condition and the overall health and age of the subject.

10. Co-Administration

[0311] In certain embodiments, pharmaceutical compositions of the present invention are co-administered with at least one other pharmaceutical composition for treating SMA and/or for treating one or more symptom associated with SMA. In certain embodiments, such other pharmaceutical composition is selected from trichostatin-A, valproic acid, riluzole, hydroxyurea, and a butyrate or butyrate derivative. In certain embodiments, pharmaceutical compositions of the present invention are co-administered with trichostatin A. In certain embodiments, pharmaceutical compositions of the present invention are co-administered with a derivative of quinazoline, for example as described in Thurmond, et al., J. Med Chem. 2008, 51, 449-469. In certain embodiments, a pharmaceutical composition of the present invention and at least one other pharmaceutical composition are co-administered at the same time. In certain embodiments, a pharmaceutical composition of the present invention and at least one other pharmaceutical composition are co-administered at different times.

[0312] In certain embodiments, pharmaceutical compositions of the present invention are co-administered with a gene therapy agent. In certain such embodiments, the gene therapy agent is administered to the CSF and the pharmaceutical composition of the present invention is administered systemically. In certain such embodiments, the gene therapy agent is administered to the CSF and the pharmaceutical composition of the present invention is administered to the CSF and systemically. In certain embodiments, a pharmaceutical composition of the present invention and a gene therapy agent are co-administered at the same time. In certain embodiments, a pharmaceutical composition of the present invention and a gene therapy agent are co-administered at different times. Certain gene therapy approaches to SMA treatment have been reported (e.g., Coady et al., PLoS ONE 2008 3(10): e3468; Passini et al., J Clin Invest 2010 Apr. 1, 120(4): 1253-64).

[0313] In certain embodiments, pharmaceutical compositions of the present invention are co-administered with at least one other therapy for SMA. In certain embodiments, such other therapy for SMA is surgery. In certain embodiments, such other therapy is physical therapy, including, but not limited to exercises designed to strengthen muscles necessary for breathing, such as cough therapy. In certain embodiments, other therapy is a physical intervention, such as a feeding tube or device for assisted breathing.

[0314] In certain embodiments, pharmaceutical compositions of the present invention are co-administered with one or more other pharmaceutical compositions that reduce an undesired side-effect of the pharmaceutical compositions of the present invention.

11. Phenotypic Effects

[0315] In certain embodiments, administration of at least one pharmaceutical composition of the present invention results in a phenotypic change in the subject. In certain embodiments, such phenotypic changes include, but are not limited to: increased absolute amount of SMN mRNA that includes exon 7; increase in the ratio SMN mRNA that includes exon 7 to SMN mRNA lacking exon 7; increased absolute amount of SMN protein that includes exon 7; increase in the ratio SMN protein that includes exon 7 to SMN protein lacking exon 7; improved muscle strength, improved electrical activity in at least one muscle; improved respiration; weight gain; and survival. In certain embodiments, at least one phenotypic change is detected in a motoneuron of the subject. In certain embodiments, administration of at least one pharmaceutical composition of the present invention results in a subject being able to sit-up, to stand, and/or to walk. In certain embodiments, administration of at least one pharmaceutical composition of the present invention results in a subject being able to eat, drink, and/or breathe without assistance. In certain embodiments, efficacy of treatment is assessed by electrophysiological assessment of muscle. In certain embodiments, administration of a pharmaceutical composition of the present invention improves at least one symptom of SMA and has little or no inflammatory effect. In certain such embodiment, absence of inflammatory effect is determined by the absence of significant increase in Aifl levels upon treatment.

[0316] In certain embodiments, administration of at least one pharmaceutical composition of the present invention delays the onset of at least one symptom of SMA. In certain embodiments, administration of at least one pharmaceutical composition of the present invention slows the progression of at least one symptom of SMA. In certain embodiments, administration of at least one pharmaceutical composition of the present invention reduces the severity of at least one symptom of SMA.

[0317] In certain embodiments, administration of at least one pharmaceutical composition of the present invention results in an undesired side-effect. In certain embodiments, a treatment regimen is identified that results in desired amelioration of symptoms while avoiding undesired side-effects.

12. Dosage Units

[0318] In certain embodiments pharmaceutical compositions of the present invention are prepared as dosage units for administration. Certain such dosage units are at concentrations selected from 0.01 mg to 100 mg. In certain such embodiments, a pharmaceutical composition of the present invention comprises a dose of antisense compound selected from 0.01 mg, 0.1 mg, 0.5 mg, 1 mg, 5 mg, 10 mg, 20 mg, 25 mg, 50 mg, 75 mg, 100 mg, 150 mg, and 200 mg. In certain embodiments, a pharmaceutical composition is comprises a dose of oligonucleotide selected from 0.1 mg, 0.5 mg, 1 mg, 5 mg, 10 mg, 25 mg, and 50 mg.

13. Kits

[0319] In certain embodiments, the present invention provides kits comprising at least one pharmaceutical composition. In certain embodiments, such kits further comprise a means of delivery, for example a syringe or infusion pump.

Nonlimiting Disclosure and Incorporation by Reference

[0320] While certain compounds, compositions and methods described herein have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds described herein and are not intended to limit the same. Each of the references, GenBank accession numbers, and the like recited herein is hereby incorporated by reference in its entirety.

[0321] Although the sequence listing accompanying this filing identifies each sequence as either "RNA" or "DNA" as required, in reality, those sequences may be modified with any combination of chemical modifications. One of skill in the art will readily appreciate that such designation as "RNA" or "DNA" to describe modified oligonucleotides is, in certain instances, arbitrary. For example, an oligonucleotide comprising a nucleoside comprising a 2'-OH sugar moiety and a thymine base could be described as a DNA having a modified sugar (2'-OH for the natural 2'-H of DNA) or as an RNA having a modified base (thymine (methylated uracil) for natural uracil of RNA).

[0322] Accordingly, nucleic acid sequences provided herein, including, but not limited to those in the sequence listing, are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases. By way of further example and without limitation, an oligomeric compound having the nucleobase sequence "ATCGATCG" encompasses any oligomeric compounds having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence "AUCGAUCG" and those having some DNA bases and some RNA bases such as "AUCGATCG" and oligomeric compounds having other modified bases, such as "AT.sup.meCGAUCG," wherein .sup.meC indicates a cytosine base comprising a methyl group at the 5-position.

EXAMPLES

[0323] The following examples illustrate certain embodiments of the present disclosure and are not limiting. Moreover, where specific embodiments are provided, the inventors have contemplated generic application of those specific embodiments. For example, disclosure of an oligonucleotide having a particular motif provides reasonable support for additional oligonucleotides having the same or similar motif. And, for example, where a particular high-affinity modification appears at a particular position, other high-affinity modifications at the same position are considered suitable, unless otherwise indicated.

Example 1

Effect of Antisense Oligonucleotides Targeting Human SMN2 in an SMA Type III Mice Model

[0324] Taiwan strain of SMA type III mice were obtained from The Jackson Laboratory (Bar Harbor, Me.). These mice lack mouse SMN and are homozygous for human SMN2 (mSMN -/-; hSMN2+/+). These mice have been described in Hsieh-Li H M, et al., Nature Genet. 24, 66-70 2000. Antisense oligonucleotides targeting human SMN2 were designed and tested in these mice.

[0325] The newly designed antisense oligonucleotides in the Table below were designed as uniform 2'-MOE oligonucleotides with mixed backbone chemistry. Each antisense oligonucleotide is 18 nucleosides in length and each nucleoside has a 2'-MOE sugar modification. The internucleoside linkages throughout each gapmer are either phosphodiester or phosphorothioate linkages. The internucleoside linkages of each oligonucleotide is denoted in the Backbone Chemistry column, where `o` indicates a phosphodiester linkage and `s` indicates a phosphorothioate linkage. All cytosine residues throughout each oligonucleotide are 5-methylcytosines. Each antisense oligonucleotide listed in the Table below is targeted to the human SMN2 genomic sequence, designated herein as SEQ ID NO: 25 (GENBANK Accession No. NT_006713.14 truncated from nucleotides 19939708 to Ser. No. 19/967,777) at the target region 27062-27079.

TABLE-US-00006 TABLE 1 Modified oligonucleotides targeting human SMN2 with mixed backbone chemistry ISIS Backbone SEQ No Sequence Chemistry ID NO 449320 TCACTTTCATAATGCTGG ssoooooooooooooss 1 449323 TCACTTTCATAATGCTGG sosososososososos 1 605918 TCACTTTCATAATGCTGG sssososososososss 1 605919 TCACTTTCATAATGCTGG sooossssssssoooss 1

Treatment

[0326] Mice were administered 500 .mu.g of ISIS oligonucleotide by intracerebroventricular (ICV) bolus injection. Control mice were administered PBS alone (dose of 0).

Tolerability Assay

[0327] At 3 hours post injection, each mouse was evaluated according to 7 different criteria. The 7 criteria are (1) the mouse was bright, alert, and responsive; (2) the mouse was standing or hunched without stimuli; (3) the mouse shows any movement without stimuli (4) the mouse demonstrates forward movement after its lifted; (5) the mouse demonstrates any movement after its lifted; (6) the mouse responds to a tail pinch; (7) regular breathing. For each of the 7 different criteria, each mouse was given a sub-score of 0 if it met the criteria or 1 if it did not. After all of the 7 criteria were evaluated, the sub-scores were summed for each mouse and then averaged for each group. For example, if a mouse was bright, alert, and responsive 3 hours after the ICV dose, and met all other criteria, it would get a summed score of 0. If another mouse was not bright, alert, and responsive 3 hours after the ICV dose but met all other criteria, it would receive a score of 1. Saline treated mice generally receive a score of 0. As presented in the Table below, the ISIS oligonucleotides were deemed tolerable in the transgenic mice.

TABLE-US-00007 TABLE 2 Scoring for tolerability to ISIS oligonucleotides in SMA Type III mice ISIS No 3 hr Score 449320 0 449323 0 605918 3 605919 0

Exon 7 Splicing

[0328] Animals were sacrificed 2 weeks after the injection and brain and lumbar sections of the spinal cords were collected from each animal. Real time PCR was performed on each sample to determine the amount of human SMN2 message including exon 7 (exon 7.sup.+) and the amount of human SMN2 message lacking exon 7(exon 7.sup.-). Expression levels for exon 7.sup.+ and exon 7.sup.- were normalized to total SMN2 levels. The data presented in the Table below are expressed as exon 7.sup.+ and exon 7.sup.- levels divided by total SMN levels, and were further normalized to the levels obtained in the PBS control group (designated as 1).

[0329] Administration of ISIS oligonucleotides targeting SEQ ID NO: 25 at start site 27062, but with different backbone chemistries, all resulted in a striking increase in inclusion of exon 7.

TABLE-US-00008 TABLE 3 Effect of ISIS oligonucleotides targeting SMN2 on splicing in SMA Type III Mice Brain Lumbar Cord exon 7.sup.+ exon 7.sup.- exon 7.sup.+ exon 7.sup.- PBS 1.0 1.0 1.0 1.0 449320 2.4 0.60 2.7 0.66 449323 4.3 0.24 4.5 0.33 605918 4.4 0.23 4.5 0.28 605919 4.2 0.26 4.0 0.38

Example 2

Effect of Antisense Oligonucleotides Targeting Human SMN2 in SMA Type III Mice and WT Mice

[0330] Additional antisense oligonucleotides targeting human SMN2 were designed and tested in SMA Type III mice or a WT (C57BL/6) strain.

[0331] The newly designed antisense oligonucleotides in the Table below were designed as uniform 2'-MOE oligonucleotides with mixed backbone chemistry. Each antisense oligonucleotide is 18 nucleosides in length wherein each nucleoside has a 2'-MOE sugar modification. The internucleoside linkages throughout each gapmer are either phosphodiester or phosphorothioate linkages. The internucleoside linkages of each oligonucleotide is denoted in the Backbone Chemistry column, where `o` indicates a phosphodiester linkage and `s` indicates a phosphorothioate linkage. All cytosine residues throughout each oligonucleotide are 5-methylcytosines. Each antisense oligonucleotide listed in the Table below is targeted to the human SMN2 genomic sequence, designated herein as SEQ ID NO: 25 (GENBANK Accession No. NT_006713.14 truncated from nucleotides 19939708 to Ser. No. 19/967,777) at the target region 27062-27079.

TABLE-US-00009 TABLE 4 Modified oligonucleotides targeting human SMN2 with mixed backbone chemistry ISIS Backbone SEQ No Sequence chemistry ID NO 605918 TCACTTTCATAATGCTGG sssososososososss 1 605919 TCACTTTCATAATGCTGG sooossssssssoooss 1 663006 TCACTTTCATAATGCTGG ssosososososososs 1 663007 TCACTTTCATAATGCTGG ssosososososoooss 1 663008 TCACTTTCATAATGCTGG soosoosoosoosooss 1 663009 TCACTTTCATAATGCTGG soooosssssssoooss 1 663010 TCACTTTCATAATGCTGG soooossssssooooss 1 663011 TCACTTTCATAATGCTGG sooooosssssooooss 1 663012 TCACTTTCATAATGCTGG sooooossssoooooss 1 663013 TCACTTTCATAATGCTGG sssooooooooooosss 1 663014 TCACTTTCATAATGCTGG ssssoooooooooosss 1 663015 TCACTTTCATAATGCTGG ssssooooooooossss 1 663016 TCACTTTCATAATGCTGG sssssoooooooossss 1 663017 TCACTTTCATAATGCTGG sssssooooooosssss 1 663018 TCACTTTCATAATGCTGG ssssssoooooosssss 1 663019 TCACTTTCATAATGCTGG ssssssooooossssss 1 669541 TCACTTTCATAATGCTGG sooosssssssssooss 1

Treatment

[0332] SMA Type III mice were administered 35 .mu.g of ISIS oligonucleotide by intracerebroventricular (ICV) bolus injection. WT mice were administered 700 .mu.g of ISIS oligonucleotide by intracerebroventricular (ICV) bolus injection. Control mice in each strain were administered PBS alone (dose of 0).

Tolerability Assay

[0333] At 3 hours post injection, each mouse was evaluated according to 7 different criteria. The 7 criteria are (1) the mouse was bright, alert, and responsive; (2) the mouse was standing or hunched without stimuli; (3) the mouse shows any movement without stimuli (4) the mouse demonstrates forward movement after its lifted; (5) the mouse demonstrates any movement after its lifted; (6) the mouse responds to a tail pinch; (7) regular breathing. For each of the 7 different criteria, each mouse was given a sub-score of 0 if it met the criteria or 1 if it did not. After all of the 7 criteria were evaluated, the sub-scores were summed for each mouse and then averaged for each group. For example, if a mouse was bright, alert, and responsive 3 hours after the ICV dose, and met all other criteria, it would get a summed score of 0. If another mouse was not bright, alert, and responsive 3 hours after ICV dose but met all other criteria, it would receive a score of 1. Saline treated mice generally receive a score of 0. As presented in the Table below, the ISIS oligonucleotides were deemed tolerable in the mice.

TABLE-US-00010 TABLE 5 Scoring for tolerability to ISIS oligonucleotides in SMA Type III mice dosed with 35 .mu.g of ISIS Oligonucleotide ISIS No 3 hr Score 605918 0 605919 0 663006 0 663007 0 663008 0 663009 0 663010 0 663011 0 663012 0 663013 0 663014 0 663015 0 663016 0 663017 0 663018 0 663019 0 669541 0

TABLE-US-00011 TABLE 6 Scoring for tolerability to ISIS oligonucleotides in WT mice dosed with 700 .mu.g of ISIS Oligonucleotide ISIS No 3 hr Score 605918 1 605919 3.3 663006 0 663007 0 663008 0 663009 0 663010 1.5 663011 0 663012 0 663013 0 663014 0 663015 0 663016 0.5 663017 2 663018 3 663019 2.75 669541 3

Exon 7 Splicing

[0334] Animals were sacrificed 2 weeks after the injection and brain and lumbar sections of the spinal cords were collected from each animal. Real time PCR was performed on each sample to determine the amount of human SMN2 message including exon 7 (exon 7.sup.+) and the amount of human SMN2 message lacking exon 7(exon 7.sup.-). Expression levels for exon 7.sup.+ and exon 7.sup.- were normalized to total SMN2 levels. The data presented in the Table below exon 7.sup.+ and exon 7.sup.- levels divided by total SMN levels, and were further normalized to the levels obtained in the PBS control group (designated as 1).

[0335] Administration of many of the ISIS oligonucleotides targeting SEQ ID NO: 25 around target region 27062-27079, but with different backbone chemistries, resulted in an increase in inclusion of exon 7.

TABLE-US-00012 TABLE 7 Effect of ISIS oligonucleotides targeting SMN2 on splicing in SMA Type III Mice Brain Lumbar Cord exon 7.sup.+ exon 7.sup.- exon 7.sup.+ exon 7.sup.- PBS 1.0 1.0 1.0 1.0 605918 2.0 0.6 1.9 0.6 605919 1.6 0.7 1.3 0.8 663006 1.5 0.8 1.6 0.8 663007 1.2 0.8 1.2 0.9 663008 1.4 0.8 1.3 0.9 663009 1.3 0.8 1.4 0.7 663010 1.0 0.8 1.2 0.8 663011 1.2 0.7 1.1 0.8 663012 1.3 0.8 1.1 0.8 663013 1.1 0.9 1.0 0.8 663014 1.4 0.7 1.1 0.8 663015 1.3 0.7 1.1 0.8 663016 1.2 0.7 1.2 0.7 663017 1.5 0.8 1.3 0.7 663018 1.6 0.6 1.3 0.7 663019 1.4 0.7 1.4 0.7 669541 1.9 0.5 2.0 0.5

Example 3

Design of Antisense Oligonucleotides Targeting Human SMN2

[0336] Additional antisense oligonucleotides targeting human SMN2 were designed targeting the human SMN2 genomic sequence, designated herein as SEQ ID NO: 25 (GENBANK Accession No. NT_006713.14 truncated from nucleotides 19939708 to Ser. No. 19/967,777) around target region 27062-27079.

[0337] The newly designed antisense oligonucleotides in the Table below were designed as uniform 2-MOE oligonucleotides with mixed backbone chemistry. Each antisense oligonucleotide is 15, 18, or 20 nucleosides in length wherein each nucleoside has a 2'-MOE sugar modification. The internucleoside linkages throughout each gapmer are either phosphodiester or phosphorothioate linkages. The internucleoside linkages of each oligonucleotide is denoted in the Backbone Chemistry column, where `o` indicates a phosphodiester linkage and `s` indicates a phosphorothioate linkage. All cytosine residues throughout each oligonucleotide are 5-methylcytosines. "Start site" indicates the 5'-most nucleoside to which the gapmer is targeted in the human gene sequence. "Stop site" indicates the 3'-most nucleoside to which the gapmer is targeted human gene sequence.

TABLE-US-00013 TABLE 8 Modified oligonucleotides targeting human SMN2 with mixed backbone chemistry Start Stop SEQ OligoID Site Site OligoSeq Backbone Chemistry ID NO 710554 27061 27075 TTTCATAATGCTGGC sososososososs 2 710555 27061 27075 TTTCATAATGCTGGC ssossossosooss 2 710556 27061 27075 TTTCATAATGCTGGC ssossossossoss 2 710557 27061 27075 TTTCATAATGCTGGC sssosssosssoss 2 710558 27061 27075 TTTCATAATGCTGGC ssssossssossss 2 449321 27062 27079 TCACTTTCATAATGCTGG ssooosssoooooooss 1 449322 27062 27079 TCACTTTCATAATGCTGG ssooooooosssoooss 1 710543 27062 27079 TCACTTTCATAATGCTGG ssosssosssosssoss 1 710544 27062 27079 TCACTTTCATAATGCTGG ssossossossossoss 1 710548 27062 27081 ATTCACTTTCATAATGCTGG sosososososososooss 7 710549 27062 27081 ATTCACTTTCATAATGCTGG soosossossossosooss 7 710550 27062 27081 ATTCACTTTCATAATGCTGG sossossossossosooss 7 710551 27062 27081 ATTCACTTTCATAATGCTGG sossossossossossoss 7 710552 27062 27081 ATTCACTTTCATAATGCTGG sossosssosssosssoss 7 710553 27062 27081 ATTCACTTTCATAATGCTGG sossssossssossssoss 7

Example 4

Design of Antisense Oligonucleotides Targeting Human SMN2

[0338] Additional antisense oligonucleotides targeting human SMN2 were designed targeting the human SMN2 genomic sequence, designated herein as SEQ ID NO: 25.

[0339] The newly designed antisense oligonucleotides in the Table below were designed as uniform 2-MOE oligonucleotides with mixed backbone chemistry. Each antisense oligonucleotide is 18 nucleosides in length wherein each nucleoside has a 2'-MOE sugar modification. The internucleoside linkages throughout each gapmer are either phosphodiester or phosphorothioate linkages. The internucleoside linkages of each oligonucleotide is denoted in the Backbone Chemistry column, where `o` indicates a phosphodiester linkage and `s` indicates a phosphorothioate linkage. All cytosine residues throughout each oligonucleotide are 5-methylcytosines. "Start site" indicates the 5'-most nucleoside to which the gapmer is targeted in the human gene sequence. "Stop site" indicates the 3'-most nucleoside to which the gapmer is targeted human gene sequence.

TABLE-US-00014 TABLE 9 Modified oligonucleotides targeting human SMN2 with mixed backbone chemistry Start Stop SEQ OligoID Site Site OligoSeq Backbone Chemistry ID NO 758133 27060 27077 ACTTTCATAATGCTGGCA ssosssosssosssoss 26 758134 27060 27077 ACTTTCATAATGCTGGCA ssossossossossoss 26 758135 27060 27077 ACTTTCATAATGCTGGCA sssososososososss 26 758136 27060 27077 ACTTTCATAATGCTGGCA ssosososososososs 26 758137 27061 27078 CACTTTCATAATGCTGGC SSOSSSOSSSOSSSOSS 27 758138 27061 27078 CACTTTCATAATGCTGGC SSOSSOSSOSSOSSOSS 27 758139 27061 27078 CACTTTCATAATGCTGGC SSSOSOSOSOSOSOSSS 27 758140 27061 27078 CACTTTCATAATGCTGGC SSOSOSOSOSOSOSOSS 27

Example 5

Effect of Antisense Oligonucleotides Targeting Human SMN2 in an SMA Type III Mice Model

[0340] Groups of 3 to 4 wild type and SMN transgenic mice were treated and analyzed as described in Example 2. The antisense oligonucleotides that were administered to the mice are listed in the tables below. The results are presented, as described in Example 2, in the tables below.

TABLE-US-00015 TABLE 10 Scoring for tolerability of ISIS oligonucleotides in WT mice dosed with 700 .mu.g of ISIS Oligonucleotide ISIS No 3 hr Score 494323 0 710543 2 710544 1 710548 0 710549 0 710550 1 710551 0 710552 1 710553 0 710554 0 710555 0 710556 0 710557 0 710558 0 758133 0 758134 0 758135 0 758136 2 758137 0 758138 0 758139 0 758140 0

TABLE-US-00016 TABLE 11 Scoring for tolerability of ISIS oligonucleotides in SMA Type III mice dosed with 35 .mu.g of ISIS Oligonucleotide ISIS No 3 hr Score 494323 0 710543 0 710544 0 710548 0 710549 0 710550 0 710551 0 710552 0 710553 0

TABLE-US-00017 TABLE 12 Effect of 35 .mu.g of ISIS Oligonucleotide targeting SMN2 on splicing in SMA Type III Mice Brain Lumbar Cord Isis No. exon 7.sup.+ exon 7.sup.- exon 7.sup.+ exon 7.sup.- PBS 1.0 1.0 1.0 1.0 494323 2.6 0.9 2.0 0.8 710543 2.7 0.7 2.1 0.7 710544 2.5 0.9 1.9 0.7 710548 2.1 1.0 1.4 0.8 710549 2.4 0.8 1.7 0.7 710550 2.4 0.9 1.9 0.7 710551 2.4 0.8 2.1 0.7 710552 2.4 0.8 1.9 0.7 710553 3.1 0.8 2.0 0.7

Sequence CWU 1

1

27118DNAArtificial sequenceSynthetic oligonucleotide 1tcactttcat aatgctgg 18215DNAArtificial sequenceSynthetic oligonucleotide 2tttcataatg ctggc 15315DNAArtificial sequenceSynthetic oligonucleotide 3tgctggcaga cttac 15415DNAArtificial sequenceSynthetic oligonucleotide 4cataatgctg gcaga 15515DNAArtificial sequenceSynthetic oligonucleotide 5tcataatgct ggcag 15615DNAArtificial sequenceSynthetic oligonucleotide 6ttcataatgc tggca 15720DNAArtificial sequenceSynthetic oligonucleotide 7attcactttc ataatgctgg 20815DNAArtificial sequenceSynthetic oligonucleotide 8ctttcataat gctgg 15912DNAArtificial sequenceSynthetic oligonucleotide 9tcataatgct gg 121015DNAArtificial sequenceSynthetic oligonucleotide 10actttcataa tgctg 151112DNAArtificial sequenceSynthetic oligonucleotide 11ttcataatgc tg 121215DNAArtificial sequenceSynthetic oligonucleotide 12cactttcata atgct 151312DNAArtificial sequenceSynthetic oligonucleotide 13tttcataatg ct 12 1415DNAArtificial sequenceSynthetic oligonucleotide 14tcactttcat aatgc 151512DNAArtificial sequenceSynthetic oligonucleotide 15ctttcataat gc 121615DNAArtificial sequenceSynthetic oligonucleotide 16ttcactttca taatg 151712DNAArtificial sequenceSynthetic oligonucleotide 17actttcataa tg 121815DNAArtificial sequenceSynthetic oligonucleotide 18attcactttc ataat 151912DNAArtificial sequenceSynthetic oligonucleotide 19cactttcata at 122015DNAArtificial sequenceSynthetic oligonucleotide 20gattcacttt cataa 152112DNAArtificial sequenceSynthetic oligonucleotide 21tcactttcat aa 122212DNAArtificial sequenceSynthetic oligonucleotide 22ttcactttca ta 122312DNAArtificial sequenceSynthetic oligonucleotide 23attcactttc at 122415DNAArtificial sequenceSynthetic oligonucleotide 24agtaagattc acttt 152528070DNAHomo sapiens 25ccacaaatgt gggagggcga taaccactcg tagaaagcgt gagaagttac tacaagcggt 60cctcccggcc accgtactgt tccgctccca gaagccccgg gcggcggaag tcgtcactct 120taagaaggga cggggcccca cgctgcgcac ccgcgggttt gctatggcga tgagcagcgg 180cggcagtggt ggcggcgtcc cggagcagga ggattccgtg ctgttccggc gcggcacagg 240ccaggtgagg tcgcagccag tgcagtctcc ctattagcgc tctcagcacc cttcttccgg 300cccaactctc cttccgcagc ctcgggacag catcaagtcg atccgctcac tggagttgtg 360gtccgcgttt ttctacgtct tttcccactc cgttccctgc gaaccacatc cgcaagctcc 420ttcctcgagc agtttgggct ccttgatagc gttgagtgga ggccctgccg cgacttggca 480gtagcttatt ttgttcactc ctctctggct ggtgtggggg aggtgggggc attaggccag 540ggtgaagcag gggaaccact taggagtctg ttaagatgat ctgaacttca gaacaagatg 600ttattaacag agtgaaagta tttggattct gggtatattt tgaaatcgga ggcaacaggt 660ttttcagata gattcgataa cggaggttat cctgaatagt tgaaaagata aagttgcctt 720ttgctgaggt gggaaagaga agattgccag tagagcaggt ttctcaggag ttcagtcttg 780ggcatagcat ggtaggggtg aatttggctg gagtgagttg gagagtagga gaagagaaat 840ccaaggcaac atttgaccag cctgggcaac atagtgtgac tccgagtctg caaaaattag 900acgggtgttg tggtgcgcgt ctgtggtctc agctacctgg aaggttcagg ccttggaagg 960ctcagggagg tggaggctgc agtgatctgt gattgcgcct ctgcactcca gcctgggcga 1020cagagccaga ccctgtctta aaacaaaata aacggccggg cgcggtggct caagcctgta 1080atcccagcac tttgggaggc cgaggcggcc ggatcacaag gtcaggagat cgagaccatc 1140ctggctaaca cggtgaaacc ccgtctctac tacaaataca aaaaattagc cgggcgtggt 1200gacgggcgcc tgtagtccca gctactcggg aggctgaggc aggagaatgt catgaagccg 1260ggaggcggag cttgcagtga gccgagatcg cgccactgca ctccagcctg ggcgatagag 1320caagactccg tctcaaataa ataaataaat aaataaataa ataataaaaa catcggtagg 1380catatttcaa ggaattctat ttaaaaaaaa tttttttaga gacaagttcg ctctctgtgg 1440cccaggctgg agtacagtgg catgatccta gcccatggca gcgttgatct cttggcctca 1500agcgaccctc ctttggagtc gctgggccta aaggagtgag ccaccacgaa attttattat 1560aaatggaggg tagagaaatt gggcaataaa tggaggggga agtgagttaa gaggaatttt 1620aattatgtgt gtgtggtttt aaaagagggg ggtcttgctc tgttgcccag gctgctgggg 1680tgccagtggc gcaatcatga atcactacag ccttggactc ctggcctcaa gctatcctcc 1740cacctctgcc tcccaaagta ctgggattac tagtgtgagc cactgcacta agataggagc 1800aacatgtttc agcatgtttg tgggttgata ggaaagatga gaatgggaaa gttgatgtcg 1860gaaagaagac aatggctaga gcaatgtcct agagtaggta agaagggatg gatttggcct 1920ttgttggaaa cattagcggt tcttttggtg acagctatat agttaacaca tctatgatac 1980gtgaatgggc agataggatg gcaggagatt ttgaaagttc tcttgattct tactgttctc 2040ttagtgaaag aagcaaggtt atcagctaga agctgggatg ggagaggaaa gagaagatgg 2100gaagtagata gttctttaga agagtgggca agggttggac tagggaagtt tagtggaaat 2160attgctaggc aacataaaga gcctacttga gattcgtggt catgagttga aggagaccag 2220acagcaagat tgtgtatgag ggcacccaca gagtaaatgg agagttgaaa ttaatgcagt 2280tgtgatttta ccacgtggat atgaagaagt gagggggaga agtacaaagg agttctctta 2340atgattgacc atggaattta agctggctaa gaaaggaagt gagaggccgg gcgcggtggc 2400tcacgcctgt aatcccagca ctttgggaga ctgaggtggg tggattacct gaggtcagga 2460gtttgagacc aacctggccg atatggcgaa accccatctc taataaaaat acagaaaaat 2520tagccgggaa tggtggcagg tgcctgtaat cccagctact caagaggctg tggcaggagt 2580atcccttgga cccaggaggt ggaggttgca gtgagccgag atcacgccac tgtactccag 2640cctggacgat atagtgagac ttcacctcaa aaaaaaaaaa aaagaaagga agtgaggatt 2700ttaagaccct gagagacagt ttaaaaagtg ggaggatcgg ccgggcgctg tggctgacac 2760ctgtaatccc agcactttgg gaggccgagt tgggcagatc acaaggtcag gagttcgaga 2820ccagcctggc caatatggtg aaaccttgtc tctactaaaa atacaaaaat tagccgggca 2880tggtgtcacg tgtctataat cccagctact cgggaggctg aggcagaaaa attgcttgaa 2940cctgggaggc agaggttgca gacagctgag atcactccat tgcactccag cctgggcaac 3000aagagcaaaa ctttgtcttt aaaaaaaaaa aaaaaaaaag aatacaaaaa ttagccgggc 3060gtggtggcgc gtgcctataa tcccagctac ttgggaggct gaggcaggag aatcagttga 3120acacgggagg cgaggtttgc agtgagccga gattgcgcca ctgcactcca gcctgggcga 3180cagagcagga ctcctcttgg aaaaaaaaaa ttagctgggc atggtggcag gtgcctgtag 3240tctcagctac tagggaggct gaggcaggaa aatcacttga acccgggatg tggagtttgc 3300agtgacccga gatcgtgcca ctgtactcca tcctgggcga caaaatgaga ctctgcctca 3360aaaaaaaaaa aaaaaaaaag tgggaggatc aatgtactgc cagtcctaat gaagtggaat 3420gattgtcccc atcaaatcac tagtaggagt aagttgcaga gcctagaagg tgatggttaa 3480gagagtggga ttcttgaaac tgcatttatg gagaggttgt ggttattggt tataataaat 3540aaatacagtt gaagtgagtg agtagctgag atttggggat gtatcagttc attcttacac 3600tgctacaaag acatacctga gaccaggtat ttataaagat aagaggttta atcagctcac 3660agttctgctg cctgtacagg cttctcttgt ggaggcctaa ggaaacttac agtcatggtg 3720gaaggtgaag gggaaacaag cacagtcttc acatggccag caggagagag agagaagggg 3780gaagtgctac atactttaaa acaaccagat cttgtgagaa cgcttatcag gaaacagcac 3840ttggggatgg tgctaaatca ttagaaatca cccccatgat ccagtcgcct cctaccatgc 3900ccacctccaa cactggggat cacaattcag catgagattt gggtaggaac acagagctgc 3960accacatcag aggatgtaca agattgtggt ggagaggagt ttagagacct gcaaatatag 4020ggtaattgaa gggatcatct acatggatat ttaaatcacc aaaaattatg acaggagtag 4080tgttggagag agaactgcga tgtaaacatt aaggaatgag gaagagtgac tcggtaggct 4140gtaggtgact gcaataggaa acgataatag actgtgagtc tggtgacaag attttccttc 4200tttctttttt tccccccccc cgagacaggg cctctttttg ttgcccaggt gggagtgcag 4260tggcgcgatc acggctcact acaacctcct cccaagctca agggattctc ccacttcagc 4320ctctcaagta gctggaacta caggtgctga ccaccatgcc tggctacttt ttgtcaggat 4380tttcaaggct gggaattttg agaggggaat ggaggagaat aatctgaaag tgcaagtaag 4440gagcagggaa gatttctttt ttcttttttt tttttttttt tgagtcggag tctggctcag 4500tcgcccaggc tggagtgcag tggcgagatc tccgctcact gcaagctccg cctcccgtgt 4560tcacgccatt ctcctccttc agcctcccga gtagctggga ctacaggcgc ccgccaccac 4620gcccagctaa ttgttttttt gtatttttag tagagacggg gtttcaccgt gttagccagg 4680atggtctcaa tctcctgact ttgtgatccg cccaccccgg cctcccaaag cgcttgggat 4740tacaggcgtg agccaccgcg ccagccagag cagggaagat ttcttcccca catctccagt 4800aggtacagtg atatgaagtg tgtggaggag aaaagaggaa acatctatca tttgagatgg 4860ctgcgaaagg aaaaggcatc ctcagggagc tagattttac ttagagcaag aaatgaaggg 4920atgattcaga ggttaaaaga gtggatttta tgaattactc aagggagcac agtggaagtt 4980tcaggaagtg gtaggagaag gtagaagatg gcagggtgtt gggaataatt tgagaaatct 5040gagctactgg aaatgactga gaatcagata taaaggcagt cctggtggtc cgttctggct 5100gccgttgctg tgtaacgaat ctgccaaaac ttagtggctt gaaacaacaa agaacatttt 5160attatctctc attgtttctg tgggttagga atttgtgaga gccgtgctgg gcagttttcg 5220tgcggctgtc tcgtggttgc acctacatag ttgctagagc tacagtagct ggggactgag 5280cagctaggga ttggcaggct atctcttttt ttcatgtagt ctcatgaaga tttctttatg 5340tggtttcaat gtgtgggctg gtttggattt ccttatagca tggtggcctc agttggattg 5400ctgttttgtg atccttttca tccctccttg tcctgtcccc agacaaccac tgatctactt 5460tctgtcacca tagattagcc tgcattttta agaattttta taaacgtgga atgatagagt 5520accttttttg tcacgtttct tttatttatc atagctattt tgattttcat ccattttatt 5580gctgagtagt atcccattgc atgtatatac tatactgtat tcattcgctt gcttgtgaac 5640atttgggctt tttccagttt gggactgtta acaagtagag ccactatgaa tattagtgta 5700taagacttca tatagccaag gctggcagat cgcttgagcc caggagtttg agaccagcct 5760gggaaacatg gtgaaacctc tatttttatt ttaaaatcaa aaattaaaaa ttttctataa 5820aaaattttaa agaagacttt gtatagacat acgctttcat ttttcttgag tgaatactta 5880ggtctcaggg tagatgtatt ttaagtcttt aaggagctgt caaactcttc ctcaaagtgg 5940tggttgtacc atgttacttt ttaatataac agagattaat tgagcaaaga aaaattcaaa 6000agttggacag cccccacaac taaataggtt cagaacagct cccccatttt gcattttgac 6060cagcaatgta tgaaagttcc atttgctcag tgtccctgca aacacctggt atggtcagtc 6120tttttaattt taggcattat aatagatata gtggcttctt gtgattttaa ttagcatttc 6180ctaatgacca gtgctgctgt tgatcatttc atgagtgtat ttgccatccg tatatctttt 6240ttggtgaagt gtctattcaa atcatttggg tttttttttt ttttgttttt tttttttgga 6300gacagtgtct cactctgtca cccaggctgt tgtgcagtgg tgcaatcaca cagcctactg 6360cagcctccac ctcctgcgct cagtcttctt gtctcagcct tctgagtagc tgaaattacg 6420agcacacgcc acaatgcctg gctaattttt taaaattttg tagaaacaag gtctcattat 6480gttgcctggg cttgtcgtga actcctgggc tcaagcaatc ttcctgcctc agcctcccaa 6540agattgggat tgcaagtatg agccactgca cccggccaac ttacccatct tttaattgaa 6600tttttttgtt gttgaggttt gagagttctt catgtttgct gggtacaata tctttatcag 6660ataggtaact tgcatgtatt ttctcccggt ttacactttg gtttttcatt ttgttaacaa 6720cgtcttttta agaacagaaa atcttaattt tgctgaaatc taatttttca gttttttctt 6780tgatggtttt gagagaggag gtaaaaaaag actaggtaag ccgatagtta gacagagtcc 6840tcggtagaac ttcccttcta acaaaaagca gcccaagaaa tcacttctct tctaacaagg 6900agcagcctgg aagatcgggc tgtaaacatg tataaggaag cagctctggc acagaggggg 6960agcttcctgg gtaatcagca agcttcacat acgtaaggtg ggtatgtgaa gtaaacacag 7020tatgtgaagt aaacacagtg gaccttagta catactcaga taaggaagct ggaagcttgc 7080atgttgtgag ttgttggggt tgcctgcagc tgcacggaga gaaaggggta cctggggcca 7140ggcatgtcca ccatggtggc tccacctccc cttatttagc acatgcacaa taggaaagag 7200ataagcaatg tggagtagct caggccaagg acctgcctgc ataataaaag gttggggtgg 7260gggatgccag agattcacgc tctgtgcaga tggcaacacc tggtcctaac tggttttttg 7320ctccctatgt gtagataagc tacccccttc ccattagctc atttataaaa atgcttgcat 7380ttcactgtgg aatgggaact cttttcagga cctctctctg caggagagag ctagtctctt 7440tcttttgcct attaaacttc tgctctagcc tcacaccctt ggtgtgtcag cgtccttgat 7500ttcctcagcg tgagaccaag aacctcgggt gccaccccag gcaacaaggc catttcagtt 7560tgttcttttg ttataggcaa tccatgatca cagatttttc tctctttttt ttttttacac 7620agtttagagt tttagtttta cacttaggtc tgtaatccat tttgtattaa ttcttatatg 7680tggctcagtg taggtggaaa tttggtttgt ttttgcataa ggatttccaa tagttttacc 7740accatttctt gaaactacta tgctttctct attaaaccac atttgtaact ttagttaaaa 7800tcagtcacat atatcacagg gctatttctg actctcaatt ctgttacatt gtctattagt 7860gtatattgat gtcagtacta cacttttaat tactattgct tcagggtatg tcttgtaaac 7920caaaaataaa attataggcc ccccccgccc ctgcacaacc aactgaatgg acccatcctc 7980tcagccaagg gcattccaaa attaacctga aaaactagtt caagccatga tgggaagggg 8040gagttggaca tgtctcatca caccctacta ccttttggaa ttactgatag aacagactct 8100taaagtctga aaagaaacat ttacaaccta ccctctctga agcctgctac ctgggagctt 8160catctgcatg ataaaacctt ggtctccaca accccttatg gtaacccaaa cattcctttc 8220tgttgataat aactctttca actagttgcc aattagaaaa tctttaaatc ttcctatgac 8280ctagaaacct ccctaccccc actttgagtt gtcctgcctt tcctgacaga actcatgtac 8340atcttacata tattgattga tgcctcatgt ctccctaaaa tgtataaaac aaagctgtac 8400cccaccacct tggggacatg tcatcaggac ctcctgtggc tgtgtcatag gagcgtcttt 8460aactttggca aaataaactt tctaaattga ttgaaacctg tcttagctac ttctggttta 8520cagtcttaaa gttagataat gtaaattgtc cagctttggt ttatttttgt ccttagtagt 8580tccatataaa ttttagaatc agcttttcaa tttaatacac tactttcctc ttagatccac 8640aattaaatat atttgatgct aacaattctg ttttatgttt ttcgtttttt ttttttgaga 8700caagagtttc gctcttgttg cccaggctgg agtgcagtgg cgcgatcttg gctcaccaca 8760acctccacct cccaggttca agcaattctt ctgcctcagc ctcccgagta gctgggatta 8820caggcatgcg ccaccacgcc cggctaattt tgtattttta gtagagacgg ggtttcacca 8880tgttgatcag gctggtcttg aactcctgac ctcaggtgat ccacccacct cggcctccca 8940aagtgttggg attacaggcg tgaaccacca tgcctggcca gttctgttat ttttaaaacc 9000caagtttccc tggtcatatc ttggttggat gaagcgtatt ttcaatagat taccctggaa 9060aggctagtga gtacggtatt cttctacatt ttagactttt cttagtcttg ctacttcaag 9120gacagctagg ctgcatataa aattcttggc tcatactttt tccccataaa tttctatgag 9180aaagtctaat gataactgat tttctttatt ttgtaactta gtctttttgc ttagaggctc 9240tctgaggatg ggagggggtt cttcctccca tccctaggaa tttttctttt ttttaaattc 9300ctaatcacta gaccaccagg aagattgttt gttttgtttt gtttttattc ttcagggacc 9360ccatttatac atacgttaaa taaatactgt ttgccaatgt atcaaccatt ttgcttctta 9420tttatttttg ttcctttggt tctttttcat ggctttgctt tggtgctcct tagattttca 9480gtcagatgta tttgtccttg ggtaccttgt aatcagtatt accttttctt ctgtcgcttt 9540gttttctgtt cgttttgaaa ttacttgttt cctggtctgg caataacagt tgagatatga 9600ggagtttgag ctgccatctg tctatgtatc ttgctttaag actgcactct tctattgata 9660tcactggcct tgattttgtg atttctttat ttcttcagga ccacccttca ttttctactg 9720tttgcttcct ttttttttga gatggagtct cactctgtca ctcaggctgg agtgcagtga 9780tcttggctca ttgcaacctc tgcctcccgg gttccagcaa ttctcctgcc tcagcctccc 9840aagtatctgg gactacaggt gtgcaccacc atgcccggct aagttttgta tttttaatag 9900agacggggtt ttgccacatt ggcaggctgg tctcaaactc ctgatgtcaa gtgatccacc 9960caccccaccc acctctgcat cccaaagtgc tgggattaca ggaatgagct gccgtgccca 10020gcctcccccc tacccccctt tttttctttc gagacagaga ttataggtgt gagccactgg 10080acccagcctg tttttattcc ttttaccaaa tctccaagga atatcttccc ttccaagtgc 10140gaatgtaacc ttaagtcagt taacctcttt gtgattactt ttcttatctg caaagtgact 10200taatgatctt aagtactttt tttttttgag acagggtctc actgtcaccc tggctggagt 10260gcagtggcac gatctctgat ctccactcac tgcaatctcc tcttccctgg ttcaagcggc 10320cctcccacct tagccttctg ggtagctggg actacagatg tgaaccacca cgcccagcta 10380atttttgtac tttttgtaga gatggggttt tgccatgttg cccaggctgg gattattaag 10440tactttttat catacagcaa gattgacatt ttatattgga atacatttgt ctctatataa 10500cggagattaa caggaaaatg acaagcctgg gtgcggtggc tcatgcctgt aatcccagca 10560ctttgggagg ctgaggtggg aggatcactt gaggtcagga gttcgagacc agttttgcca 10620agatgatgaa agcccatgtc tactaaaaat acaaaaatta gcccagcttg atggtgggcg 10680cctataatcc cagctatttg agagactgag gcaggagaat cacttgaacc tgggcagcag 10740aggttgcagt gagccgagat catgccactg cactccagcc tgggtggcat agcgagactc 10800ttgtctcaag agaaaacaaa acaaaacaaa aaaaaaacag gaaaatgaca aaaagtaata 10860ttacaactca gtgaatttta taacaaactt ttttggaatt cattgactaa tactatacca 10920aatccaaaat actctctagt ataccaaatc caactctacc ctatagtata aattggattc 10980tatttggact tgtctcacta atccctcata cagtgtgttt tattttttat tgaagtaaaa 11040aaatttgtca ttttaaccat ttttaagtat atagttcagt aatattaagt atgttcatgt 11100tgttgcgcaa tagatcttcg gaagtttttc gtcttgcaac ctgaaactct acccattagc 11160aaattcccat ttctccttac acttagccct tggtaatcat cattcttttt tttttttttt 11220tgagatggag ttttactctt gttgcccagg ctggagtgca atggtgcaat ctcgactcac 11280cacaacctcc gcctcccagg ttcaagcaat tctacctcag cctcccgagt agctgggatt 11340acagtcatgc accaccacgc ccggctaatt ttgtattttt agtagagaag gggtttctcc 11400atgttgaggc tggtctcgaa ctcctgacct caggtgatct gcccacctcg gcctcccaaa 11460gtgctgggat tacaggcgtg agccactgcg cctggcccat tctttctaat tctataaatt 11520tgactactta gttaccttac ataaataaat tcttatagtt agtgttattt ttgcttccat 11580gccttttttg ttgttgttca tgctcttact tggaatgcgt tctattttgt ctacctatgc 11640acatcctgtt gggttttttt tttttttggg ggtttttttt gttttttttt gttttttttt 11700cccagacaag gtctcaattt gttacccagg ctggagtgca gcggcgccat ctccactcac 11760tgcatcctca acttcctggg cccaggtgat cctctcgcct cagcccctgc aggtagctgg 11820gactataggc atgtgccacc atgcccagct aaatttggtt tttttgtttg tttgtttttg 11880agacagagtc tcactctgtc acccaggctg gagtgcagtg gcacaatctc agctcactgc 11940aatctctgcc gcccgggttc aagtgattct cctgcctcag cctcccaagc agctgggatt 12000acaggtgact gccaccacgc cagctaagtt ttgtagtttt agtagagatg gggtttcacc 12060ttgttggcca tgctggtctc gaactcctga cctcgtgatc tgcctgcttc tgcctcccaa 12120agtgctggaa ttacaggcat gagccaccac gcccggccag aatttttgta tttttagtag 12180acacaaggtt cttaccctgt tgcctaggct ggtctggaag tcctggactc aagcaattca 12240cctgccttgg cctcccaaaa tgctgggatt acaagccacc atgcccggcc taaatcctgt 12300tgttttgttt tgttttattt tgttttgttt tgttttgttt gttttttgag acagagtctc 12360gctatgtctc tcaggctgta gtgcagtggc gcgatcttgg ctcactgcca cctctgcctc 12420ccaggttcaa gtgattctcc tgcctcagcc tcccaagtag ctgggattac aggcatgtgc 12480tactatgtcc ggctaatttt tgtattttta gtagagacag

ggtttcacca tgttggccag 12540gctggtctcg aactcctgac ctcgtgatcc acccacctcg gccacccaaa gtgctgggat 12600tacaggcgtg agtggttttt atttcttagg ccggtttcct ccatatgatc ttgcagtaga 12660cattaatttc tttccttttt aattaaaata ctgtttgtat ttcacatttt gatgtttgtt 12720aagatttgtt ttatattgtt ttttgttttg tcttgtgtga tagtcttaaa tccctagtta 12780gataataact ggagagtacc atgtttctat atatctctca gtgacttgca cagtgctagc 12840agatagtgct aaaaaattat ttattattat tattattttg ttattgttgt tgttgttgtt 12900agacagggtc ttcctctgtc acccaggcta gagggcaatg ggatgatcat agcttactgc 12960agcctccaac aactgggctc atgtaattct cctgcctcag cttcccaagt agctgggatt 13020acaggcatga gccaccatgt ctggacaaaa atatttccag gtgcagtggc tcatgcctgt 13080aattcccaca cttgggaggc cgagcgaggc tggaggatca cttgagccta ggagttcaag 13140accagcttgg ctaagatggc gagaccccgt ccctacaaaa aattttaaaa actagccagg 13200catggtggca tgcacctata ttcccaacta ctcagtgggc tgaggtggga gggtcatttg 13260aacacaggaa tttgagggga gaaaaaaaga agagagaaag agaagtgaag gaaggaagaa 13320aggaaggagg gagggagaga agaaagaaac gaaagaaagg aaaagaaaag gaaggaaaga 13380aaattggtac caggaaagca ggaaagggaa atggaagtaa aaaaataata ataataataa 13440aatgaaaatt ggttagtcac tattaacaat ttgtatcctt ataatctgga aacattataa 13500tttcaaaaga aaaaatattc tttggatcat aggttctgag gtcagaacag cattcccgta 13560gtctagatga agtcaagttt tatctgatct taattgaaat aaatatagct ggccttgaac 13620aaatctactc atggtatgtg gataggaatt aaattgtagg ggcattcact tgatggcatt 13680cattcttaga acatttacct atgtctagct tttggagtaa agtcacataa cctctaacca 13740ggtaagtttc ctgtggcttt atttaggatt ttaaatactc attttcagtg taattttgtt 13800atgtgtggat taagatgact cttggtacta acatacattt tctgattaaa cctatctgaa 13860catgagttgt ttttatttct taccctttcc agagcgatga ttctgacatt tgggatgata 13920cagcactgat aaaagcatat gataaagctg tggcttcatt taaggtatga aatgcttgct 13980tagtcgtttt cttattttct cgttattcat ttggaaagga attgataaca tacgataaag 14040tgttaaagta catgttattc agttttcatt ttgaagatta gatggtagta tgagttagtt 14100aaatcaggtg atatcctcct ttagaagttg atagcctata tatgtcatcc tttgtggagg 14160caatttaaat aaaatttaaa acatttattc ctggctgggt atggtggctc actcctgtaa 14220tcccagcact ttgagaggct gaggcgggtg gatcacctga ggtcaggagt ttgagaccag 14280cctggccaac atggtgaaac cccgtcttta ctaaaaatac aaaaattagc caagcatggt 14340ggcacgtgcc tgtaatccca gctgcttggg acactgaggc aggagaattg cttgaacctg 14400gggggcagag gttgcaatga ttgcaccact gcactccagc ctgggcgata gagtgagact 14460ccatctcaga aaacgaacaa acaatgtatt ccttttagta tttttacatt gtatcaaact 14520atggaagtcc tctaattgag attaataaga aaaagacaat ctgaattata attttaaaca 14580tttaacaagc atgtagtaaa ataatgatga agataaatag cattagtaca gcaattaata 14640tttgtagcat gctgacagtg ctctgtgtgc gtttcatata ttaaattact ctaatcatcc 14700caaatcctgt aagttgggta tcaattcaag tgttcctatt gggtaggaat atacagttct 14760tttaggaaat gtagtatggt tctgtgtctc aaacaggaca cttacacagt tggccaacat 14820catcaccttc tccattctct gagatgttta gtcttactga gcactaaata tgggtcatca 14880atagtccaga ctaccttgag caaacaatag tccagactac cttgagcaaa cagagcatat 14940actcatacag tgtataaaga gcaccaagca tacagatttc atgtctttct catagttact 15000cttgtaacat gagctaaaga tcagacctct atgtcacctt tgtaactgat ttctagattt 15060tttttttttt ttgagatggg gtcttgccct gtcacccagg ctggagtgta gtggcgtgat 15120catgcctcat tggagccttc aactcatgag ctcaaacaat cctcctacct cagcttcctg 15180agtagttggg accacaggtg tgtgccacca cacccagctc atttttgtat tctttgtaga 15240gatgcagtct caccctgttg cccacgctgg cctggaactc ctgagctcaa aagatccctc 15300cgccttgacc ttccaaagtg ctgggattac aagcatgaac cactgcaccc ggcctagatt 15360tttaaatgtg ctttccagta tacactgaaa ctagaagtcg actaaagaat taccaagaga 15420attctataaa atagagattg aaatggggct cgatgtggga tgggttggtg atattgcagg 15480gagaagtaat ctgagtaaag gaggaaaaga actgatttgg gaaaacgata gttttagtag 15540tgagtttgag tatgaattaa gttgagattg aatttgaatt aagttgaggt tgaatatgaa 15600ttaagttgag gttgagtttg aggtatgaat taagatgtga aattgatcat tggaaatgtt 15660agattgagaa aagtcacagc tggattaata gcttcagaag tgtgtttgca gacagttgca 15720actaaagtaa taagaataga tggccttggc cgggcgcggt ggctcacgcc tgtaatccca 15780gtactttggg aggctgaggc gagcaaatca cgaggtcagg agttcaagac cagcctggcc 15840cacatggtga aaccccgtct ttattaaaaa tacaaaaatt agctgtgcac agtggtgcac 15900gcctgtaatc ccagctactc gggaggctga gacaggagaa tcgcttgaac ctgggaggtg 15960gaggttgcag tgagctgaga tcagtgtgac tgcactccag cccggtgaca gagtgagact 16020ctgtgtaaaa aaataaaata aataaaataa tggccgtaag caagtaaaga aggatggcca 16080gctcttattg ggaatgccta aatctaaggc ttgatcagaa gtaatgaaac cgttggggcc 16140ctacattgct atgacatcca aagggccatg aatatcagga agaaagataa ttaacagggt 16200ctaatgttac agagaggttg agagcaagga gatttgatta aaagggtctt tagagctgat 16260gtcaggtgta tgatgccttt aagagcagtt tttatagtgc agggggtggt caaaagagaa 16320aataggtgct ttctgaggtg acggagcctt gagactagct tatagtagta actgggttat 16380gtcgtgactt ttattctgtg caccaccctg taacatgtac atttttattc ctattttcgt 16440agcatgctct aaagaatggt gacatttgtg aaacttcggg taaaccaaaa accacaccta 16500aaagaaaacc tgctaagaag aataaaagcc aaaagaagaa tactgcagct tccttacaac 16560aggttatttt aaaatgttga gatttaactt caaaggatgt ctcattagtc cttatttaat 16620agtgtaaaat gtctttaact taagtgatta gtacagtgtt tctattgaca tatacttata 16680caacttcaaa aacaactatt aaattttctg ttatttagga acatgcatat tagtcatgaa 16740agtataaaga attagatggg aatgataaat gctaaaatca ggacatgtgt tccatttgtg 16800aatggaaggc agggagaagg tgccgtttgg aaggagtacc caagagccgt aagctgaatt 16860ggcagtgttt tacatcttaa gctgagagat agattttttt ttcccctttt tctttaaaaa 16920ctctaaaact gttaattcca aggaacccag aagtctaggt agattatttc tgctagttaa 16980aagcagtagt cctgaaagct gaatattttg gtgtcttttg agccaacttt agtttcatca 17040ttaccaaggg ggaagagagc taacagttga tgagcacttg ctctaggcca gtccagagtg 17100ctgggcacca tacgcatttt atctccctcc cgctattcac aacaaatatg ggaggtagtt 17160tatattatag ccatctaata agatggggaa actaagactc aaagagattc agaaacttgt 17220ccatgattat aaatgtaaga gagttggaat tcagatttat gtatttagac cccaagcctt 17280tctcattaca tcattttgcc ttccaaatct ctaccctcta tccttcacct ccccactgat 17340caaaacgaga tgatagtttg ccctcttcaa aagaaatgtg tgcatgtata tatctttgat 17400ttcttttgta gtggaaagtt ggggacaaat gttctgccat ttggtcagaa gacggttgca 17460tttacccagc taccattgct tcaattgatt ttaagagaga aacctgtgtt gtggtttaca 17520ctggatatgg aaatagagag gagcaaaatc tgtccgatct actttcccca atctgtgaag 17580tagctaataa tatagaacaa aatgctcaag aggtaaggat acaaaaaaaa aaaaattcaa 17640tttctggaag cagagactag atgagaaact gttaaacagt atacacagtt gtcagtttga 17700tccaccgagg cattaatttt ttcttaatca cacccttata acaaaaacct gcatattttt 17760tctttttaaa gaatgaaaat gaaagccaag tttcaacaga tgaaagtgag aactccaggt 17820ctcctggaaa taaatcagat aacatcaagc ccaaatctgc tccatggaac tcttttctcc 17880ctccaccacc ccccatgcca gggccaagac tgggaccagg aaaggtaaac cttctatgaa 17940agttttccag aaaatagtta atgtcgggac atttaacctc tctgttaact aatttgtagc 18000tctcccatga aacttttgta gcttaaatac acaagaattt tttgaaaagg aaataagata 18060atgatgcaaa atagttaatt ttttaaaaaa atgttagaca ctgcagtgga tgcaacaaaa 18120tactttatat gaaagattta tccagttaac ttttgtggag tattaggtat tagactaata 18180attagcacac ttacttaagt tagaaagtat aataatgcgc cggacgcggt agctcacgcc 18240tgtaatccca gcactttggg aggccaaggt gggcggatca caaggtcagg agatcgagac 18300catcctggct aacacggtga aaccccatct ctactgaaaa tacaaaaaaa tttgccgggc 18360gtgatggcgg gcacctgtag tcccagctac tcgggaggct gaggcaggag gatggtgtga 18420accccggagg cagagcttgc agtgagtcaa gatcgtgcca ctgcactcca acctgggcga 18480cagaatgaga ctccatctca aacaaaaaaa caaaacaaaa caaaaaaaag tgtaataata 18540atttatcatt agctggatga tatgctgttg tttcccatgt cacctgtata agatatgtaa 18600aataagaaca cattatttac atctaatata gataaaatcc tgaggcgctc tcagattgtt 18660ttgtagagtt caaatgtaaa tattgttttc atttatggtc cttttggtta taagtaacag 18720aaatcaactc taaaaagatt tttattatag gttagattat gtcatggaac cttaaggctt 18780gtccctttct agttcttttg tgtaaagcgg tgatttcttc catggaggga atggtattta 18840ggcaattttt tttttttttt cgagatggag tcttgctctg tcgctcaggc tggagtgcag 18900tggcaccatt tcagctcact gcaacttcca cctcctgggt tcaagtgatt ctcctgcttc 18960agcctcccaa gtagctgaga ttacaggcac ccgccaccac acccggctta ttttgtattt 19020ttagtagaga tggggtttca ccatgttggc caggctggtc ttgaactcct gacctcaagt 19080gatctcccca ccttggcctt ccaaagtgct aggattacag gcgcctagcc taggcagtca 19140ttttcaaaaa acaagcatga ctcaccaaaa gttttaagat tttctgtgat aatgttctta 19200ttgaggctta cattatatta cagtttcttg aatctaaaat gatgtaccct cttagaatat 19260atacatcatg cttcattggt ctcagggggc tgatttttat aaggagagat ttgctagttt 19320tcacaatatg tcctctaagt tggcatgtat agctaaacag gctttcataa aaatatacaa 19380tttagttaat gaaatttggg atatagtctt ttatgattga aataattttg ctaaatagac 19440tgtctctgat ttattaggta atcaccactc ttattttgtt ttacttcctt aatgtctaca 19500tagaaaggaa atgagaaaaa tccagaggtt gtcatttgac ttatgagtct gtttgacttc 19560aggatttggt acatgaaatt tcacttaatc tttttgatat gtataaaaca aatattctgg 19620gtaattattt ttatcctttt ggttttgagt cctttttatt cctatcatat tgaaattggt 19680aagttaattt tcctttgaaa tattccttat agccaggtct aaaattcaat ggcccaccac 19740cgccaccgcc accaccacca ccccacttac tatcatgctg gctgcctcca tttccttctg 19800gaccaccagt aagtaaaaaa gagtataggt tagattttgc tttcacatac aatttgataa 19860ttagcagaat agaggattgt aaaatgtcat tgtagaacat cccttgggcc agattctaat 19920gggtagaaat ttgaactaaa cctctgggtt ttgtttgttt ttaatgcctt tctgttaccc 19980agatgcagtg ctcttgtagt cccaagtcta agctctaggt tgccttcttt cctggcagaa 20040gttggtgtct atgccataag gaggtagttc ctgttagaag ggatttaatt ataccttata 20100taaggaatta gtgtttgccc ttctaggtat agttggatgt tagcttctga tgtaaactgg 20160atttcttttt ctttctctct cttttttttt ttttgttttg gaggcagagt tttgcccttg 20220taccccaggc tggagtgcag tggtgtgatc tcagctcaca gcaacctccg cctcctgggt 20280tcaagcaatt ctgcctcggc ctcccaagta gctgggatta caggcgactg ccaccacacc 20340cggctaattt ttgttttatt agtagagatg gggtttcacc atgttggcca gactgatctt 20400gaactcctga cctcaggtga tccacccgcc ttggcctccc aaagcgctgg gattacaggc 20460gtgagctgcc gcacccagct gtaaactgga tttctaatgg tagattttta ggtattaaca 20520atagataaaa agatactttt tggcatactg tgtattggga tggggttaga acaggtgttc 20580tacccaagac atttacttaa aatcgccctc gaaatgctat gtgagctgtg tgtgtgtgtg 20640tgtgtgtgtg tgtattaagg aaaagcatga aagtatttat gcttgatttt ttttttttac 20700tcatagcttc atagtggaac agatacatag tctaaatcaa aatgtttaaa ctttttatgt 20760cacttgctgt cttttcgtcc tcgttaaatt taattttgtt ggtcttttgt tgttattggt 20820tggttttctc caaatgctag ctatgttaag aaatttaagg ccaggtacag tggctcatgc 20880ctgtaatccc ggcattttag aaggctgagg caggaggatc acttgagctc aggagtttga 20940gaccagtctg ggcaacatag caagacctcg tctttgttta ggggaaaaaa aagaaattta 21000agtaggagat tatataagca aaaatacaat taatttccag cattcactat ataatataaa 21060tctccagact ttactttttt gtttactgga tataaacaat atctttttct gtctccagat 21120aattccccca ccacctccca tatgtccaga ttctcttgat gatgctgatg ctttgggaag 21180tatgttaatt tcatggtaca tgagtggcta tcatactggc tattatatgg taagtaatca 21240ctcagcatct tttcctgaca atttttttgt agttatgtga ctttgttttg taaatttata 21300aaatactact tgcttctctc tttatattac taaaaaataa aaataaaaaa atacaactgt 21360ctgaggctta aattactctt gcattgtccc taagtataat tttagttaat tttaaaaagc 21420tttcatgcta ttgttagatt attttgatta tacacttttg aattgaaatt atactttttc 21480taaataatgt tttaatctct gatttgaaat tgattgtagg gaatggaaaa gatgggataa 21540tttttcataa atgaaaaatg aaattctttt tttttttttt ttttttttga gacggagtct 21600tgctctgttg cccaggctgg agtgcaatgg cgtgatcttg gctcacagca agctctgcct 21660cctggattca cgccattctc ctgcctcagc ctcagaggta gctgggacta caggtgcctg 21720ccaccacgcc tgtctaattt tttgtatttt tttgtaaaga cagggtttca ctgtgttagc 21780caggatggtc tcaatctcct gaccccgtga tccacccgcc tcggccttcc aagagaaatg 21840aaattttttt aatgcacaaa gatctggggt aatgtgtacc acattgaacc ttggggagta 21900tggcttcaaa cttgtcactt tatacgttag tctcctacgg acatgttcta ttgtatttta 21960gtcagaacat ttaaaattat tttattttat tttatttttt tttttttttt gagacggagt 22020ctcgctctgt cacccaggct ggagtacagt ggcgcagtct cggctcactg caagctccgc 22080ctcccgggtt cacgccattc tcctgcctca gcctctccga gtagctggga ctacaggcgc 22140ccgccaccac gcccggctaa ttttttttta tttttagtag agacggggtt tcaccgtggt 22200ctcgatctcc tgacctcgtg atccacccgc ctcggcctcc caaagtgctg ggattacaag 22260cgtgagccac cgcgcccggc ctaaaattat ttttaaaagt aagctcttgt gccctgctaa 22320aattatgatg tgatattgta ggcacttgta tttttagtaa attaatatag aagaaacaac 22380tgacttaaag gtgtatgttt ttaaatgtat catctgtgtg tgcccccatt aatattctta 22440tttaaaagtt aaggccagac atggtggctt acaactgtaa tcccaacagt ttgtgaggcc 22500gaggcaggca gatcacttga ggtcaggagt ttgagaccag cctggccaac atgatgaaac 22560cttgtctcta ctaaaaatac caaaaaaaat ttagccaggc atggtggcac atgcctgtaa 22620tccgagctac ttgggaggct gtggcaggaa aattgcttta atctgggagg cagaggttgc 22680agtgagttga gattgtgcca ctgcactcca cccttggtga cagagtgaga ttccatctca 22740aaaaaagaaa aaggcctggc acggtggctc acacctataa tcccagtact ttgggaggta 22800gaggcaggtg gatcacttga ggttaggagt tcaggaccag cctggccaac atggtgacta 22860ctccatttct actaaataca caaaacttag cccagtggcg ggcagttgta atcccagcta 22920cttgagaggt tgaggcagga gaatcacttg aacctgggag gcagaggttg cagtgagccg 22980agatcacacc gctgcactct agcctggcca acagagtgag aatttgcgga gggaaaaaaa 23040agtcacgctt cagttgttgt agtataacct tggtatattg tatgtatcat gaattcctca 23100ttttaatgac caaaaagtaa taaatcaaca gcttgtaatt tgttttgaga tcagttatct 23160gactgtaaca ctgtaggctt ttgtgttttt taaattatga aatatttgaa aaaaatacat 23220aatgtatata taaagtattg gtataattta tgttctaaat aactttcttg agaaataatt 23280cacatggtgt gcagtttacc tttgaaagta tacaagttgg ctgggcacaa tggctcacgc 23340ctgtaatccc agcactttgg gaggccaggg caggtggatc acgaggtcag gagatcgaga 23400ccatcctggc taacatggtg aaaccccgtc tctactaaaa gtacaaaaac aaattagccg 23460ggcatgttgg cgggcacctt ttgtcccagc tgctcgggag gctgaggcag gagagtggcg 23520tgaacccagg aggtggagct tgcagtgagc cgagattgtg ccagtgcact ccagcctggg 23580cgacagagcg agactctgtc tcaaaaaata aaataaaaaa gaaagtatac aagtcagtgg 23640ttttggtttt cagttatgca accatcacta caatttaaga acattttcat caccccaaaa 23700agaaaccctg ttaccttcat tttccccagc cctaggcagt cagtacactt tctgtctcta 23760tgaatttgtc tattttagat attatatata aacggaatta tacgatatgt ggtcttttgt 23820gtctggcttc tttcacttag catgctattt tcaagattca tccatgctgt agaatgcacc 23880agtactgcat tccttcttat tgctgaatat tctgttgttt ggttatatca cattttatcc 23940attcatcagt tcatggacat ttaggttgtt tttatttttg ggctataatg aataatgttg 24000ctatgaacat tcgtttgtgt tctttttgtt tttttggttt tttgggtttt ttttgttttg 24060tttttgtttt tgagacagtc ttgctctgtc tcctaagctg gagtgcagtg gcatgatctt 24120ggcttactgc aagctctgcc tcccgggttc acaccattct cctgcctcag cccgacaagt 24180agctgggact acaggcgtgt gccaccatgc acggctaatt ttttgtattt ttagtagaga 24240tggggtttca ccgtgttagc caggatggtc tcgatctcct gacctcgtga tctgcctgcc 24300taggcctccc aaagtgctgg gattacaggc gtgagccact gcacctggcc ttaagtgttt 24360ttaatacgtc attgccttaa gctaacaatt cttaaccttt gttctactga agccacgtgg 24420ttgagatagg ctctgagtct agcttttaac ctctatcttt ttgtcttaga aatctaagca 24480gaatgcaaat gactaagaat aatgttgttg aaataacata aaataggtta taactttgat 24540actcattagt aacaaatctt tcaatacatc ttacggtctg ttaggtgtag attagtaatg 24600aagtgggaag ccactgcaag ctagtataca tgtagggaaa gatagaaagc attgaagcca 24660gaagagagac agaggacatt tgggctagat ctgacaagaa aaacaaatgt tttagtatta 24720atttttgact ttaaattttt tttttattta gtgaatactg gtgtttaatg gtctcatttt 24780aataagtatg acacaggtag tttaaggtca tatattttat ttgatgaaaa taaggtatag 24840gccgggcacg gtggctcaca cctgtaatcc cagcactttg ggaggccgag gcaggcggat 24900cacctgaggt cgggagttag agactagcct caacatggag aaaccccgtc tctactaaaa 24960aaaatacaaa attaggcggg cgtggtggtg catgcctgta atcccagcta ctcaggaggc 25020tgaggcagga gaattgcttg aacctgggag gtggaggttg cggtgagccg agatcacctc 25080attgcactcc agcctgggca acaagagcaa aactccatct caaaaaaaaa aaaataaggt 25140ataagcgggc tcaggaacat cattggacat actgaaagaa gaaaaatcag ctgggcgcag 25200tggctcacgc cggtaatccc aacactttgg gaggccaagg caggcgaatc acctgaagtc 25260gggagttcca gatcagcctg accaacatgg agaaaccctg tctctactaa aaatacaaaa 25320ctagccgggc atggtggcgc atgcctgtaa tcccagctac ttgggaggct gaggcaggag 25380aattgcttga accgagaagg cggaggttgc ggtgagccaa gattgcacca ttgcactcca 25440gcctgggcaa caagagcgaa actccgtctc aaaaaaaaaa ggaagaaaaa tattttttta 25500aattaattag tttatttatt ttttaagatg gagttttgcc ctgtcaccca ggctggggtg 25560caatggtgca atctcggctc actgcaacct ccgcctcctg ggttcaagtg attctcctgc 25620ctcagcttcc cgagtagctg tgattacagc catatgccac cacgcccagc cagttttgtg 25680ttttgttttg ttttttgttt tttttttttg agagggtgtc ttgctctgtc ccccaagctg 25740gagtgcagcg gcgcgatctt ggctcactgc aagctctgcc tcccaggttc acaccattct 25800cttgcctcag cctcccgagt agctgggact acaggtgccc gccaccacac ccggctaatt 25860tttttgtgtt tttagtagag atggggtttc actgtgttag ccaggatggt ctcgatctcc 25920tgaccttttg atccacccgc ctcagcctcc ccaagtgctg ggattatagg cgtgagccac 25980tgtgcccggc ctagtcttgt atttttagta gagtcgggat ttctccatgt tggtcaggct 26040gttctccaaa tccgacctca ggtgatccgc ccgccttggc ctccaaaagt gcaaggcaag 26100gcattacagg catgagccac tgtgaccggc aatgttttta aattttttac atttaaattt 26160tattttttag agaccaggtc tcactctatt gctcaggctg gagtgcaagg gcacattcac 26220agctcactgc agccttgacc tccagggctc aagcagtcct ctcacctcag tttcccgagt 26280agctgggact acagtgataa tgccactgca cctggctaat ttttattttt atttatttat 26340ttttttttga gacagagtct tgctctgtca cccaggctgg agtgcagtgg tgtaaatctc 26400agctcactgc agcctccgcc tcctgggttc aagtgattct cctgcctcaa cctcccaagt 26460agctgggatt agaggtcccc accaccatgc ctggctaatt ttttgtactt tcagtagaaa 26520cggggttttg ccatgttggc caggctgttc tcgaactcct gagctcaggt gatccaactg 26580tctcggcctc ccaaagtgct gggattacag gcgtgagcca ctgtgcctag cctgagccac 26640cacgccggcc taatttttaa attttttgta gagacagggt ctcattatgt tgcccagggt 26700ggtgtcaagc tccaggtctc aagtgatccc cctacctccg cctcccaaag ttgtgggatt 26760gtaggcatga gccactgcaa gaaaacctta actgcagcct aataattgtt ttctttggga 26820taacttttaa agtacattaa aagactatca acttaatttc tgatcatatt ttgttgaata 26880aaataagtaa aatgtcttgt gaaacaaaat gctttttaac atccatataa agctatctat 26940atatagctat ctatatctat atagctattt tttttaactt cctttatttt ccttacaggg 27000ttttagacaa aatcaaaaag aaggaaggtg ctcacattcc ttaaattaag gagtaagtct 27060gccagcatta tgaaagtgaa tcttactttt gtaaaacttt atggtttgtg gaaaacaaat 27120gtttttgaac atttaaaaag ttcagatgtt agaaagttga aaggttaatg taaaacaatc 27180aatattaaag aattttgatg ccaaaactat tagataaaag gttaatctac atccctacta 27240gaattctcat acttaactgg ttggttgtgt ggaagaaaca tactttcaca ataaagagct 27300ttaggatatg atgccatttt atatcactag taggcagacc agcagacttt tttttattgt 27360gatatgggat aacctaggca tactgcactg tacactctga catatgaagt gctctagtca 27420agtttaactg gtgtccacag aggacatggt ttaactggaa ttcgtcaagc ctctggttct 27480aatttctcat ttgcaggaaa tgctggcata gagcagcact aaatgacacc actaaagaaa 27540cgatcagaca gatctggaat gtgaagcgtt atagaagata

actggcctca tttcttcaaa 27600atatcaagtg ttgggaaaga aaaaaggaag tggaatgggt aactcttctt gattaaaagt 27660tatgtaataa ccaaatgcaa tgtgaaatat tttactggac tctattttga aaaaccatct 27720gtaaaagact gaggtggggg tgggaggcca gcacggtggt gaggcagttg agaaaatttg 27780aatgtggatt agattttgaa tgatattgga taattattgg taattttatg agctgtgaga 27840agggtgttgt agtttataaa agactgtctt aatttgcata cttaagcatt taggaatgaa 27900gtgttagagt gtcttaaaat gtttcaaatg gtttaacaaa atgtatgtga ggcgtatgtg 27960gcaaaatgtt acagaatcta actggtggac atggctgttc attgtactgt ttttttctat 28020cttctatatg tttaaaagta tataataaaa atatttaatt tttttttaaa 280702618DNAArtificial sequenceSynthetic oligonucleotide 26actttcataa tgctggca 182718DNAArtificial sequenceSynthetic oligonucleotide 27cactttcata atgctggc 18

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed