Language selection

Search

Patent 2842187 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2842187
(54) English Title: HETEROCYCLIC COMPOUNDS AND USES THEREOF
(54) French Title: COMPOSES HETEROCYCLIQUES ET LEURS UTILISATIONS
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 471/04 (2006.01)
  • A61K 31/4725 (2006.01)
  • A61P 37/00 (2006.01)
  • C07D 487/04 (2006.01)
(72) Inventors :
  • CASTRO, ALFREDO C. (United States of America)
  • EVANS, CATHERINE A. (United States of America)
  • JANARDANANNAIR, SOMARAJANNAIR (United States of America)
  • LESCARBEAU, ANDRE (United States of America)
  • LIU, TAO (United States of America)
  • SNYDER, DANIEL A. (United States of America)
  • TREMBLAY, MARTIN R. (United States of America)
  • REN, PINGDA (United States of America)
  • LIU, YI (United States of America)
  • LI, LIANSHENG (United States of America)
  • CHAN, KATRINA (United States of America)
(73) Owners :
  • INFINITY PHARMACEUTICALS INC. (United States of America)
(71) Applicants :
  • INFINITY PHARMACEUTICALS INC. (United States of America)
(74) Agent: OSLER, HOSKIN & HARCOURT LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2012-07-18
(87) Open to Public Inspection: 2013-01-24
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2012/047186
(87) International Publication Number: WO2013/012915
(85) National Entry: 2014-01-16

(30) Application Priority Data:
Application No. Country/Territory Date
61/509,458 United States of America 2011-07-19
61/509,474 United States of America 2011-07-19
61/509,409 United States of America 2011-07-19

Abstracts

English Abstract

Compounds and pharmaceutical compositions that modulate kinase activity, including PI3 kinase activity, and compounds, pharmaceutical compositions, and methods of treatment of diseases and conditions associated with kinase activity, including PI3 kinase activity, are described herein.


French Abstract

L'invention concerne des composés et des compositions pharmaceutiques qui modulent l'activité de kinase, notamment l'activité de kinase PI3, et des composés, des compositions pharmaceutiques et des procédés de traitement de maladies et d'affections associées à l'activité de kinase, notamment l'activité de kinase PI3.

Claims

Note: Claims are shown in the official language in which they were submitted.




CLAIMS
WHAT IS CLAIMED IS:

1. A compound of Formula (I):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
W b5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or heteroaryl, each of
which is substituted with 0-4 occurrences of R2;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl,
nitro, phosphate, urea or carbonate;
X is absent or is -(CH(R9)),-;
Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R9)-, -C(=O)-(CHR9),-, -C(=O)-, -
N(R9)-C(=O)-, or -N(R9)-
C(=O)NH-, -N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when W b5 is N, no more than one of X or Y is absent;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl,
acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro; wherein
each of the above substituents can be substituted with 0, 1, 2, or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
W d is Image
282



wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and R17 are independently hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, oxo, or NR'R" wherein
R' and R" are taken together with nitrogen to form a cyclic moiety.
2. The compound of claim 1, wherein Cy is aryl substituted with 0-1
occurrences of R3 and 0-3 occurrences of R5.
3. The compound of claim 2, wherein Cy is phenyl substituted with 1
occurrence of R3 and 0 occurrences of R5.
4. The compound of claim 3, wherein the compound of Formula (I) has a
structure of Formula (II):
Image
5. The compound of claim 4, wherein the compound of Formula (II) has a
structure of Formula (IIa) or (IIb):
Image
6. The compound of claim 5, wherein the compound of Formula (II) has a
structure of Formula (IIIa) or (IIIb):
283



Image
7. The compound of claim 6, wherein the compound of Formula (II) has a
structure of Formula (IIIb-1):
Image
8. The compound of claim 1, wherein Cy is heteroaryl substituted with 0-1
occurrences of le and 0-3 occurrences
of R5.
9. The compound of claim 8, wherein Cy is 5-membered heteroaryl substituted
with 0-1 occurrences of R3 and 0-3
occurrences of R5.
10. The compound of claim 9, wherein Cy is isothiazolyl substituted with 0-
1 occurrences of le and 0 occurrence
of R5.
11. The compound of claim 10, wherein the compound of Formula (I) has the
structure of Formula (IVa) or
Formula (IVb):
284



Image
12. The compound of claim 11, wherein the compound of Formula (IVa) or
Formula (IVb) has the structure of
Formula (Va) or Formula (Vb):
Image
13. The compound of claim 12, wherein the compound of Formula (I) has a
structure of Formula (Va-1):
Image
14. The compound of claim 9, wherein Cy is thiophenyl substituted with 0-1
occurrences of R3 and 0 occurrence of
R5.
15. The compound of claim 14, wherein the compound of Formula (I) has the
structure of Formula (VIa) or
Formula (VIb):
Image
16. The compound of claim 15, wherein the compound of Formula (VIa) or
Formula (VIb) has the structure of
Formula (VIIa) or (VIIb):
285


Image
17. The compound of claim 15, wherein the compound of Formula (VIa) or
Formula (VIb) has the structure of
Formula (VIIIa) or Formula (VIIIb):
Image
18. The compound of claim 17, wherein the compound of Formula (VIIa) or
Formula (VIIb) has the structure of
Formula (IXa) or Formula (IXb):
Image
19. The compound of claim 18, wherein the compound of Formula (I) has a
structure of Formula (IXa-1):
Image
20. The compound of any one of claims 1-19, wherein each R5 is
independently selected from CH3, OCH3, CF3,
and halo.
21. The compound of any one of claims 1-20, wherein R3 is selected from
alkyl, cycloalkyl, halo, aryl, and
heteroaryl.
22. The compound of any one of claims 1-21, wherein R3 is selected from
methyl, chloro, and pyrazolo.
286




Image
288


Image
289

Image
or a pharmaceutically acceptable form thereof.
40. A compound of Formula (XI):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein:

290


W b5 is N, CHR8, or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is -(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)2-, -C(=O)-, or -O-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen,or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, heteroaryl, aryl, hydroxyl, or nitro;
wherein each of the above substituents can be
substituted with 0, 1, 2, or 3 R17;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R"
are taken together with nitrogen to
form a cyclic moiety;
X is absent or is -(CH(R16))z-;
Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R16)-, -C(=O)-(CHR16)-, -C(=O)-, -
N(R16)-C(=O)-, or -N(R16)-
C(=O)NH-,-N(R16)C(R16)2-, or -C(=O)-N(R16)-(CHR16)z-;
each z is an integer of 1, 2, 3, or 4;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or heteroaryl; and
W d is Image
wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and R17 are independently hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, oxo, or NR'R" wherein
R' and R" are taken together with nitrogen to form a cyclic moiety.
4 1. The compound of claim 40, wherein Cy is a 6-membered ring.
42. The compound of claim 40 or claim 4 1, wherein Cy is a 6-membered aryl
ring.
43. The compound of any one of claims 40-42, wherein W b5 is CH.
44. The compound of any one of claims 40-43, wherein X-Y is -CH2-N(CH3).
45. A compound of Formula (XII):
291



Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
W b5 is CR8; CHR8, or N,
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl, aryl or
heteroaryl, each of which is
substituted with 0-4 R2;
R2 is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano,
hydroxyl, nitro, phosphate, urea or
carbonate;
X is absent or is -(CH(R9))z-;
Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R9)-, -C(=O)-(CHR9),-, -C(=O)-, -
N(R9)-C(=O)-, -N(R9)-
C(=O)NH-,-N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido, amino, acyl,
acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido, halo, cyano,
aryl, heteroaryl, hydroxyl, or nitro; wherein
each of the above substituents can be substituted with 0, 1, 2, or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
Wd is Image
wherein one of X5 and X2 is N and one of X5 and X2 is C;
X3 and X4 are each independently CR13 or N;
X2 and X3 are not both N; and
each R10, R11, R12, R13, and R17 is independently hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
292


alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, oxo, or NR'R" wherein R' and
R" are taken together with nitrogen to form a cyclic moiety.
46. The compound of claim 45, wherein Cy is aryl substituted with 0-1
occurrences of R3 and 0-3 occurrences of
R5.
47. The compound of claim 46, wherein Cy is phenyl substituted with 1
occurrence of R3 and 0 occurrences of R5.
48. The compound of claim 47, wherein the compound of Formula (XII) has a
structure of Formula (XIII):
Image
49. The compound of claim 48, wherein the compound of Formula (XIII) has a
structure of Formula (XIIIa) or
(XIIIb):
Image
50. The compound of claim 49, wherein the compound of Formula (XIIIa) or
(XIIIb) has a structure of Formula
(XIVa) or (XIVb):
293


Image
51. The compound of claim 50, wherein the compound of Formula (XIII) has a
structure of Formula (XIVb-1):
Image
52. The compound of claim 45, wherein Cy is heteroaryl substituted with 0-1
occurrences of R3 and 0-3
occurrences of R5.
53. The compound of claim 52, wherein Cy is 5-membered heteroaryl
substituted with 0-1 occurrences of R3 and 0-
3 occurrences of R5.
54. The compound of claim 53, wherein Cy is isothiazolyl substituted with 0-
1 occurrences of R3 and 0 occurrence
of R5.
55. The compound of claim 53, wherein the compound of Formula (XII) has a
structure of Formula (XVa) or
(XVb):
Image
294


Formula (XVa) Formula (XVb).
56. The compound of claim 55, wherein the compound of Formula (XVa) or
(XVb) has a structure of Formula
(XVIa) or (XVIb):
Image
57. The compound of claim 56, wherein the compound of Formula (XII) has a
structure of Formula (XVIa-1):
Image
58. The compound of claim 53, wherein Cy is thiophenyl substituted with 0-1
occurrences of R3 and 0 occurrence
of R5.
59. The compound of claim 58, wherein the compound of Formula (XII) has a
structure of Formula (XVIIa) or
(XVIIb):
Image
60. The compound of claim 59, wherein the compound of Formula (XVIIa) or
(XVIIb) has a structure of Formula
(XVIIIa) or (XVIIIb):
295


Image
61. The compound of claim 59, wherein the compound of Formula (XVIIa) or
(XVIIb) has a structure of Formula
(XIXa) or (XIXb):
Image
62. The compound of claim 61, wherein the compound of Formula (XVIIa) or
(XVIIb) has the structure of Formula
(XXa) or (XXb):
Image
63. The compound of claim 62, wherein the compound of Formula (XII) has a
structure of Formula (XXa-1):
Image
64. The compound of any one of claims 45-63, wherein each R5 is
independently selected from CH3, OCH3, CF3,
and halo.
65. The compound of any one of claims 45-64, wherein R3 is selected from
hydrogen, alkyl, cycloalkyl, halo, aryl,
and heteroaryl.
296


66. The compound of any one of claims 45-65, wherein R3 is selected from
methyl, chloro, and pyrazolo.
67. The compound of any one of claims 45-66, wherein B is aryl substituted
with 0-3 occurrences of R2.
68. The compound of any one of claims 45-67, wherein B is phenyl
substituted with 0-3 occurrences of R2.
69. The compound of any one of claims 45-68, wherein B is unsubstituted
phenyl.
70. The compound of any one of claims 45-68, wherein B is phenyl
substituted with 1 occurrence of R2.
71. The compound of any one of claims 45-70, wherein R2 is halo or alkyl.
72. The compound of any one of claims 45-66, wherein B is cycloalkyl.
73. The compound of any one of claims 45-66, wherein B is a heterocyclyl.
74. The compound of any one of claims 45-73, wherein W d is Image
75. The compound of any one of claims 45-73, wherein Wd is Image
76. The compound of any one of claims 45-73, wherein Wd is Image
77. The compound of any one of claims 45-76, wherein Y is absent, -O-, -
NH(R9)-, or -S(=O)2-.
78. The compound of any one of claims 45-77, wherein X-Y is -CH2-N(CH3).
79. The compound of any one of claims 45-77, wherein X-Y is (S)-CH(CH3)-NH-
.
80. The compound of any one of claims 45-77, wherein X-Y is (R)-CH(CH3)-NH-
.
81. The compound of any one of claims 45-80, wherein R8 is hydrogen.
82. The compound of claim 45, wherein the compound is
297


Image
298



Image
or a pharmaceutically acceptable form thereof.
83. A compound of Formula (XXIII):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein
W b5 is N or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is -(L)-R1';
L is a bond, -S-, -N(R16)-, -C(R15)2-, -C(=O)-, or -O-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, heterocyclylalkyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido,
halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, substituted
nitrogen,or NR'R" wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido, amino, acyl,
acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro; wherein
each of the above substituents can be substituted with 0, 1, 2, or 3 R17;
each R5is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido,
halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R'
and R" are taken together with nitrogen
to form a cyclic moiety;
X is absent or is -(CH(R14))z;
299


Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R9)-, -C(=O)-(CHR14),-, -C(=O)-, -
N(R9)-C(=O)-, or -N(R9)-
C(=O)NH-,-N(R9)C(R14)2-, or C(=O)-(CHR14)z-;
each z is an integer of 1,2,3, or 4;
each R9 and R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, or heteroaryl;
each R14 and R15 is independently hydrogen, alkyl, aryl, heteroaryl or halo;
and
W d is Image
wherein R10, R11, R12, and R17 are independently hydrogen, alkyl, heteroalkyl,
alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocycloalkyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea,
carbonate, oxo, or NR'R" wherein R' and R"
are taken together with nitrogen to form a cyclic moiety.
84. The compound of claim 83, wherein the compound of of Formula (XXIII)
has a structure of Formula (XXIV):
Image
85. The compound of claim 83 or claim 84, wherein R3 is halo, alkyl,
heteroalkyl, haloalkyl, alkenyl, alkynyl,
cycloalkyl, heteroaryl, or heterocyclyl.
86. The compound of any one of claims 83-85, wherein R3 is alkyl or halo.
87. The compound of any one of claims 83-86, wherein L is -C(=O)- or -O-.
88. The compound of any one of claims 83-87, wherein R1' is substituted
alkyl, substituted nitrogen, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety.
89. The compound of claim 88, wherein the cyclic moiety is a morpholino
group.
90. The compound of any one of claims 83-89, wherein X is -CH2- or -CH(CH3)-
.
91. The compound of any one of claims 83-90, wherein the CH carbon of the -
CH(CH3)- moiety has a (S)
stereochemical configuration.
92. The compound of any one of claims 83-90, wherein the CH carbon of the -
CH(CH3)- moiety has a (R)
stereochemical configuration.
93. The compound of any one of claims 83-92, wherein Y is absent, -O-, -
NH(R9)-, or -S(=O)2-.
94. The compound of any one of claims 83-93, wherein R9 is methyl or
hydrogen.
300


95. The compound of any one of claims 83-94, wherein X-Y is -CH2-N(CH3).
96. The compound of any one of claims 83-94, wherein X-Y is (S) -CH(CH3)-NH-
.
97. The compound of any one of claims 83-94, wherein X-Y is (R) -CH(CH3)-NH-
.
98. The compound of any one of claims 83-97, wherein R10, R11, and R12 are
independently selected from
hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo,
cyano, and hydroxyl.
99. The compound of any one of claims 83-98, wherein R10, R11, and R12 are
independently selected hydrogen,
amino, and chloro.
100. The compound of any one of claims 83-99, wherein R10 is amino or
chloro.
101. A compound of Formula (XXII):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein:
W b5 is N, CHR8 or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is -(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)2-, -C(=O)-, or -O-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl, heterocyclyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido,
amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate,
substituted nitrogen,or NR'R" wherein R' and R"
are taken together with nitrogen to form a cyclic moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido, amino, acyl,
acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro; wherein
each of the above substituents can be substituted with 0,1,2, or 3 R17;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo,

301


cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R"
are taken together with nitrogen to form
a cyclic moiety;
X is absent or is -(CH(R9))z;
Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R9)-, -C(=O)-(CHR9)z-, -C(=O)-, -
N(R9)-C(=O)-, or -N(R9)-
C(=O)NH-,-N(R9)C(R9)2-, or -C(=O)-N(R9)-(CHR9)z-;
each z is an integer of 1,2,3, or 4;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or heteroaryl; and
W d is Image
wherein one of X5 and X2 is N and one of X5 and X2 is C;
wherein X3 and X4 are each independently selected from CR13 and N; and
each R10, R11, R12, R13, and R17 is independently hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, or NR'R" wherein R' and R" are
taken together with nitrogen to form a cyclic moiety.
102. The compound of claim 101, wherein Cy is a 6-membered ring.
103. The compound of claim101 or claim 102, wherein Cy is a 6-membered aryl
ring.
104. The compound of any one of claims 101-103, wherein W b5 is CH.
105. The compound of any one of claims 101-104, wherein W d is Image
106. The compound of any one of claims 101-104, wherein W d is Image
107. The compound of any one of claims 101-104, wherein W d is Image

302


108. A compound of Formula (XXV):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its pharmaceutically
acceptable forms thereof, wherein
W b1 and W b2 are each independently CR3, S, O, N or NR13, wherein at least
one of W b1 and W b2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
W b5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
X is absent or is -(CH(R9))z-;
Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R9)-, -C(=O)-(CHR9)z-, -C(=O)-, -
N(R9)-C(=O)-, or -
N(R9)-C(=O)NH-,-N(R9)C(R9)2-, or -C(=O)-N(R9)-(CHR9)z;
each z is independently an integer of 1, 2, 3, or 4;
wherein when W b5 is N, no more than one of X or Y is absent;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl,
nitro, phosphate, urea or carbonate;
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
W d is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more R b,
R11 or R12;
wherein each R b is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl, alkynyl,
cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate, oxo,
or NR'R", wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.

303


109. The compound of claim 0, wherein W b1 and W b2 are CR3.
110. The compound of claim 0, wherein W b1 is S and W b2 is CR3.
111. The compound of claim 0, wherein W b1 is S and W b2 is N.
112. The compound of claim 0, wherein the compound of Formula (XXV) has a
structure of Formula
(XXVI):
Image
113. The compound of claim 112, wherein the compound of Formula (XXVI) has
a structure of Formula (XXVIa) or
(XXVIb):
Image
114. The compound of claim 113, wherein the compound of Formula (XXVI) has
a structure of Formula (XXVIIa)
or (XXVIIb):
Image
115. The compound of claim 0, wherein the compound of Formula (XXV) has a
structure of Formula (XXVIII):

304


Image
116. The compound of claim 115, wherein the compound of Formula (XXVIII)
has a structure of Formula (XXIX):
Image
117. The compound of claim 0, wherein the compound of Formula (XXV) has a
structure of Formula (XXX):
Image
118. The compound of claim 117, wherein the compound Formula (XXX) has a
structure of Formula (XXXI):
Image
119. The compound of claim 118, wherein the compound of Formula (XXX) has a
structure of Formula (XXXII):

305


Image
120. The compound of claim 119, wherein the compound of Formula (XXXII) has
a structure of Formula (XXXIII):
Image
121. The compound of any one of claims 0-120, wherein each R3 is
independently hydrogen, alkyl, cycloalkyl,
heterocyclyl, fluoroalkyl, alkoxy, halo, cyano, heteroaryl or aryl.
122. The compound of any one of claims 0-121, wherein R3 is hydrogen,
alkyl, fluoroalkyl, alkoxy or aryl.
123. The compound of any one of claims 0-122, wherein p is 0 or 1.
124. The compound of any one of claims 0-123, wherein B is aryl substituted
with 0-4 occurrences of R2.
125. The compound of any one of claims 0-124, wherein B is phenyl
substituted with 0-4 occurrences or R2.
126. The compound of any one of claims 0-125, wherein X is -(CH(R9))z-.
127. The compound of any one of claims 0-126, wherein z is 1.
128. The compound of any one of claims 0-127, wherein Y is absent or -N(R9)-
.
129. The compound of any one of claims 0-128, wherein W d is heteroaryl.
130. The compound of any one of claims 0-129, wherein W d is selected from:
Image

306


Image
131. The compound of any one of claims 0-130, wherein R12 is hydrogen,
alkyl,haloalkyl, halo, cyano, amino or
amido.
132. The compound of any one of claims 0-131, wherein R12 is hydrogen.
133. The compound of any one of claims 0-132, wherein R11 is hydrogen or
amino.
134. The compound of any one of claims 0-133, wherein R b is hydrogen or
amino.
135. The compound of claim 0, wherein the compound is

307


Image

308


Image
or a pharmaceutically acceptable form thereof.
136. A compound of Formula (XXXIV) or (XXXV):
Image
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or their pharmaceutically
acceptable forms thereof, wherein:
W b1 and W b2 are each independently CR3, S, O, N or NR13, wherein at least
one of W b1 and W b2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
W b5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl, acyloxy,
sulfonamido, halo, cyano, hydroxyl, or nitro;
R1 is -(L)-R1';
L is a bond, -S-, -N(R15)-, -C(=O)-, or -O-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
X is absent or is -(CH(R16))z;

309


Y is absent, -O-, -S-, -S(=O)-, -S(=O)2-, -N(R16)-, -C(=O)-(CHR16),-, -C(=O)-,
-N(R16)-C(=O)-, or -
N(R16)-C(=O)NH-,-N(R16)C(R16)2-, or -C(=O)-N(R16)-(CHR16)z-;
each z is an integer of 1,2,3, or 4;
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R13 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl;
W d is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more R b,
R11 or R12;
wherein each R b is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl, alkynyl,
cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate, oxo,
or NR'R", wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.
137. The compound of claim 136, wherein W b1 is CR3 and W b2 is CR3.
138. The compound of claim 136 or claim 137, wherein W b5 is CH.
139. The compound of any one of claims 136-138, wherein L is a bond, -
N(R15)- or -C(=O)-.
140. A pharmaceutical composition comprising a compound according to any
one of claims 1-139 and one or more
pharmaceutically acceptable excipients.
141. A method of treating a PI3K mediated disorder in a subject, the method
comprising administering a
therapeutically effective amount of a compound according to any one of claims
1-139 or a pharmaceutical composition
according to claim 140 to said subject.
142. The method of claim 141, wherein the disorder is cancer, an
inflammatory disease or an auto-immune disease.

310

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
HETEROCYCLIC COMPOUNDS AND USES THEREOF
[0001]
This application claims priority to U.S. Provisional Application Nos.
61/509,409, filed July 19,
2011, 61/509,458, filed July 19, 2011, 61/509,474, filed July 19, 2011, and
61/562,247, filed November 21, 2011,
the entireties of which are incorporated herein by reference.
BACKGROUND
[0002]
The activity of cells can be regulated by external signals that stimulate or
inhibit intracellular
events. The process by which stimulatory or inhibitory signals are transmitted
into and within a cell to elicit an
intracellular response is referred to as signal transduction. Over the past
decades, cascades of signal transduction
events have been elucidated and found to play a central role in a variety of
biological responses. Defects in various
components of signal transduction pathways have been found to account for a
vast number of diseases, including
numerous forms of cancer, inflammatory disorders, metabolic disorders,
vascular and neuronal diseases (Gaestel et
al. Current Medicinal Chemistry (2007) 14:2214-2234).
[0003]
Kinases represent a class of important signaling molecules. Kinases can
generally be
classified into protein kinases and lipid kinases, and certain kinases exhibit
dual specificities. Protein kinases are
enzymes that phosphorylate other proteins and/or themselves (i.e.,
autophosphorylation). Protein kinases can be
generally classified into three major groups based upon their substrate
utilization: tyrosine kinases which
predominantly phosphorylate substrates on tyrosine residues (e.g., erb2, PDGF
receptor, EGF receptor, VEGF
receptor, src, abl), serine/threonine kinases which predominantly
phosphorylate substrates on serine and/or threonine
residues (e.g., mTorCl, mTorC2, ATM, ATR, DNA-PK, Akt), and dual-specificity
kinases which phosphorylate
substrates on tyrosine, serine and/or tlu-eonine residues.
[0004]
Lipid kinases are enzymes that catalyze the phosphorylation of lipids. These
enzymes, and the
resulting phosphorylated lipids and lipid-derived biologically active organic
molecules play a role in many different
physiological processes, including cell proliferation, migration, adhesion,
and differentiation. Certain lipid kinases
are membrane associated and they catalyze the phosphorylation of lipids
contained in or associated with cell
membranes. Examples of such enzymes include phosphoinositide(s) kinases (e.g.,
P13-kinases, P14-Kinases),
diacylglycerol kinases, and sphingosine kinases.
[0005]
The phosphoinositide 3-kinases (PI3Ks) signaling pathway is one of the most
highly mutated
systems in human cancers. PI3K signaling is also a key factor in many other
diseases in humans. PI3K signaling is
involved in many disease states including allergic contact dermatitis,
rheumatoid arthritis, osteoarthritis,
inflammatory bowel diseases, chronic obstructive pulmonary disorder,
psoriasis, multiple sclerosis, asthma,
disorders related to diabetic complications, and inflammatory complications of
the cardiovascular system such as
acute coronary syndrome.
[0006]
PI3Ks are members of a unique and conserved family of intracellular lipid
kinases that
phosphorylate the 3'-OH group on phosphatidylinositols or phosphoinositides.
The PI3K family comprises 15
kinases with distinct substrate specificities, expression patterns, and modes
of regulation. The class I PI3Ks (p110a,
p11013, p1106, and p1107) are typically activated by tyrosine kinases or G-
protein coupled receptors to generate

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
PIP3, which engages downstream effectors such as those in the Akt/PDK1
pathway, mTOR, the Tec family kinases,
and the Rho family GTPases. The class II and III PI3Ks play a key role in
intracellular trafficking through the
synthesis of PI(3)P and P1(3 ,4)P2. The P13 Ks are protein kinases that
control cell growth (mTORC1) or monitor
genomic integrity (ATM, ATR, DNA-PK, and hSmg-1).
[0007] The delta ((5) isoform of class I PI3K has been implicated, in
particular, in a number of
diseases and biological processes. PI3K-6 is expressed primarily in
hematopoietic cells including leukocytes such as
T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages.
PI3K-6 is integrally involved in
mammalian immune system functions such as T-cell function, B-cell activation,
mast cell activation, dendritic cell
function, and neutrophil activity. Due to its integral role in immune system
function, PI3K-6 is also involved in a
number of diseases related to undesirable immune response such as allergic
reactions, inflammatory diseases,
inflammation mediated angiogenesis, rheumatoid arthritis, and auto-immune
diseases such as lupus, asthma,
emphysema and other respiratory diseases. Other class I PI3K involved in
immune system function includes PI3K-
y, which plays a role in leukocyte signaling and has been implicated in
inflammation, rheumatoid arthritis, and
autoimmune diseases such as lupus.
[0008] Unlike PI3K-6, the beta (f3) isoform of class I PI3K appears to
be ubiquitously expressed.
PI3K-f3 has been implicated primarily in various types of cancer including
PTEN-negative cancer (Edgar et al.
Cancer Research (2010) 70(3):1164-1172), and HER2-overexpressing cancer such
as breast cancer and ovarian
cancer.
SUMMARY
[0009] Described herein are compounds capable of selectively inhibiting
certain isoform(s) of class I
PI3K without substantially affecting the activity of the remaining isoforms of
the same class. For example, non-
limiting examples of inhibitors capable of selectively inhibiting PI3K-6
and/or PI3K-y, but without substantially
affecting the activity of PI3K-f3 are disclosed. Such inhibitors can be
effective in ameliorating disease conditions
associated with PI3K-6/y activity.
[0010] In one aspect, provided herein are compounds are provided of
Formula (I):
0
=
W 5 X
Wd
Formula (I)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
2

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
Wb5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea or carbonate;
X is absent or is -(CH(R9)),-;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-, -N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when Wb5 is N, no more than one of X or Y is absent;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino,
acyl, acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
R13 Nj
Xi
I
R12N R1
Wd iS R11 /
wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and R17
are independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, carboxylic
acid, oxo, or NR'R" wherein R' and R" are taken together with nitrogen to form
a cyclic moiety.
[00 1 1] In some embodiments, the compound of Formula (I) has the
structure of Formula (II):
R3 0
---
x
1
88 Y
Wd
Formula (II) .
3

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[0012] For example, the compound of Formula (II) can have a structure of
Formula (Ha) or (MI):
R3 0
0 B
N
1
x
1
R8 Y
Wd
Formula (Ha) or
R3 0
NB
le 1
x
1
R8 Y
Wd
Formula (Hb)
[0013] In other embodiments, the compound of Formula (II) has the
structure of Formula (IIIa) or
(11th):
R3 0
1. R8 N13
1 R9
_
7
Wd ,
Formula (IIIa) or
R3 0
N13
le 1 R9
R8 7
Wd ,
Formula (Mb).
[0014] For example, the compound of Formula (I) has the structure of
Formula (IVa) or Formula
(IVb):
4

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
3
NB
N\ ,1
S wb5 Sw105-
Wd or Wd
Formula (IVa) Formula (IVb).
[0015]
In some embodiments, the compound of Formula (IVa) or Formula (IVb) has the
structure of
Formula (Va) or Formula (Vb):
R3 0 0
N'{'
\S R9 S R9
Wb
Wd or Wd
Formula (Va) Formula (Vb).
[0016]
In other embodiments, the compound of Formula (I) has the structure of Formula
(VIa) or
Formula (VIb):
,B
N-B
eL
S"N
Wb5 X Wb5 X
Y,
Wd or Wd
Formula (VIa) Formula (VIb).
[0017]
In other embodiments, the compound of Formula (VIa) or (VIb) has the structure
of Formula
(VIIa) or (VIIb):
0 0
R3\ ji
N,B
j\R9 ,õ, S"\b\,õ...R9
wb5
"
\-(\
Wd or Wd
Formula (VIIa) Formula (VIIb).
[0018] In some embodiments, the compound of Formula (VIIa) or (VIIb) has
the structure of Formula
(Villa) or (VIIIb):

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
R3\ it
B
N
CL_ 1
S'X S'X
1 1
Y, Y,
Wd or Wd
Formula (Villa) Formula (VIIIb).
[0019] In some embodiments, the compound of Formula (Vila) or (VIIb) has
the structure of Formula
(IXa) or (IXb):
0
R3\ ji 0
,B
(....FN
e N_L,B
R9 S- N,õ..R9
Y, Y,
Wd or Wd
Formula (IXa) Formula (IXb).
[0020] In another aspect, provided herein are compounds of Formula (X)
or (XI):
ii
0
Ni N
0 1
1
--j= --j
W/)( Wb5 )(1
I I
Y Y
Wd or \Aid
Formula (X) Formula (XI),
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or their
pharmaceutically acceptable forms thereof, wherein:
Wb5 is N, CHR8, or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is -(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)2-, -C(=0)-, or -0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
6

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfonamido, halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above
substituents can be substituted with 0, 1, 2, or 3 R17;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea, carbonate, or
NR'R" wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is ¨(CH(R16)),¨;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R16)-, -C(=0)-(CHR16),-, -C(=0)-,
-N(R16)-C(=0)-, or
-N(R16)-C(=0)NH-,-N(R16)C(R16)2-, or -C(=0)-N(R16)-(CHR16)z-;
each z is an integer of 1,2, 3, or 4;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl; and
..111W
Ri N j x
I 1
R12N R1
Wd iS R11 /
wherein X1 is N or CR14;
wherein R10, k' 1, R12, R13, R14, and R17
are independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, carboxylic
acid, oxo, or NR'R" wherein R' and R" are taken together with nitrogen to form
a cyclic moiety.
[0021] In a first aspect, provided herein are compounds of Formula
(XII):
0
B
0 N
-
Wb5 X
I
Y
Wd
Formula (XII)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
Wb5 is Cle, CHR8, or N,
7

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl, aryl or
heteroaryl, each of
which is substituted with 0-4 R2;
R2 is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea or carbonate;
X is absent or is ¨(CH(R9)),-;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, aryl, heteroaryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino,
acyl, acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
JUNA,
Y
. X X
Rio..... 0
y50 ..-4
i
, .2
N R12
Wd iS R11 ,
wherein one of X5 and X2 is N and one of X5 and X2 is C;
X3 and X4 are each independently CR13 or N;
X2 and X3 are not both N; and
each R10, R11, R12, R13, and R17 is independently hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, oxo, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety.
[0022] In
some embodiments, the compound of Formula (XII) has the structure of Formula
(XIII):
R3 0
140
1
R8 Y
Wd
Formula (XIII) .
8

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[0023] For example, the compound of Formula (XIII) can have a structure
of Formula (XIIIa) or
(XIIIb):
R3 0
0 B
N
1
x
1
R8 Y
Wd
Formula (XIIIa) or
R3 0
0 NB
1
x
1
R8 Y
Wd
Formula (XIIIb)
[0024] In other embodiments, the compound of Formula (XIIIa) or (XIIIb)
has the structure of
Formula (XIVa) or (XIVb):
R3 0
N13
1. 1 R9
R8 7
Wd ,
Formula (XIVa) or
R3 0
N13
le 1
R9
R8 7\
Wd ,
Formula (XIVb).
[0025] For example, the compound of Formula (XII) has the structure of
Formula (XVa) or Formula
(XVb):
9

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
3
,B ,B
N I
S wicnx
Wb X
Wd or Wd
Formula (XVa) Formula (XVb).
[0026] In some embodiments, the compound of Formula (XVa) or (XVb) has
the structure of Formula
(XVIa) or Formula (XVIb):
R3 0 0
,B
N
\S R9
Wb Wb
Wd or Wd
Formula (XVIa) Formula (XVIb).
[0027] In other embodiments, the compound of Formula (XII) has the
structure of Formula (XVIIa) or
Formula (XVIIb):
,B
N-B
CL
S"N S'N
Wb5 X Wb5 X
Y,
Wd or Wd
Formula (XVIIa) Formula (XVIIIb).
[0028] In other embodiments, the compound of Formula (XVIIa) or (XVIIb)
has the structure of
Formula (XVIIIa) or (XVIIIb):
0 0
R3\ ji ,B
N,B
CL
'j\R9 v v b5j\, wb5 R9
Wd or Wd
Formula (XVIIIa) Formula (XVIIIb).
[0029] In some embodiments, the compound of Formula (XVIIIa) or (XVIIIb)
has the structure of
Formula (XIXa) or (XIXb):

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
R3\
N
er_ 1 N
eL_ 1
S'X S'X
1 1
Y, Y,
Wd or Wd
Formula (XIXa) Formula (XIXb).
[0030] In some embodiments, the compound of Formula (XVIIIa) or (XVIIIb)
has the structure of
Formula (XXa) or (XXl3):
0
R3\ ji 0
,B
(....FN
e N_L,B
R9 S- R9
Y, Y,
Wd or Wd
Formula (XXa) Formula (XXl3).
[0031] In another aspect, provided herein is a compound of formula (XXI)
or (XXII):
ii
0
Ni N
0 1
1
--j= --j
W/)( Wb5 )(1
I I
Y Y
Wd or \Aid
Formula (XXI) Formula (XXII),
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein:
Wb5 is N, CHR8 or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is -(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)27, -C(=0)-, or -0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
11

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, oxo, or
NR'R" wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is ¨(CH(R9)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is an integer of 1,2, 3, or 4;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl; and
JINV
XLI.XX
Rb0KQ5O 3
N R-
Wd iS R11
wherein one of X5 and X2 is N and one of X5 and X2 is C;
wherein X3 and X4 are each independently selected from CR13 and N; and
each R10, R11, R12 an R13
is independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, or NR'R" wherein
R' and R" are taken together with nitrogen to form a cyclic moiety.
[0032] In another aspect, provided herein is a compound of Formula
(XXIII):
R1
N
Wb5
Wd
Formula (XXIII)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Wb5 is N or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
12

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R1 is -(L)-R1';
L is a bond, -S-, -N(R16)-, -C(R15)2-, -C(=0)-, or -0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, heterocyclylalkyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy,
amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, suffinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 isindependently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido,
amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R"
wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is -(CH(R14)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR14),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)C(R14)2-, or
each z is an integer of 1,2, 3, or 4;
each R9 and R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, or
heteroaryl;
each R14 and R15 is independently hydrogen, alkyl, aryl, heteroaryl or halo;
and
N-N)N
Rio_yN R12
wd is R11
wherein R10, R11, R12, and R17 are each independently hydrogen, alkyl,
heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido,
amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy,
nitro, phosphate, urea, carbonate, oxo, or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.
[0033] In some embodiments, the compound of Formula (XXIII) is a
compound of Formula (XXIV):
R3 R1
N
X
Wd
Formula (XXIV).
[0034] In one aspect, provided herein are compounds of Formula (XXV):
13

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
CF3 0
R3 B
N
p0
Wbi Wb5 X
1
Y
\
Wd
Formula (XXV)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Wbi and Wb2 are each independently CR3, S, 0, N or NR13, wherein at least one
of Wbi and Wb2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
Wb5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
X is absent or is ¨(CH(R9)),-;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when Wb5 is N, no more than one of X or Y is absent;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea or carbonate;
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
Wd is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more
Rb, R11 or R12;
wherein each Rb is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl,
alkynyl, cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate,
oxo, or NR'R", wherein R' and R" are taken together with nitrogen to form a
cyclic moiety.
14

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[0035] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula (XXVI):
cF3 0
0
---
x
1
R8
,A,
Y\
"d
Formula (XXVI).
[0036] In some embodiments, the compound of Formula (XXVI) has a
structure of Formula (XXVIa)
or (XXVIb):
CF3 0 CF3 0
B B
I\J N
le 1 le
x x
1 1
R8 \( R 8 Y\
Wd or Wd
Formula (XXVIa) Formula (XXVIb).
[0037] In some embodiments, the compound of Formula (XXVI) has a
structure of Formula (XXVIIa)
or (XXVIIb):
CF3 0 CF3 0
N N
el R9 el
R9
R8 Y. R8 CI
Wd or Wd
Formula (XXVIIa) Formula (XXVIIb).
[0038] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula
(XXVIII):
0
F3C
,B
---'1\1-
N 1 &\
Swb5 x
I
Wd
Formula (XXVIII).

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
[0039] In some embodiments, the compound of Formula (XXVIII) has a
structure of Formula
(XXIX):
0
F3C
NrB
\,Sw R9
b
Wd
Formula (XXIX).
[0040] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula (XXX):
0
F3C
/L1\113
Wb5 X,
Wd
Formula (XXX).
[0041] In
certain embodiments, the compound of Formula (XXX) has a structure of Formula
(XXXI):
F3C 0
N,B
Y,
Wd
Formula (XXXI).
[0042] In certain embodiments, the compound of Formula (XXX) has a
structure of Formula (XXXII):
F3C 0
I N-B
Y,
Wd
Formula (XXXII).
16

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[0043] In some embodiments, the compound of Formula (XXXII) has a
structure of Formula
(XXXIII):
0
F3C
N,B
R9
Y,
Wd
Formula (XXXIII).
[0044] In another aspect, provided herein are compounds of Formula
(XXXIV) or (XXXV):
CF3 CF3
1
R3
N/ R1 N
PO PO
Wb2 - w b2
Wb1 Wb5 Wb1 Wb5
Wd or Wd
Formula (XXXIV) Formula (XXXV)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or their
pharmaceutically acceptable forms thereof, wherein:
Wbi and Wb2 are each independently CR3, S, 0, N or NR13, wherein at least one
of Wbi and Wb2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
Wb5 is Cle, CHle, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
R1 is ¨(L)-Rr;
L is a bond, -S-, -N(R15)-, -C(=0)-, or ¨0-;
Rr is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen, or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
X is absent or is ¨(CH(R16)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R16)-, -C(=0)-(CHR16),-, -C(=0)-,
-N(R16)-C(=0)-, or
-N(R16)-C(=0)NH-,-N(R16)C(R16)2-, or -C(=0)-N(R16)-(CHR16)z-;
each z is an integer of 1,2, 3, or 4;
17

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R13 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl;
Wd is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more
Rb, R11 or R12;
wherein each Rb is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl,
alkynyl, cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate,
oxo, or NR'R", wherein R' and R" are taken together with nitrogen to form a
cyclic moiety.
[0045]
In certain embodiments, a compound as disclosed herein selectively modulates
phosphatidyl
inosito1-3 kinase (PI3 kinase) delta isoform. In certain embodiments, the
compound selectively inhibits the delta
isoform over the beta isoform. By way of non-limiting example, the ratio of
selectivity can be greater than a factor
of about 10, greater than a factor of about 50, greater than a factor of about
100, greater than a factor of about 200,
greater than a factor of about 400, greater than a factor of about 600,
greater than a factor of about 800, greater than
a factor of about 1000, greater than a factor of about 1500, greater than a
factor of about 2000, greater than a factor
of about 5000, greater than a factor of about 10,000, or greater than a factor
of about 20,000, where selectivity can
be measured by IC50, among other means. In certain embodiments, the PI3 kinase
delta isoform IC50 activity of a
compound as disclosed herein can be less than about 1000 nM, less than about
100 nM, less than about 10 nM, or
less than about 1 nM.
[0046]
In certain embodiments, provided herein is a composition (e.g., a
pharmaceutical
composition) comprising a compound as described herein and a pharmaceutically
acceptable excipient. In some
embodiments, provided herein is a method of inhibiting a phosphatidyl inosito1-
3 kinase (PI3 kinase), comprising
contacting the PI3 kinase with an effective amount of a compound or
pharmaceutical composition as described
herein. In certain embodiments, a method is provided for inhibiting a
phosphatidyl inosito1-3 kinase (PI3 kinase)
wherein said PI3 kinase is present in a cell. The inhibition can take place in
a subject suffering from a disorder
selected from cancer, bone disorder, inflammatory disease, immune disease,
nervous system disease (e.g., a
neuropsychiatric disorder), metabolic disease, respiratory disease,
thrombosis, and cardiac disease. In certain
embodiments, a second therapeutic agent is administered to the subject.
[0047]
In certain embodiments, a method is provided of selectively inhibiting a
phosphatidyl inositol-
3 kinase (PI3 kinase) delta isoform over PI3 kinase beta isoform wherein the
inhibition takes place in a cell. Non-
limiting examples of the methods disclosed herein can comprise contacting PI3
kinase delta isoform with an
18

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
effective amount of a compound or pharmaceutical composition as disclosed
herein. In an embodiment, such
contact can occur in a cell.
[0048]
In certain embodiments, a method is provided of selectively inhibiting a
phosphatidyl inositol-
3 kinase (PI3 kinase) delta isoform over PI3 kinase beta isoform wherein the
inhibition takes place in a subject
suffering from a disorder selected from cancer, bone disorder, inflammatory
disease, immune disease, nervous
system disease (e.g., a neuropsychiatric disorder), metabolic disease,
respiratory disease, thrombosis, and cardiac
disease, said method comprising administering an effective amount of a
compound or pharmaceutical composition
to said subject. In certain embodiments, provided herein is a method of
treating a subject suffering from a disorder
associated with phosphatidyl inosito1-3 kinase (PI3 kinase), said method
comprising selectively modulating the
phosphatidyl inosito1-3 kinase (PI3 kinase) delta isoform over PI3 kinase beta
isoform by administering an amount
of a compound or pharmaceutical composition to said subject, wherein said
amount is sufficient for selective
modulation of PI3 kinase delta isoform over PI3 kinase beta isoform.
[0049] In some embodiments, provided herein is a method of making a
compound as described herein.
[0050] In certain embodiments, provided herein is a reaction mixture
comprising a compound as
described herein.
[0051] In certain embodiments, provided herein is a kit comprising a
compound as described herein.
[0052] In some embodiments, a method is provided for treating a disease
or disorder described herein, the
method comprising administering a therapeutically effective amount of a
compound or pharmaceutical composition
described herein to a subject.
[0053] In some embodiments, a method is provided for treating a PI3K
mediated disorder in a subject, the
method comprising administering a therapeutically effective amount of a
compound or pharmaceutical composition
described herein to a subject.
[0054] In some embodiments, provided herein is a use of a compound or a
pharmaceutical composition
described herein for the treatment of a disease or disorder described herein
in a subject.
[0055] In some embodiments, provided herein is a use of a compound or a
pharmaceutical composition
described herein for the treatment of a PI3K mediated disorder in a subject.
[0056] In some embodiments, provided herein is a use of a compound or a
pharmaceutical composition
described herein in the manufacture of a medicament for the treatment of a
disease or disorder described herein in a
subject.
[0057] In certain embodiments, provided herein is use of a compound or a
pharmaceutical composition
described herein in the manufacture of a medicament for the treatment of a
PI3K mediated disorder in a subject.
INCORPORATION BY REFERENCE
[0058]
All publications, patents, and patent applications mentioned in this
specification are herein
incorporated by reference to the same extent as if each individual
publication, patent, or patent application was
specifically and individually indicated to be incorporated by reference. In
case of conflict, the present application,
including any definitions herein, will control.
19

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
DETAILED DESCRIPTION
[0059] In one embodiment, provided are heterocyclyl compounds, and
pharmaceutically acceptable
forms, including, but not limited to, salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives
thereof.
[0060] In another embodiment, provided are methods of treating and/or
managing various diseases
and disorders, which comprises administering to a patient a therapeutically
effective amount of a compound
provided herein, or a pharmaceutically acceptable form (e.g., salts, hydrates,
solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof. Examples of diseases and disorders
are described herein.
[0061] In another embodiment, provided are methods of preventing various
diseases and disorders,
which comprises administering to a patient in need of such prevention a
prophylactically effective amount of a
compound provided herein, or a pharmaceutically acceptable form (e.g., salts,
hydrates, solvates, isomers, prodrugs,
and isotopically labeled derivatives) thereof. Examples of diseases and
disorders are described herein.
[0062] In other embodiments, a compound provided herein, or a
pharmaceutically acceptable form
(e.g., salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, is administered in
combination with another drug ("second active agent") or treatment. Second
active agents include small molecules
and large molecules (e.g., proteins and antibodies), examples of which are
provided herein, as well as stem cells.
Other methods or therapies that can be used in combination with the
administration of compounds provided herein
include, but are not limited to, surgery, blood transfusions, immunotherapy,
biological therapy, radiation therapy,
and other non-drug based therapies presently used to treat, prevent or manage
various disorders described herein.
[0063] Also provided are pharmaceutical compositions (e.g., single unit
dosage forms) that can be
used in the methods provided herein. In one embodiment, pharmaceutical
compositions comprise a compound
provided herein, or a pharmaceutically acceptable form (e.g., salts, hydrates,
solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, and optionally one or more second
active agents.
[0064] While specific embodiments have been discussed, the specification
is illustrative only and not
restrictive. Many variations of this disclosure will become apparent to those
skilled in the art upon review of this
specification.
[0065] Unless defined otherwise, all technical and scientific terms used
herein have the same meaning
as is commonly understood by one of skill in the art to which this
specification pertains.
[0066] As used in the specification and claims, the singular form "a",
"an" and "the" includes plural
references unless the context clearly dictates otherwise.
[0067] As used herein, "agent" or "biologically active agent" or "second
active agent" refers to a
biological, pharmaceutical, or chemical compound or other moiety. Non-limiting
examples include simple or
complex organic or inorganic molecules, a peptide, a protein, an
oligonucleotide, an antibody, an antibody
derivative, an antibody fragment, a vitamin, a vitamin derivative, a
carbohydrate, a toxin, or a chemotherapeutic
compound, and metabolites thereof. Various compounds can be synthesized, for
example, small molecules and
oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic
compounds based on various core
structures. In addition, various natural sources can provide compounds for
screening, such as plant or animal

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
extracts, and the like. A skilled artisan can readily recognize that there is
no limit as to the structural nature of the
agents of this disclosure.
[0068]
The term "agonist" as used herein refers to a compound or agent having the
ability to initiate
or enhance a biological function of a target protein or polypeptide, such as
increasing the activity or expression of
the target protein or polypeptide. Accordingly, the term "agonist" is defined
in the context of the biological role of
the target protein or polypeptide. While some agonists herein specifically
interact with (e.g., bind to) the target,
compounds and/or agents that initiate or enhance a biological activity of the
target protein or polypeptide by
interacting with other members of the signal transduction pathway of which the
target polypeptide is a member are
also specifically included within this definition.
[0069]
The terms "antagonist" and "inhibitor" are used interchangeably, and they
refer to a
compound or agent having the ability to inhibit a biological function of a
target protein or polypeptide, such as by
inhibiting the activity or expression of the target protein or polypeptide.
Accordingly, the terms "antagonist" and
"inhibitor" are defined in the context of the biological role of the target
protein or polypeptide. While some
antagonists herein specifically interact with (e.g., bind to) the target,
compounds that inhibit a biological activity of
the target protein or polypeptide by interacting with other members of the
signal transduction pathway of which the
target protein or polypeptide are also specifically included within this
definition. Non-limiting examples of
biological activity inhibited by an antagonist include those associated with
the development, growth, or spread of a
tumor, or an undesired immune response as manifested in autoimmune disease.
[0070]
An "anti-cancer agent", "anti-tumor agent" or "chemotherapeutic agent" refers
to any agent
useful in the treatment of a neoplastic condition. One class of anti-cancer
agents comprises chemotherapeutic
agents. "Chemotherapy" means the administration of one or more
chemotherapeutic drugs and/or other agents to a
cancer patient by various methods, including intravenous, oral, intramuscular,
intraperitoneal, intravesical,
subcutaneous, transdermal, buccal, or inhalation or in the form of a
suppository.
[0071]
The term "cell proliferation" refers to a phenomenon by which the cell number
has changed
as a result of division. This term also encompasses cell growth by which the
cell morphology has changed (e.g.,
increased in size) consistent with a proliferative signal.
[0072]
The term "co-administration," "administered in combination with," and their
grammatical
equivalents, as used herein, encompasses administration of two or more agents
to subject so that both agents and/or
their metabolites are present in the subject at the same time. Co-
administration includes simultaneous
administration in separate compositions, administration at different times in
separate compositions, or administration
in a composition in which both agents are present.
[0073]
The term "effective amount" or "therapeutically effective amount" refers to
that amount of a
compound or pharmaceutical composition described herein that is sufficient to
effect the intended application
including, but not limited to, disease treatment, as illustrated below. The
therapeutically effective amount can vary
depending upon the intended application (in vitro or in vivo), or the subject
and disease condition being treated, e.g.,
the weight and age of the subject, the severity of the disease condition, the
manner of administration and the like,
which can readily be determined by one of ordinary skill in the art. The term
also applies to a dose that will induce a
21

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
particular response in target cells, e.g., reduction of platelet adhesion
and/or cell migration. The specific dose will
vary depending on, for example, the particular compounds chosen, the dosing
regimen to be followed, whether it is
administered in combination with other agents, timing of administration, the
tissue to which it is administered, and
the physical delivery system in which it is carried.
[0074]
As used herein, the terms "treatment", "treating", "palliating" and
"ameliorating" are used
interchangeably herein. These terms refer to an approach for obtaining
beneficial or desired results including, but
not limited to, therapeutic benefit and/or a prophylactic benefit. By
therapeutic benefit is meant eradication or
amelioration of the underlying disorder being treated. Also, a therapeutic
benefit is achieved with the eradication or
amelioration of one or more of the physiological symptoms associated with the
underlying disorder such that an
improvement is observed in the patient, notwithstanding that the patient can
still be afflicted with the underlying
disorder. For prophylactic benefit, the pharmaceutical compositions can be
administered to a patient at risk of
developing a particular disease, or to a patient reporting one or more of the
physiological symptoms of a disease,
even though a diagnosis of this disease may not have been made.
[0075]
A "therapeutic effect," as that term is used herein, encompasses a therapeutic
benefit and/or a
prophylactic benefit as described above. A prophylactic effect includes
delaying or eliminating the appearance of a
disease or condition, delaying or eliminating the onset of symptoms of a
disease or condition, slowing, halting, or
reversing the progression of a disease or condition, or any combination
thereof.
[0076]
"Signal transduction" is a process during which stimulatory or inhibitory
signals are
transmitted into and within a cell to elicit an intracellular response. A
"modulator" of a signal transduction pathway
refers to a compound which modulates the activity of one or more cellular
proteins mapped to the same specific
signal transduction pathway. A modulator can augment (agonist) or suppress
(antagonist) the activity of a signaling
molecule.
[0077]
The term "selective inhibition" or "selectively inhibit" as applied to a
biologically active
agent refers to the agent's ability to selectively reduce the target signaling
activity as compared to off-target
signaling activity, via direct or interact interaction with the target. For
example, a compound that selectively inhibits
one isoform of PI3K over another isoform of PI3K has an activity of at least
2X against a first isoform relative to the
compound's activity against the second isoform (e.g., at least about 3X, 5X,
10X, 20X, 50X, or 100X).
[0078]
"Radiation therapy" means exposing a patient, using routine methods and
compositions known
to the practitioner, to radiation emitters such as, but not limited to, alpha-
particle emitting radionuclides (e.g.,
actinium and thorium radionuclides), low linear energy transfer (LET)
radiation emitters (i.e., beta emitters),
conversion electron emitters (e.g., strontium-89 and samarium-153-EDTMP), or
high-energy radiation, including
without limitation x-rays, gamma rays, and neutrons.
[0079]
"Subject" to which administration is contemplated includes, but is not limited
to, humans (i.e.,
a male or female of any age group, e.g., a pediatric subject (e.g., infant,
child, adolescent) or adult subject (e.g.,
young adult, middle¨aged adult or senior adult)) and/or other primates (e.g.,
cynomolgus monkeys, rhesus monkeys);
mammals, including commercially relevant mammals such as cattle, pigs, horses,
sheep, goats, cats, and/or dogs;
and/or birds, including commercially relevant birds such as chickens, ducks,
geese, quail, and/or turkeys.
22

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[0080] The term "in vivo" refers to an event that takes place in a
subject's body.
[0081]
The term "in vitro" refers to an event that takes places outside of a
subject's body. For
example, an in vitro assay encompasses any assay conducted outside of a
subject. In vitro assays encompass cell-
based assays in which cells, alive or dead, are employed. In vitro assays also
encompass a cell-free assay in which
no intact cells are employed.
[0082]
As used herein, "pharmaceutically acceptable esters" include, but are not
limited to, alkyl,
alkenyl, alkynyl, aryl, aralkyl, and cycloalkyl esters of acidic groups,
including, but not limited to, carboxylic acids,
phosphoric acids, phosphinic acids, sulfonic acids, sulfinic acids and boronic
acids.
[0083]
As used herein, "pharmaceutically acceptable enol ethers" include, but are not
limited to,
derivatives of formula -C=C(OR) where R can be selected from alkyl, alkenyl,
alkynyl, aryl, aralkyl and cycloalkyl.
Pharmaceutically acceptable enol esters include, but are not limited to,
derivatives of formula -C=C(OC(0)R) where
R can be selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl and
cycloalkyl.
[0084]
As used herein, a "pharmaceutically acceptable form" of a disclosed compound
includes, but is
not limited to, pharmaceutically acceptable salts, hydrates, solvates,
isomers, prodrugs, and isotopically labeled
derivatives of disclosed compounds. In one embodiment, a "pharmaceutically
acceptable form" includes, but is not
limited to, pharmaceutically acceptable salts, isomers, prodrugs and
isotopically labeled derivatives of disclosed
compounds.
[0085]
In certain embodiments, the pharmaceutically acceptable form is a
pharmaceutically acceptable
salt. As used herein, the term "pharmaceutically acceptable salt" refers to
those salts which are, within the scope of
sound medical judgment, suitable for use in contact with the tissues of
subjects without undue toxicity, irritation,
allergic response and the like, and are commensurate with a reasonable
benefit/risk ratio. Pharmaceutically
acceptable salts are well known in the art. For example, Berge et al.
describes pharmaceutically acceptable salts in
detail in J Pharmaceutical Sciences (1977) 66:1-19. Pharmaceutically
acceptable salts of the compounds provided
herein include those derived from suitable inorganic and organic acids and
bases. Examples of pharmaceutically
acceptable, nontoxic acid addition salts are salts of an amino group formed
with inorganic acids such as
hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and
perchloric acid or with organic acids such as
acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic
acid or malonic acid or by using other methods
used in the art such as ion exchange. Other pharmaceutically acceptable salts
include adipate, alginate, ascorbate,
aspartate, benzenesulfonate, besylate, benzoate, bisulfate, borate, butyrate,
camphorate, camphorsulfonate, citrate,
cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate,
fumarate, glucoheptonate,
glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide,
2¨hydroxy¨ethanesulfonate,
lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate,
methanesulfonate, 2¨naphthalenesulfonate,
nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate,
persulfate, 3¨phenylpropionate, phosphate, picrate,
pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate,
p¨toluenesulfonate, undecanoate, valerate
salts, and the like. In some embodiments, organic acids from which salts can
be derived include, for example, acetic
acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid,
malonic acid, succinic acid, fumaric acid,
23

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid,
methanesulfonic acid, ethanesulfonic acid,
p-toluenesulfonic acid, salicylic acid, and the like.
[0086] Pharmaceutically acceptable salts derived from appropriate bases
include alkali metal, alkaline
earth metal, ammonium and N'(C1_4alky1)4- salts. Representative alkali or
alkaline earth metal salts include sodium,
lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese,
aluminum, and the like. Further
pharmaceutically acceptable salts include, when appropriate, nontoxic
ammonium, quaternary ammonium, and
amine cations formed using counterions such as halide, hydroxide, carboxylate,
sulfate, phosphate, nitrate, lower
alkyl sulfonate and aryl sulfonate. Organic bases from which salts can be
derived include, for example, primary,
secondary, and tertiary amines, substituted amines including naturally
occurring substituted amines, cyclic amines,
basic ion exchange resins, and the like, such as isopropylamine,
trimethylamine, diethylamine, triethylamine,
tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically
acceptable base addition salt is
chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
[0087] In certain embodiments, the pharmaceutically acceptable form is a
"solvate" (e.g., a hydrate).
As used herein, the term "solvate" refers to compounds that further include a
stoichiometric or non-stoichiometric
amount of solvent bound by non-covalent intermolecular forces. The solvate can
be of a disclosed compound or a
pharmaceutically acceptable salt thereof. Where the solvent is water, the
solvate is a "hydrate". Pharmaceutically
acceptable solvates and hydrates are complexes that, for example, can include
1 to about 100, or 1 to about 10, or
one to about 2, about 3 or about 4, solvent or water molecules. It will be
understood that the term "compound" as
used herein encompasses the compound and solvates of the compound, as well as
mixtures thereof.
[0088] In certain embodiments, the pharmaceutically acceptable form is a
prodrug. As used herein,
the term "prodrug" refers to compounds that are transformed in vivo to yield a
disclosed compound or a
pharmaceutically acceptable form of the compound. A prodrug can be inactive
when administered to a subject, but
is converted in vivo to an active compound, for example, by hydrolysis (e.g.,
hydrolysis in blood). In certain cases, a
prodrug has improved physical and/or delivery properties over the parent
compound. Prodrugs are typically
designed to enhance pharmaceutically and/or pharmacokinetically based
properties associated with the parent
compound. The prodrug compound often offers advantages of solubility, tissue
compatibility or delayed release in a
mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-
9, 21-24 (Elsevier, Amsterdam). A
discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel
Delivery Systems," A.C.S. Symposium
Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B.
Roche, American Pharmaceutical
Association and Pergamon Press, 1987, both of which are incorporated in full
by reference herein. Exemplary
advantages of a prodrug can include, but are not limited to, its physical
properties, such as enhanced water solubility
for parenteral administration at physiological pH compared to the parent
compound, or it enhances absorption from
the digestive tract, or it can enhance drug stability for long¨term storage.
[0089] The term "prodrug" is also meant to include any covalently bonded
carriers, which release the
active compound in vivo when such prodrug is administered to a subject.
Prodrugs of an active compound, as
described herein, can be prepared by modifying functional groups present in
the active compound in such a way that
the modifications are cleaved, either in routine manipulation or in vivo, to
the parent active compound. Prodrugs
24

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
include compounds wherein a hydroxy, amino or mercapto group is bonded to any
group that, when the prodrug of
the active compound is administered to a subject, cleaves to form a free
hydroxy, free amino or free mercapto group,
respectively. Examples of prodrugs include, but are not limited to, acetate,
formate and benzoate derivatives of an
alcohol or acetamide, formamide and benzamide derivatives of an amine
functional group in the active
compound and the like. Other examples of prodrugs include compounds that
comprise -NO, -NO2, -ONO, or -
0NO2 moieties. Prodrugs can typically be prepared using well-known methods,
such as those described in Burger's
Medicinal Chemistry and Drug Discovery, 172-178, 949-982 (Manfred E. Wolff
ed., 5th ed., 1995), and Design of
Prodrugs (H. Bundgaard ed., Elselvier, New York, 1985).
[0090] For example, if a disclosed compound or a pharmaceutically
acceptable form of the compound
contains a carboxylic acid functional group, a prodrug can comprise a
pharmaceutically acceptable ester formed by
the replacement of the hydrogen atom of the acid group with a group such as
(C1-C8)alkyl, (C2-
C12)alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-
methyl-1-(alkanoyloxy)-ethyl
having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6
carbon atoms, 1-
(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1-
(alkoxycarbonyloxy)ethyl having from 5 to
8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms,

14N-(alkoxycarbonyBamino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl,
4-crotonolactonyl, gamma-
butyrolacton-4-yl, di-N,N-(C1-C2)alkylamino(C2-C3)alkyl (such as 13-
dimethylaminoethyl), carbamoyl-(C1-C2)alkyl,
N,N-di(Ci-C2)alkylcarbamoy1-(C1-C2)alkyl and piperidino-, pyrrolidino- or
morpholino(C2-C3)alkyl.
[0091] Similarly, if a disclosed compound or a pharmaceutically
acceptable form of the compound
contains an alcohol functional group, a prodrug can be formed by the
replacement of the hydrogen atom of the
alcohol group with a group such as (C1-C6)alkanoyloxymethyl, 14(C1-
C6)alkanoyloxy)ethyl,
1-methyl-1 -((C1-C6)alkanoyloxy)ethyl (C1-C6)alkoxycarbonyloxymethyl, N-(C1-
C6)alkoxyc arbonylaminomethyl,
succinoyl, (C1-C6)alkanoyl, a-amino(C1-C4)alkanoyl, arylacyl and a-aminoacyl,
or a-aminoacyl-a-aminoacyl, where
each a-aminoacyl group is independently selected from the naturally occurring
L-amino acids, P(0)(OH)2,
-P(0)(0(C1-C6)alkyl)2 or glycosyl (the radical resulting from the removal of a
hydroxyl group of the hemiacetal
form of a carbohydrate).
[0092] If a disclosed compound or a pharmaceutically acceptable form of
the compound incorporates
an amine functional group, a prodrug can be formed by the replacement of a
hydrogen atom in the amine group with
a group such as R-carbonyl, RO-carbonyl, NRR'-carbonyl where R and R' are each
independently (C1-C1o)alkyl,
(C3-C7)cycloalkyl, benzyl, a natural a-aminoacyl or natural a-aminoacyl-
natural a-aminoacyl, ¨C(OH)C(0)0Y1
wherein Y1 is H, (C1-C6)alkyl or benzyl, -C(0Y2)Y3 wherein Y2 is (C1-C4) alkyl
and Y3 is (C1-C6)alkyl, carboxy(C1-
C6)alkyl, amino(C1-C4)alkyl or mono-N¨ or di-N,N¨(C1-C6)alkylaminoalkyl,
¨C(Y4)Y5 wherein Y4 is H or
methyl and Y5 is mono-N¨ or di-N,N¨(C1-C6)alkylamino, morpholino, piperidin- 1
-y1 or pyrrolidin- 1 -yl.
[0093] In certain embodiments, the pharmaceutically acceptable form is
an isomer. "Isomers" are
different compounds that have the same molecular formula. "Stereoisomers" are
isomers that differ only in the way
the atoms are arranged in space. As used herein, the term "isomer" includes
any and all geometric isomers and
stereoisomers. For example, "isomers" include geometric double bond cis¨ and
trans¨isomers, also termed E¨ and

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Z¨ isomers; R¨ and S¨enantiomers; diastereomers, (d)¨isomers and (1)¨isomers,
racemic mixtures thereof; and other
mixtures thereof, as falling within the scope of this disclosure.
[0094] Geometric isomers can be represented by the symbol ¨ which
denotes a bond that can
be a single, double or triple bond as described herein. Provided herein are
various geometric isomers and mixtures
thereof resulting from the arrangement of substituents around a carbon-carbon
double bond or arrangement of
substituents around a carbocyclic ring. Substituents around a carbon-carbon
double bond are designated as being in
the "Z" or "E' configuration wherein the terms "Z" and "E" are used in
accordance with IUPAC standards. Unless
otherwise specified, structures depicting double bonds encompass both the "E"
and "Z" isomers.
[0095] Substituents around a carbon-carbon double bond alternatively can
be referred to as "cis" or
"trans," where "cis" represents substituents on the same side of the double
bond and "trans" represents substituents
on opposite sides of the double bond. The arrangement of substituents around a
carbocyclic ring can also be
designated as "cis" or "trans." The term "cis" represents substituents on the
same side of the plane of the ring, and
the term "trans" represents substituents on opposite sides of the plane of the
ring. Mixtures of compounds wherein
the substituents are disposed on both the same and opposite sides of plane of
the ring are designated "cis/trans."
[0096] "Enantiomers" are a pair of stereoisomers that are non-
superimposable mirror images of each
other. A mixture of a pair of enantiomers in any proportion can be known as a
"racemic" mixture. The term "( )" is
used to designate a racemic mixture where appropriate. "Diastereoisomers" are
stereoisomers that have at least two
asymmetric atoms, but which are not mirror-images of each other. The absolute
stereochemistry is specified
according to the Cahn-Ingold-Prelog R-S system. When a compound is an
enantiomer, the stereochemistry at each
chiral carbon can be specified by either R or S. Resolved compounds whose
absolute configuration is unknown can
be designated (+) or (-) depending on the direction (dextro- or levorotatory)
which they rotate plane polarized light
at the wavelength of the sodium D line. Certain of the compounds described
herein contain one or more asymmetric
centers and can thus give rise to enantiomers, diastereomers, and other
stereoisomeric forms that can be defined, in
terms of absolute stereochemistry at each asymmetric atom, as (R)- or (S)-.
The present chemical entities,
pharmaceutical compositions and methods are meant to include all such possible
isomers, including racemic
mixtures, optically substantially pure forms and intermediate mixtures.
Optically active (R)- and (S)- isomers can
be prepared, for example, using chiral synthons or chiral reagents, or
resolved using conventional techniques.
[0097] The "enantiomeric excess" or "% enantiomeric excess" of a
composition can be calculated
using the equation shown below. In the example shown below, a composition
contains 90% of one enantiomer, e.g.,
the S enantiomer, and 10% of the other enantiomer, e.g., the R enantiomer.
ee = (90-10)/100 = 80%.
[0098] Thus, a composition containing 90% of one enantiomer and 10% of
the other enantiomer is
said to have an enantiomeric excess of 80%. Some compositions described herein
contain an enantiomeric excess of
at least about 50%, about 75%, about 90%, about 95%, or about 99% of the S
enantiomer. In other words, the
compositions contain an enantiomeric excess of the S enantiomer over the R
enantiomer. In other embodiments,
some compositions described herein contain an enantiomeric excess of at least
about 50%, about 75%, about 90%,
26

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
about 95%, or about 99% of the R enantiomer. In other words, the compositions
contain an enantiomeric excess of
the R enantiomer over the S enantiomer.
[0099]
For instance, an isomer/enantiomer can, in some embodiments, be provided
substantially free
of the corresponding enantiomer, and can also be referred to as "optically
enriched," "enantiomerically enriched,"
"enantiomerically pure" and "non-racemic," as used interchangeably herein.
These terms refer to compositions in
which the percent by weight of one enantiomer is greater than the amount of
that one enantiomer in a control
mixture of the racemic composition (e.g., greater than 1:1 by weight). For
example, an enantiomerically enriched
preparation of the S enantiomer, means a preparation of the compound having
greater than about 50% by weight of
the S enantiomer relative to the R enantiomer, such as at least about 75% by
weight, further such as at least about
80% by weight. In some embodiments, the enrichment can be much greater than
about 80% by weight, providing a
"substantially enantiomerically enriched," "substantially enantiomerically
pure" or a "substantially non-racemic"
preparation, which refers to preparations of compositions which have at least
about 85% by weight of one
enantiomer relative to other enantiomer, such as at least about 90% by weight,
and further such as at least about 95%
by weight. In certain embodiments, the compound provided herein is made up of
at least about 90% by weight of
one enantiomer. In other embodiments, the compound is made up of at least
about 95%, about 98%, or about 99%
by weight of one enantiomer.
[00100]
In some embodiments, the compound is a racemic mixture of (S)- and (R)-
isomers. In other
embodiments, provided herein is a mixture of compounds wherein individual
compounds of the mixture exist
predominately in an (S)- or (R)- isomeric configuration. For example, the
compound mixture has an (S)-
enantiomeric excess of greater than about 55%, about 60%, about 65%, about
70%, about 75%, about 80%, about
85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about
99.5%, or more. In other
embodiments, the compound mixture has an (S)-enantiomeric excess of greater
than about 55% to about 99.5%,
greater than about about 60% to about 99.5%, greater than about 65% to about
99.5%, greater than about 70% to
about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to
about 99.5%, greater than about
85% to about 99.5%, greater than about 90% to about 99.5%, greater than about
95% to about 99.5%, greater than
about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than
about 98% to greater than about
99.5%, greater than about 99% to about 99.5%, or more.
[00101]
In other embodiments, the compound mixture has an (R)-enantiomeric purity of
greater than
about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%,
about 90%, about 95%, about
96%, about 97%, about 98%, about 99%, about 99.5% or more. In some other
embodiments, the compound mixture
has an (R)-enantiomeric excess of greater than about 55% to about 99.5%,
greater than about about 60% to about
99.5%, greater than about 65% to about 99.5%, greater than about 70% to about
99.5%, greater than about 75% to
about 99.5%, greater than about 80% to about 99.5%, greater than about 85% to
about 99.5%, greater than about
90% to about 99.5%, greater than about 95% to about 99.5%, greater than about
96% to about 99.5%, greater than
about 97% to about 99.5%, greater than about 98% to greater than about 99.5%,
greater than about 99% to about
99.5% or more.
27

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00102]
In other embodiments, the compound mixture contains identical chemical
entities except for
their stereochemical orientations, namely (S)- or (R)- isomers. For example,
if a compound disclosed herein has -
CH(R)- unit, and R is not hydrogen, then the -CH(R)- is in an (S)- or (R)-
stereochemical orientation for each of the
identical chemical entities. In some embodiments, the mixture of identical
chemical entities is a racemic mixture of
(S)- and (R)- isomers. In another embodiment, the mixture of the identical
chemical entities (except for their
stereochemical orientations), contain predominately (S)-isomers or
predominately (R)- isomers. For example, the
(S)- isomers in the mixture of identical chemical entities are present at
about 55%, about 60%, about 65%, about
70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about
97%, about 98%, about 99%,
about 99.5% ,or more, relative to the (R)- isomers. In some embodiments, the
(S)- isomers in the mixture of
identical chemical entities are present at an (S)-enantiomeric excess of
greater than about 55% to about 99.5%,
greater than about about 60% to about 99.5%, greater than about 65% to about
99.5%, greater than about 70% to
about 99.5%, greater than about 75% to about 99.5%, greater than about 80% to
about 99.5%, greater than about
85% to about 99.5%, greater than about 90% to about 99.5%, greater than about
95% to about 99.5%, greater than
about 96% to about 99.5%, greater than about 97% to about 99.5%, greater than
about 98% to greater than about
99.5%, greater than about 99% to about 99.5% or more.
[00103]
In another embodiment, the (R)- isomers in the mixture of identical chemical
entities (except
for their stereochemical orientations), are present at about 55%, about 60%,
about 65%, about 70%, about 75%,
about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%,
about 99%, about 99.5%, or
more, relative to the (S)- isomers. In some embodiments, the (R)- isomers in
the mixture of identical chemical
entities (except for their stereochemical orientations), are present at a (R)-
enantiomeric excess greater than about
55% to about 99.5%, greater than about about 60% to about 99.5%, greater than
about 65% to about 99.5%, greater
than about 70% to about 99.5%, greater than about 75% to about 99.5%, greater
than about 80% to about 99.5%,
greater than about 85% to about 99.5%, greater than about 90% to about 99.5%,
greater than about 95% to about
99.5%, greater than about 96% to about 99.5%, greater than about 97% to about
99.5%, greater than about 98% to
greater than about 99.5%, greater than about 99% to about 99.5%, or more.
[00104]
Enantiomers can be isolated from racemic mixtures by any method known to those
skilled in
the art, including chiral high pressure liquid chromatography (HPLC), the
formation and crystallization of chiral
salts, or prepared by asymmetric syntheses. See, for example, Enantiomers,
Racemates and Resolutions (Jacques,
Ed., Wiley Interscience, New York, 1981); Wilen et al., Tetrahedron 33:2725
(1977); Stereochemistry of Carbon
Compounds (E.L. Eliel, Ed., McGraw¨Hill, NY, 1962); and Tables of Resolving
Agents and Optical Resolutions p.
268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972).
[00105]
In certain embodiments, the pharmaceutically acceptable form is a tautomer. As
used herein,
the term "tautomer" is a type of isomer that includes two or more
interconvertable compounds resulting from at least
one formal migration of a hydrogen atom and at least one change in valency
(e.g., a single bond to a double bond, a
triple bond to a single bond, or vice versa). "Tautomerization" includes
prototropic or proton-shift tautomerization,
which is considered a subset of acid-base chemistry.
"Prototropic tautomerization" or "proton-shift
tautomerization" involves the migration of a proton accompanied by changes in
bond order. The exact ratio of the
28

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
tautomers depends on several factors, including temperature, solvent, and pH.
Where tautomerization is possible
(e.g., in solution), a chemical equilibrium of tautomers can be reached.
Tautomerizations (i.e., the reaction
providing a tautomeric pair) can be catalyzed by acid or base, or can occur
without the action or presence of an
external agent. Exemplary tautomerizations include, but are not limited to,
keto¨to¨enol; amide¨to¨imide; lactam¨
to¨lactim; enamine¨to¨imine; and enamine¨to¨(a different) enamine
tautomerizations. A specific example of
keto-enol tautomerization is the interconversion of pentane-2,4-dione and 4-
hydroxypent-3-en-2-one tautomers.
Another example of tautomerization is phenol-keto tautomerization. A specific
example of phenol-keto
tautomerization is the interconversion of pyridin-4-ol and pyridin-4(1H)-one
tautomers.
[00106]
Unless otherwise stated, structures depicted herein are also meant to include
compounds which
differ only in the presence of one or more isotopically enriched atoms. For
example, compounds having the present
structures except for the replacement of a hydrogen by a deuterium or tritium,
or the replacement of a carbon by 13C-
or 14C-enriched carbon are within the scope of this disclosure.
[00107]
The disclosure also embraces isotopically labeled compounds which are
identical to those
recited herein, except that one or more atoms are replaced by an atom having
an atomic mass or mass number
different from the atomic mass or mass number usually found in nature.
Examples of isotopes that can be
incorporated into disclosed compounds include isotopes of hydrogen, carbon,
nitrogen, oxygen, phosphorus, fluorine
and chlorine, such as 2H, 3H, 13C, 14C, 15N, 180, 170, 31F,, 32F,, 35s,
r and 36C1, respectively. Certain isotopically-
labeled disclosed compounds (e.g., those labeled with 3H and 14C) are useful
in compound and/or substrate tissue
distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes
can allow for ease of preparation and
delectability. Further, substitution with heavier isotopes such as deuterium
(i.e., 2H) can afford certain therapeutic
advantages resulting from greater metabolic stability (e.g., increased in vivo
half-life or reduced dosage
requirements). Isotopically labeled disclosed compounds can generally be
prepared by substituting an isotopically
labeled reagent for a non-isotopically labeled reagent. In some embodiments,
provided herein are compounds that
can also contain unnatural proportions of atomic isotopes at one or more of
atoms that constitute such compounds.
All isotopic variations of the compounds as disclosed herein, whether
radioactive or not, are encompassed within the
scope of the present disclosure.
[00108]
"Pharmaceutically acceptable carrier" or "pharmaceutically acceptable
excipient" includes
any and all solvents, dispersion media, coatings, antibacterial and antifungal
agents, isotonic and absorption
delaying agents and the like. The use of such media and agents for
pharmaceutically active substances is well
known in the art. Except insofar as any conventional media or agent is
incompatible with the active ingredient, its
use in the therapeutic compositions as disclosed herein is contemplated.
Supplementary active ingredients can also
be incorporated into the pharmaceutical compositions.
[00109]
Definitions of specific functional groups and chemical terms are described in
more detail
below. The chemical elements are identified in accordance with the Periodic
Table of the Elements, CAS version,
Handbook of Chemistry and Physics, 75th ed., inside cover, and specific
functional groups are generally defined as
described therein. Additionally, general principles of organic chemistry, as
well as specific functional moieties and
reactivity, are described in Organic Chemistry, Thomas Sorrell, University
Science Books, Sausalito, 1999; Smith
29

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
and March March's Advanced Organic Chemistry, 5th ed., John Wiley & Sons,
Inc., New York, 2001; Larock,
Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989;
and Carruthers, Some Modern
Methods of Organic Synthesis, 3rd ed., Cambridge University Press, Cambridge,
1987.
[00110]
When a range of values is listed, it is intended to encompass each value and
sub-range within
the range. For example "C1_6 alkyl" is intended to encompass, C1, C2, C3, C4,
C5, C6, C1-6, C1-5, C1-4, C1-3, C1-2, C2-
6, C2-5, C2-4, C2-3, C3-6, C3-5, C3-4, C4-6, C4_5, and C5_6 alkyl.
[00111]
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting
solely of carbon
and hydrogen atoms, containing no unsaturation, having from one to ten carbon
atoms (e.g., C1-C10 alkyl).
Whenever it appears herein, a numerical range such as "1 to 10" refers to each
integer in the given range; e.g., "1 to
carbon atoms" means that the alkyl group can consist of 1 carbon atom, 2
carbon atoms, 3 carbon atoms, etc., up
to and including 10 carbon atoms, although the present definition also covers
the occurrence of the term "alkyl"
where no numerical range is designated. In some embodiments, it is a C1-C6
alkyl group. In some embodiments,
alkyl groups have 1 to 10, 1 to 6, or 1 to 3 carbon atoms. Representative
saturated straight chain alkyls include, but
are not limited to, -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, and -n-
hexyl; while saturated branched alkyls
include, but are not limited to, -isopropyl, -sec-butyl, -isobutyl, -tert-
butyl, -isopentyl, 2-methylbutyl, 3-methylbutyl,
2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-methylhexyl, 3-methylhexyl,
4-methylhexyl, 5-methylhexyl, 2,3-
dimethylbutyl, and the like. The alkyl is attached to the parent molecule by a
single bond. Unless stated otherwise
in the specification, an alkyl group is optionally substituted by one or more
of substituents which independently
include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl,
aralkyl, aryl, aryloxy, amino, amido, amidino,
imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy,
cyano, halo, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo,
phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamidyl,
sulfoxyl, sulfonate, urea, -Si(Ra)3-, -
OR', -SRa, -0C(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2, -
N(Ra)C(0)01ta, -N(Ra)C(0)Ra, -
N(Ra)C(0)N(Ra)2,-N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0),Ra (where t is 1 or 2), -
S(0),ORa (where t is 1 or 2),
-S(0)tN(Ra)2 (where t is 1 or 2), or -0-P(=0)(01ta)2 where each It is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl,
and each of these moieties can be optionally substituted as defined herein.
[00112]
"Perhaloalkyl" refers to an alkyl group in which all of the hydrogen atoms
have been replaced
with a halogen selected from fluoro, chloro, bromo, and iodo. In some
embodiments, all of the hydrogen atoms are
each replaced with fluoro. In some embodiments, all of the hydrogen atoms are
each replaced with chloro.
Examples of perhaloalkyl groups include -CF3, -CF2CF3, -CF2CF2CF3, -CC13, -
CFC12, -CF2C1 and the like.
[00113]
"Alkyl-cycloalkyl" refers to an -(alkyl)cycloalkyl radical where alkyl and
cycloalkyl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for alkyl and cycloalkyl respectively. The "alkyl-cycloalkyl" is
bonded to the parent molecular structure
through the alkyl group. The terms "alkenyl-cycloalkyl" and "alkynyl-
cycloalkyl" mirror the above description of
"alkyl-cycloalkyl" wherein the term "alkyl" is replaced with "alkenyl" or
"alkynyl" respectively, and "alkenyl" or
"alkynyl" are as described herein.

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00114]
"Alkylaryl" refers to an -(alkyl)aryl radical where aryl and alkyl are as
disclosed herein and
which are optionally substituted by one or more of the substituents described
as suitable substituents for aryl and
alkyl respectively. The "alkylaryl" is bonded to the parent molecular
structure through the alkyl group. The terms
"-(alkenyl)aryl" and "-(alkynyl)aryl" mirror the above description of "-
(alkyl)aryl" wherein the term "alkyl" is
replaced with "alkenyl" or "alkynyl" respectively, and "alkenyl" or "alkynyl"
are as described herein.
[00115]
"Alkyl-heteroaryl" refers to an -(alkyl)heteroaryl radical where heteroaryl
and alkyl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for heteroaryl and alkyl respectively. The "alkyl-heteroaryl" is
bonded to the parent molecular structure
through the alkyl group. The terms "-(alkenyl)heteroaryl" and "-
(alkynyl)heteroaryl" mirror the above description
of "-(alkyl)heteroaryl" wherein the term "alkyl" is replaced with "alkenyl" or
"alkynyl" respectively, and "alkenyl"
or "alkynyl" are as described herein.
[00116]
"Alkyl-heterocyclyl" refers to an ¨(alkyl)heterocycyl radical where alkyl and
heterocyclyl are
as disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for heterocyclyl and alkyl respectively. The "alkyl-heterocyclyl"
is bonded to the parent molecular
structure through the alkyl group. The terms "-(alkenyl)heterocyclyl" and "-
(alkynyl)heterocyclyl" mirror the above
description of "-(alkyl)heterocyclyl" wherein the term "alkyl" is replaced
with "alkenyl" or "alkynyl" respectively,
and "alkenyl" or "alkynyl" are as described herein.
[00117]
"Alkenyl" refers to a straight or branched hydrocarbon chain radical group
consisting solely
of carbon and hydrogen atoms, containing at least one double bond, and having
from two to ten carbon atoms (i.e.,
C2-C10 alkenyl). Whenever it appears herein, a numerical range such as "2 to
10" refers to each integer in the given
range; e.g., "2 to 10 carbon atoms" means that the alkenyl group can consist
of 2 carbon atoms, 3 carbon atoms, etc.,
up to and including 10 carbon atoms. In certain embodiments, an alkenyl
comprises two to eight carbon atoms. In
other embodiments, an alkenyl comprises two to five carbon atoms (e.g., C2-05
alkenyl). The alkenyl is attached to
the parent molecular structure by a single bond, for example, ethenyl (i.e.,
vinyl), prop-1 -enyl (i.e., allyl), but-1 -enyl,
pent-1 -enyl, penta-1,4-dienyl, and the like. The one or more carbon¨carbon
double bonds can be internal (such as in
2¨butenyl) or terminal (such as in 1¨buteny1). Examples of C2-4 alkenyl groups
include ethenyl (C2), 1¨propenyl
(C3), 2¨propenyl (C3), 1¨butenyl (C4), 2¨butenyl (C4), butadienyl (C4) and the
like. Examples of C2_6 alkenyl
groups include the aforementioned C2_4 alkenyl groups as well as pentenyl
(C5), pentadienyl (C5), hexenyl (C6) and
the like. Additional examples of alkenyl include heptenyl (C7), octenyl (C8),
octatrienyl (C8) and the like. Unless
stated otherwise in the specification, an alkenyl group is optionally
substituted by one or more substituents which
independently include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy, amino,
amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl,
heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy, haloalkyl, ester, ether,
mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl,
sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ita)3-,
SRa,-0C(0)-Ita, -N(It3)2, -C(0)Ita, -C(0)0Ita, -0C(0)N(R3)2, -C(0)N(Ita)2,
-N(Ita)C(0)01ta, -N(Ita)C(0)Ita, - N(Ita)C(0)N(Ra)2, N(Ita)C(NRa)N(Ra)2, -
N(Ita)S(0)tle (where t is 1 or 2),
-S(0)tOlta (where t is 1 or 2), -S(0)N(Ita)2 (where t is 1 or 2), or ¨0-
P(=0)(01ta)2 where each Ita is independently
31

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl,
heteroaryl or heteroarylalkyl, and each of these moieties can be optionally
substituted as defined herein.
[00118]
"Alkynyl" refers to a straight or branched hydrocarbon chain radical group
consisting solely
of carbon and hydrogen atoms, containing at least one triple bond, having from
two to ten carbon atoms (i.e., C2-C10
alkynyl). Whenever it appears herein, a numerical range such as "2 to 10"
refers to each integer in the given range;
e.g., "2 to 10 carbon atoms" means that the alkynyl group can consist of 2
carbon atoms, 3 carbon atoms, etc., up to
and including 10 carbon atoms. In certain embodiments, an alkynyl comprises
two to eight carbon atoms. In other
embodiments, an alkynyl has two to five carbon atoms (e.g., C2-05 alkynyl).
The alkynyl is attached to the parent
molecular structure by a single bond, for example, ethynyl, propynyl, butynyl,
pentynyl, hexynyl, and the like.
Unless stated otherwise in the specification, an alkynyl group is optionally
substituted by one or more substituents
which independently include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy,
amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl,
heteroalkyl, heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy, haloalkyl, ester, ether,
mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl,
sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -Slta, -0C(0)-Ra, -N(Ra)2, -C(0)Ra, -
C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2,
-N(Ra)C(0)01ta, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -
N(Ra)S(0)tRa (where t is 1 or 2),
-S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-
P(=0)(01ta)2 where each It is independently
hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl,
heteroaryl or heteroarylalkyl, and each of these moieties can be optionally
substituted as defined herein.
[00119]
The term "alkoxy" refers to the group -0-alkyl, including from 1 to 10 carbon
atoms of a
straight, branched, cyclic configuration and combinations thereof, attached to
the parent molecular structure through
an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy,
cyclopropyloxy, cyclohexyloxy and the like.
"Lower alkoxy" refers to alkoxy groups containing one to six carbons. In some
embodiments, C1-C4 alkoxy is an
alkoxy group which encompasses both straight and branched chain alkyls of from
1 to 4 carbon atoms. Unless
stated otherwise in the specification, an alkoxy group is optionally
substituted by one or more substituents which
independently include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy, amino,
amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl,
heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy, haloalkyl, ester, ether,
mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl,
sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -Slta, -0C(0)-Ra, -N(Ra)2, -C(0)Ra, -
C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2,
-N(Ra)C(0)01ta, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -
N(Ra)S(0)tRa (where t is 1 or 2),
-S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-
P(=0)(01ta)2 where each It' is independently
hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl,
heteroaryl or heteroarylalkyl, and each of these moieties can be optionally
substituted as defined herein. The terms
"alkenoxy" and "alkynoxy" mirror the above description of "alkoxy" wherein the
prefix "alk" is replaced with
"alken" or "alkyn" respectively, and the parent "alkenyl" or "alkynyl" terms
are as described herein.
32

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00120]
The term "alkoxycarbonyl" refers to a group of the formula (alkoxy)(C=0)-
attached to the
parent molecular structure through the carbonyl carbon having from 1 to 10
carbon atoms. Thus a Ci-C6
alkoxycarbonyl group is an alkoxy group having from 1 to 6 carbon atoms
attached through its oxygen to a carbonyl
linker. The C1-C6 designation does not include the carbonyl carbon in the atom
count. "Lower alkoxycarbonyl"
refers to an alkoxycarbonyl group wherein the alkyl portion of the alkoxy
group is a lower alkyl group. In some
embodiments, C1-C4 alkoxy is an alkoxy group which encompasses both straight
and branched chain alkoxy groups
of from 1 to 4 carbon atoms. Unless stated otherwise in the specification, an
alkoxycarbonyl group is optionally
substituted by one or more substituents which independently include: acyl,
alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy, amino, amido, amidino, imino, azide,
carbonate, carbamate, carbonyl, heteroalkyl,
heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halo,
haloalkoxy, haloalkyl, ester, ether, mercapto,
thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate,
phosphinate, silyl, sulfinyl, sulfonyl,
sulfonamidyl, sulfoxyl, sulfonate, urea, -Si(Ra)3-, -0Ra, -Slta, -0C(0)4Ra, -
N(Ra)2, -C(0)Ita, -C(0)0Ita,
-0C(0)N(Ra)2, -C(0)N(Ra)2, -N(Ita)C (0)0Ra, -N(Ita)C (0)Ra, - N(Ra)C(0)N(Ra)2,
N(Ra)C(NRa)N(Ra)2,
-N(Ra)S(0),Ra (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0),N(Ra)2
(where t is 1 or 2), or
where each It is independently hydrogen, alkyl, haloalkyl, carbocyclyl,
carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, and
each of these moieties can be optionally
substituted as defined herein. The terms "alkenoxycarbonyl" and
"alkynoxycarbonyl" mirror the above description
of "alkoxycarbonyl" wherein the prefix "alk" is replaced with "alken" or
"alkyn" respectively, and the parent
"alkenyl" or "alkynyl" terms are as described herein.
[00121]
"Acyl" refers to R-C(0)- groups such as, but not limited to, (alkyl)-C(0)-,
(alkenyl)-C(0)-,
(alkynyl)-C (0)- , (aryl)-C(0)-, (cycloalkyl)-C (0)- ,
(heteroaryl)-C (0)- , (heteroalkyl)-C(0)-, and
(heterocycloalkyl)-C(0)-, wherein the group is attached to the parent
molecular structure through the carbonyl
functionality. In some embodiments, it is a C1-C10 acyl radical which refers
to the total number of chain or ring
atoms of the, for example, alkyl, alkenyl, alkynyl, aryl, cyclohexyl,
heteroaryl or heterocycloalkyl portion plus the
carbonyl carbon of acyl. For example, a C4-acyl has three other ring or chain
atoms plus carbonyl. If the R radical
is heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute
to the total number of chain or ring
atoms. Unless stated otherwise in the specification, the "R" of an acyloxy
group can be optionally substituted by
one or more substituents which independently include: acyl, alkyl, alkenyl,
alkynyl, alkoxy, alkylaryl, cycloalkyl,
aralkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate,
carbamate, carbonyl, heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy,
haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate,
silyl, suffinyl, sulfonyl, sulfonamidyl,
sulfoxyl, sulfonate, urea, -Si(Ra)3-, -OR', -SRa, -0C(0)-Ra, -N(Ra)2, -
C(0)Ita, -C(0)01ta, -0C(0)N(Ra)2,
-C(0)N(Ra)2, -N(Ra)C(0)01ta, -N(Ra)C(0)Ita, - N(Ra)C(0)N(Ra)2,
N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tle (where t is 1
or 2), -S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or -0-
P(=0)(01ta)2 where each Ita is
independently hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl,
aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, and each of these
moieties can be optionally substituted as
defined herein.
33

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00122] "Acyloxy" refers to a R(C=0)0- radical wherein "R" can be
alkyl, alkenyl, alkynyl,
heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, cyclohexyl, heteroaryl or
heterocycloalkyl, which are as described
herein. The acyloxy group is attached to the parent molecular structure
through the oxygen functionality. In some
embodiments, an acyloxy group is a C1-C4 acyloxy radical which refers to the
total number of chain or ring atoms of
the alkyl, alkenyl, alkynyl, aryl, cyclohexyl, heteroaryl or heterocycloalkyl
portion of the acyloxy group plus the
carbonyl carbon of acyl, i.e., a C4-acyloxy has three other ring or chain
atoms plus carbonyl. If the R radical is
heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute to
the total number of chain or ring atoms.
Unless stated otherwise in the specification, the "R" of an acyloxy group is
optionally substituted by one or more
substituents which independently include: acyl, alkyl, alkenyl, alkynyl,
alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl,
heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy,
haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate,
silyl, suffinyl, sulfonyl, sulfonamidyl,
sulfoxyl, sulfonate, urea, -Si(Ra)3-, -OR', -SRa, -0C(0)4Ra, -N(Ra)2, -C(0)1e,
-C(0)01ta, -0C(0)N(Ra)2,
-C(0)N(Ra)2, -N(Ra)C(0)01ta, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2,
N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tle (where t is 1
or 2), -S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or -0-
P(=0)(01ta)2 where each Ita is
independently hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl,
aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl and each of these
moieties can be optionally substituted as
defined herein.
[00123] "Amino" or "amine" refers to a -N(Rb)2, -N(Rb)Rb-, or -
RbN(Rb)Rb- radical group, where
each Rb is independently selected from hydrogen, alkyl, alkenyl, alkynyl,
haloalkyl, heteroalkyl (bonded through a
chain carbon), cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl
(bonded through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring carbon) or
heteroarylalkyl, unless stated otherwise in the
specification, each of which moiety can itself be optionally substituted as
described herein. When a -N(Rb)2 group
has two Rb other than hydrogen, they can be combined with the nitrogen atom to
form a 3-, 4-, 5-, 6-, or 7-
membered ring. For example, -N(Rb)2 is meant to include, but not be limited
to, 1-pyrrolidinyl and 4-morpholinyl.
Unless stated otherwise in the specification, an amino group is optionally
substituted by one or more substituents
which independently include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy,
amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl,
heteroalkyl, heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy, haloalkyl, ester, ether,
mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl,
sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -SRa, -0C(0)-Ra, -N(Ra)2, -C(0)Ra, -
C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2,
-N(Ra)C(0)01ta, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -
N(Ra)S(0)tRa (where t is 1 or 2),
-S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or -0-
P(=0)(01ta)2 where each Ita is independently
hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl,
heteroaryl or heteroarylalkyl, and each of these moieties can be optionally
substituted as defined herein.
[00124] The terms "amine" and "amino" also refer to N-oxides of the
groups
and -1\1'(Ita)(Ra)0-, Ita as described above, where the N-oxide is bonded to
the parent molecular structure through
34

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
the N atom. N-oxides can be prepared by treatment of the corresponding amino
group with, for example, hydrogen
peroxide or m-chloroperoxybenzoic acid. The person skilled in the art is
familiar with reaction conditions for
carrying out the N-oxidation.
[00125]
"Amide" or "amido" refers to a chemical moiety with formula ¨C(0)N(Rb)2 or
¨NRbC(0)Rb,
where Rb is independently selected from hydrogen, alkyl, alkenyl, alkynyl,
haloalkyl, heteroalkyl (bonded through a
chain carbon), cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl
(bonded through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring carbon) or
heteroarylalkyl, unless stated otherwise in the
specification, each of which moiety can itself be optionally substituted as
described herein. In some embodiments,
this radical is a C1-C4 amido or amide radical, which includes the amide
carbonyl in the total number of carbons in
the radical. When a ¨C(0)N(Rb)2 has two Rb other than hydrogen, they can be
combined with the nitrogen atom to
form a 3-, 4-, 5-, 6-, or 7-membered ring. For example, N(Rb)2 portion of a
¨C(0)N(Rb)2 radical is meant to
include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. Unless
stated otherwise in the specification, an
amido Rb group is optionally substituted by one or more substituents which
independently include: acyl, alkyl,
alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl, aryloxy,
amino, amido, amidino, imino, azide,
carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo,
haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate,
phosphonate, phosphinate, silyl, suffinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -SRa,
- OC (0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2, -
N(Ra)C(0)01e, -N(Ra)C(0)Ita, -
N(Ra)C(0)N(Ra)2, N(Ra)C(Nle)N(Ra)2, -N(Ra)S(0),Ra (where t is 1 or 2), -
S(0),ORa (where t is 1 or 2),
-S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-P(=0)(01ta)2 where each Ita is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl,
and each of these moieties can be optionally substituted as defined herein.
[00126]
The term "amide" or "amido" is inclusive of an amino acid or a peptide
molecule. Any amine,
hydroxy, or carboxyl side chain on the compounds described herein can be
transformed into an amide group. The
procedures and specific groups to make such amides are known to those of skill
in the art and can readily be found
in reference sources such as Greene and Wuts, Protective Groups in Organic
Synthesis, 3rd Ed., John Wiley & Sons,
New York, NY, 1999, which is incorporated herein by reference in its entirety.
[00127]
"Amidino" refers to both the ¨C(=NRb)N(Rb)2 and ¨N(Rb)-C(=NRb)- radicals,
where each Rb
is independently selected from hydrogen, alkyl, alkenyl, alkynyl, haloalkyl,
heteroalkyl (bonded through a chain
carbon), cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded
through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring carbon) or
heteroarylalkyl, unless stated otherwise in the
specification, each of which moiety can itself be optionally substituted as
described herein.
[00128]
"Aromatic" or "aryl" refers to a radical with six to ten ring atoms (e.g., C6-
C10 aromatic or C6-
C10 aryl) which has at least one ring having a conjugated pi electron system
which is carbocyclic (e.g., phenyl,
fluorenyl, and naphthyl). For example, bivalent radicals formed from
substituted benzene derivatives and having the
free valences at ring atoms are named as substituted phenylene radicals. In
other embodiments, bivalent radicals
derived from univalent polycyclic hydrocarbon radicals whose names end in "-
yl" by removal of one hydrogen atom

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
from the carbon atom with the free valence are named by adding "-idene" to the
name of the corresponding
univalent radical, e.g., a naphthyl group with two points of attachment is
termed naphthylidene. Whenever it
appears herein, a numerical range such as "6 to 10 aryl" refers to each
integer in the given range; e.g., "6 to 10 ring
atoms" means that the aryl group can consist of 6 ring atoms, 7 ring atoms,
etc., up to and including 10 ring atoms.
The term includes monocyclic or fused-ring polycyclic (i.e., rings which share
adjacent pairs of ring atoms) groups.
Unless stated otherwise in the specification, an aryl moiety can be optionally
substituted by one or more substituents
which independently include: acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl,
cycloalkyl, aralkyl, aryl, aryloxy,
amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl,
heteroalkyl, heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy, haloalkyl, ester, ether,
mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl,
sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -01ta,SRa,-0C(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)0Ra, -
0C(0)N(Ra)2, -C(0)N(Ra)2,
-N(Ra)C(0)0Ra, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -
N(Ra)S(0)tRa (where t is 1 or 2),
-S(0)tORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-
P(=0)(01e)2 where each Ra is independently
hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl,
heteroaryl or heteroarylalkyl, and each of these moieties can be optionally
substituted as defined herein.
[00129]
"Aralkyl" or "arylalkyl" refers to an (aryl)alkyl- radical where aryl and
alkyl are as disclosed
herein and which are optionally substituted by one or more of the substituents
described as suitable substituents for
aryl and alkyl respectively. The "aralkyl/arylalkyl" is bonded to the parent
molecular structure through the alkyl
group.
The terms "aralkenyl/arylalkenyl" and "aralkynyl/arylalkynyl" mirror the above
description of
"aralkyl/arylalkyl" wherein the "alkyl" is replaced with "alkenyl" or
"alkynyl" respectively, and the "alkenyl" or
"alkynyl" terms are as described herein.
[00130] "Azide" refers to a ¨N3 radical.
[00131] "Carbamate" refers to any of the following radicals: ¨0-(C=0)-
N(Rb)-, -0-(C=0)-N(Rb)2, ¨
N(Rb)-(C=0)-0-, and ¨N(Rb)-(C=0)-OR', wherein each Rb is independnently
selected from alkyl, alkenyl, alkynyl,
haloalkyl, heteroalkyl (bonded through a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl
(bonded through a ring carbon), heterocycloalkylalkyl, heteroaryl (bonded
through a ring carbon) or heteroarylalkyl,
unless stated otherwise in the specification, each of which moiety can itself
be optionally substituted as described
herein.
[00132] "Carbonate" refers to a ¨0-(C=0)-0- radical.
[00133] "Carbonyl" refers to a ¨(C=0)- radical.
[00134] "Carboxaldehyde" refers to a ¨(C=0)H radical.
[00135] "Carboxyl" refers to a ¨(C=0)0H radical.
[00136] "Cyano" refers to a ¨CN radical.
[00137] "Cycloalkyl" and "carbocycly1" each refer to a monocyclic or
polycyclic radical that contains
only carbon and hydrogen, and can be saturated or partially unsaturated.
Partially unsaturated cycloalkyl groups can
be termed "cycloalkenyl" if the carbocycle contains at least one double bond,
or "cycloalkynyl" if the carbocycle
contains at least one triple bond. Cycloalkyl groups include groups having
from 3 to 10 ring atoms (i.e., C3-C10
36

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
cycloalkyl). Whenever it appears herein, a numerical range such as "3 to 10"
refers to each integer in the given
range; e.g., "3 to 10 carbon atoms" means that the cycloalkyl group can
consist of 3 carbon atoms, 4 carbon atoms, 5
carbon atoms, etc., up to and including 10 carbon atoms. The term "cycloalkyl"
also includes bridged and spiro-
fused cyclic structures containing no heteroatoms. The term also includes
monocyclic or fused-ring polycyclic (i.e.,
rings which share adjacent pairs of ring atoms) groups. In some embodiments,
it is a C3-C8 cycloalkyl radical. In
some embodiments, it is a C3-05 cycloalkyl radical. Illustrative examples of
cycloalkyl groups include, but are not
limited to the following moieties: C3_6 carbocyclyl groups include, without
limitation, cyclopropyl (C3), cyclobutyl
(C4), cyclopentyl (C5), cyclopentenyl (C5), cyclohexyl (C6), cyclohexenyl
(C6), cyclohexadienyl (C6) and the like.
Examples of C3_8 carbocyclyl groups include the aforementioned C3_6
carbocyclyl groups as well as cycloheptyl
(C7), cycloheptadienyl (C7), cycloheptatrienyl (C7), cyclooctyl (C8),
bicyclo[2.2.1]heptanyl, bicyclo[2.2.2]octanyl,
and the like. Examples of C3_10 carbocyclyl groups include the aforementioned
C3_8 carbocyclyl groups as well as
octahydro-1H¨indenyl, decahydronaphthalenyl, spiro[4.5]decanyl and the like.
Unless stated otherwise in the
specification, a cycloalkyl group is optionally substituted by one or more
substituents which independently include:
acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, amidino, imino,
azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano,
halo, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate,
phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -SRa,
- OC (0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2, -
N(Ra)C(0)01e, -N(Ra)C(0)Ra, -
N(Ra)C(0)N(Ra)2, N(Ra)C(Nle)N(Ra)2, -N(Ra)S(0),Ra (where t is 1 or 2), -
S(0),ORa (where t is 1 or 2),
-S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-P(=0)(01e)2 where each It is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl,
and each of these moieties can be optionally substituted as defined herein.
[00138] "Cycloalkyl-alkyl" refers to a ¨(cycloalkyl)alkyl radical where
cycloalkyl and alkyl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for cycloalkyl and alkyl respectively. The "cycloalkyl-alkyl" is
bonded to the parent molecular structure
through the cycloalkyl group. The terms "cycloalkyl-alkenyl" and "cycloalkyl-
alkynyl" mirror the above
description of "cycloalkyl-alkyl" wherein the term "alkyl" is replaced with
"alkenyl" or "alkynyl" respectively, and
"alkenyl" or "alkynyl" are as described herein.
[00139] "Cycloalkyl-heterocycloalkyl" refers to a
¨(cycloalkyl)heterocycylalkyl radical where
cycloalkyl and heterocycloalkyl are as disclosed herein and which are
optionally substituted by one or more of the
substituents described as suitable substituents for heterocycloalkyl and
cycloalkyl respectively. The "cycloalkyl-
heterocycloalkyl" is bonded to the parent molecular structure through the
cycloalkyl group.
[00140] "Cycloalkyl-heteroaryl" refers to a ¨(cycloalkyl)heteroaryl
radical where cycloalkyl and
heteroaryl are as disclosed herein and which are optionally substituted by one
or more of the substituents described
as suitable substituents for heteroaryl and cycloalkyl respectively. The
"cycloalkyl-heteroaryl" is bonded to the
parent molecular structure through the cycloalkyl group.
[00141] As used herein, a "covalent bond" or "direct bond" refers to a
single bond joining two groups.
37

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00142] "Ester" refers to a radical of formula ¨COOR, where R is selected
from alkyl, alkenyl, alkynyl,
haloalkyl, heteroalkyl (bonded through a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl
(bonded through a ring carbon), heterocycloalkylalkyl, heteroaryl (bonded
through a ring carbon) or heteroarylalkyl.
Any amine, hydroxy, or carboxyl side chain on the compounds described herein
can be esterified. The procedures
and specific groups to make such esters are known to those of skill in the art
and can readily be found in reference
sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3rd
Ed., John Wiley & Sons, New York,
NY, 1999, which is incorporated herein by reference in its entirety. Unless
stated otherwise in the specification, an
ester group can be optionally substituted by one or more substituents which
independently include: acyl, alkyl,
alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl, aryloxy,
amino, amido, amidino, imino, azide,
carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl,
heterocycloalkyl, hydroxy, cyano, halo,
haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio,
thiocarbonyl, nitro, oxo, phosphate,
phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -SRa,
-0C(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)01ta, -0C(0)N(Ra)2, -C(0)N(Ra)2, -
N(Ra)C(0)01e, -N(Ra)C(0)Ita, -
N(Ra)C(0)N(Ra)2, N(Ra)C(Nita)N(Ra)2, -N(Ra)S(0),Ra (where t is 1 or 2), -
S(0),ORa (where t is 1 or 2),
-S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-P(=0)(01e)2 where each It is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl,
and each of these moieties can be optionally substituted as defined herein.
[00143] "Ether" refers to a ¨R'-O-R'- radical where each Rb is
independently selected from hydrogen,
alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through a chain
carbon), cycloalkyl, cycloalkylalkyl, aryl,
aralkyl, heterocycloalkyl (bonded through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring
carbon) or heteroarylalkyl, unless stated otherwise in the specification, each
of which moiety can itself be optionally
substituted as described herein.
[00144] "Halo", "halide", or, alternatively, "halogen" means fluoro,
chloro, bromo or iodo. The terms
"haloalkyl," "haloalkenyl," "haloalkynyl" and "haloalkoxy" include alkyl,
alkenyl, alkynyl and alkoxy structures
that are substituted with one or more halo groups or with combinations
thereof. For example, the terms "fluoroalkyl"
and "fluoroalkoxy" include haloalkyl and haloalkoxy groups, respectively, in
which the halo is fluorine, such as, but
not limited to, trifluoromethyl, difluoromethyl, 2,2,2-trifluoroethyl, 1-
fluoromethy1-2-fluoroethyl, and the like. Each
of the alkyl, alkenyl, alkynyl and alkoxy groups are as defined herein and can
be optionally further substituted as
defined herein.
[00145] "Heteroalkyl", "heteroalkenyl" and "heteroalkynyl" include alkyl,
alkenyl and alkynyl radicals,
respectively, which have one or more skeletal chain atoms selected from an
atom other than carbon, e.g., oxygen,
nitrogen, sulfur, phosphorus or combinations thereof. A numerical range can be
given, e.g., C1-C4 heteroalkyl which
refers to the chain length in total, which in this example is 4 atoms long.
For example, a ¨CH2OCH2CH3 radical is
referred to as a "C4" heteroalkyl, which includes the heteroatom center in the
atom chain length description.
Connection to the parent molecular strucuture can be through either a
heteroatom or a carbon in the heteroalkyl
chain. For example, an N-containing heteroalkyl moiety refers to a group in
which at least one of the skeletal atoms
is a nitrogen atom. One or more heteroatom(s) in the heteroalkyl radical can
be optionally oxidized. One or more
38

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
nitrogen atoms, if present, can also be optionally quaternized. For example,
heteroalkyl also includes skeletal chains
substituted with one or more nitrogen oxide (-0-) substituents. Exemplary
heteroalkyl groups include, without
limitation, ethers such as methoxyethanyl (¨CH2CH20CH3), ethoxymethanyl
(¨CH20CH2CH3),
(methoxymethoxy)ethanyl (¨CH2CH20CH20CH3), (methoxymethoxy)methanyl
(¨CH20CH20CH3) and
(methoxyethoxy)methanyl (¨CH20CH2 CH20CH3) and the like; amines such as
¨CH2CH2NHCH3, ¨
CH2CH2N(CH3)2, ¨CH2NHCH2CH3, ¨CH2N(CH2CH3)(CH3) and the like. Heteroalkyl,
heteroalkenyl, and
heteroalkynyl groups can each be optionally substituted by one or more
substituents which independently include:
acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, amidino, imino,
azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano,
halo, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate,
phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(Ra)3-, -OR', -SRa,
-0C(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)0Ra, -0C(0)N(Ra)2, -C(0)N(Ra)2, -
N(Ra)C(0)0Ra, -N(Ra)C(0)Ra, -
N(Ra)C(0)N(Ra)2, N(Ra)C(NR')N(Ra)2, -N(Ra)S(0),Ra (where t is 1 or 2), -
S(0),0Ra (where t is 1 or 2),
-S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-P(=0)(0Ra)2 where each It is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl,
and each of these moieties can be optionally substituted as defined herein.
[00146]
"Heteroalkyl-aryl" refers to a -(heteroalkyl)aryl radical where heteroalkyl
and aryl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for heteroalkyl and aryl respectively. The "heteroalkyl-aryl" is
bonded to the parent molecular structure
through an atom of the heteroalkyl group.
[00147]
"Heteroalkyl-heteroaryl" refers to a -(heteroalkyl)heteroaryl radical where
heteroalkyl and
heteroaryl are as disclosed herein and which are optionally substituted by one
or more of the substituents described
as suitable substituents for heteroalkyl and heteroaryl respectively. The
"heteroalkyl-heteroaryl" is bonded to the
parent molecular structure through an atom of the heteroalkyl group.
[00148]
"Heteroalkyl-heterocycloalkyl" refers to a -(heteroalkyl)heterocycloalkyl
radical where
heteroalkyl and heterocycloalkyl are as disclosed herein and which are
optionally substituted by one or more of the
substituents described as suitable substituents for heteroalkyl and
heterocycloalkyl respectively. The "heteroalkyl-
heterocycloalkyl" is bonded to the parent molecular structure through an atom
of the heteroalkyl group.
[00149]
"Heteroalkyl-cycloalkyl" refers to a -(heteroalkyl)cycloalkyl radical where
heteroalkyl and
cycloalkyl are as disclosed herein and which are optionally substituted by one
or more of the substituents described
as suitable substituents for heteroalkyl and cycloalkyl respectively. The
"heteroalkyl-cycloalkyl" is bonded to the
parent molecular structure through an atom of the heteroalkyl group.
[00150]
"Heteroaryl" or, alternatively, "heteroaromatic" refers to a refers to a
radical of a 5-18
membered monocyclic or polycyclic (e.g., bicyclic or tricyclic) aromatic ring
system (e.g., having 6, 10 or 14
electrons shared in a cyclic array) having ring carbon atoms and 1-6 ring
heteroatoms provided in the aromatic ring
system, wherein each heteroatom is independently selected from nitrogen,
oxygen, phosphorous and sulfur ("5-18
membered heteroaryl"). Heteroaryl polycyclic ring systems can include one or
more heteroatoms in one or both
39

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
rings. Whenever it appears herein, a numerical range such as "5 to 18" refers
to each integer in the given range; e.g.,
"5 to 18 ring atoms" means that the heteroaryl group can consist of 5 ring
atoms, 6 ring atoms, etc., up to and
including 18 ring atoms. For example, bivalent radicals derived from univalent
heteroaryl radicals whose names end
in "-yl" by removal of one hydrogen atom from the atom with the free valence
are named by adding "-idene" to the
name of the corresponding univalent radical, e.g., a pyridyl group with two
points of attachment is a pyridylidene.
[00151]
For example, an N-containing "heteroaromatic" or "heteroaryl" moiety refers to
an aromatic
group in which at least one of the skeletal atoms of the ring is a nitrogen
atom. One or more heteroatom(s) in the
heteroaryl radical can be optionally oxidized. One or more nitrogen atoms, if
present, can also be optionally
quaternized. Heteroaryl also includes ring systems substituted with one or
more nitrogen oxide (-0-) substituents,
such as pyridinyl N-oxides. The heteroaryl is attached to the parent molecular
structure through any atom of the
ring(s).
[00152]
"Heteroaryl" also includes ring systems wherein the heteroaryl ring, as
defined above, is fused
with one or more aryl groups wherein the point of attachment to the parent
molecular structure is either on the aryl
or on the heteroaryl ring, or wherein the heteroaryl ring, as defined above,
is fused with one or more cycloalkyl or
heterocycyl groups wherein the point of attachment to the parent molecular
structure is on the heteroaryl ring. For
polycyclic heteroaryl groups wherein one ring does not contain a heteroatom
(e.g., indolyl, quinolinyl, carbazolyl
and the like), the point of attachment to the parent molecular structure can
be on either ring, i.e., either the ring
bearing a heteroatom (e.g., 2¨indoly1) or the ring that does not contain a
heteroatom (e.g., 5¨indoly1). In some
embodiments, a heteroaryl group is a 5-10 membered aromatic ring system having
ring carbon atoms and 1-4 ring
heteroatoms provided in the aromatic ring system, wherein each heteroatom is
independently selected from nitrogen,
oxygen, phosphorous, and sulfur ("5-10 membered heteroaryl"). In some
embodiments, a heteroaryl group is a 5-8
membered aromatic ring system having ring carbon atoms and 1-4 ring
heteroatoms provided in the aromatic ring
system, wherein each heteroatom is independently selected from nitrogen,
oxygen, phosphorous, and sulfur ("5-8
membered heteroaryl"). In some embodiments, a heteroaryl group is a 5-6
membered aromatic ring system having
ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring
system, wherein each heteroatom is
independently selected from nitrogen, oxygen, phosphorous, and sulfur ("5-6
membered heteroaryl"). In some
embodiments, the 5-6 membered heteroaryl has 1-3 ring heteroatoms selected
from nitrogen, oxygen, phosphorous,
and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1-2 ring
heteroatoms selected from nitrogen,
oxygen, phosphorous, and sulfur. In some embodiments, the 5-6 membered
heteroaryl has 1 ring heteroatom
selected from nitrogen, oxygen, phosphorous, and sulfur.
[00153]
Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl,
benzimidazolyl,
benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl,
benzo[d]thiazolyl, benzothiadiazolyl,
benzo [b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl,
1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl,
benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl,
benzofuranyl, benzofuranonyl,
benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl),
benzothieno[3,2-d]pyrimidinyl, benzotriazolyl,
benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl,
cinnolinyl, cyclopenta[d]pyrimidinyl,
6,7-dihydro-5H- cyclopenta[4,5] thieno [2,3 -d] pyrimidinyl,
5,6-dihydrobenzo[h] quinazolinyl,

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
5,6-dihydrobenzo[h] cinnolinyl,
6,7-dihydro-5H-benzo[6,7] cyclohepta[1,2-c]pyridazinyl, dibenzofuranyl,
dibenzothiophenyl, furanyl, furazanyl, furanonyl,
furo[3,2-c]pyridinyl,
5,6,7, 8,9,10-hexahydrocycloocta[d] pyrimidinyl,
5,6,7, 8,9,10-hexahydrocycloocta[d] pyridazinyl,
5,6,7,8,9,10-hexahydrocycloocta[d]pyridinyl,isothiazolyl, imidazolyl,
indazolyl, indolyl, indazolyl, isoindolyl,
indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-
5,6,7,8-tetrahydroquinazolinyl,
naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl,
oxazolyl, oxiranyl,
5,6,6a,7, 8,9,10,10 a-octahydrobenzo[h] quinazolinyl, 1 -pheny1-1H-
pyrrolyl, phenazinyl, phenothiazinyl,
phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrrolyl, pyrazolyl,
pyrazolo[3,4-d]pyrimidinyl, pyridinyl,
pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl,
pyridazinyl, pyrrolyl, quinazolinyl,
quinoxalinyl, quinolinyl, isoquinolinyl,
tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl,
5,6,7, 8-tetrahydrobenzo[4,5] thieno [2,3 -d] pyrimidinyl,
6,7,8,9 -tetrahydro-5H - cyclohepta[4,5] thieno [2,3 -d]pyrimidinyl,
5,6,7,8-tetrahydropyrido[4,5-c]pyridazinyl,
thiazolyl, thiadiazolyl, thiapyranyl,
triazolyl, tetrazolyl, triazinyl, thieno[2,3-d]pyrimidinyl,
thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pridinyl, and thiophenyl (i.e.,
thienyl). Unless stated otherwise in the
specification, a heteroaryl moiety is optionally substituted by one or more
substituents which independently include:
acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, amidino, imino,
azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano,
halo, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate,
phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamidyl, sulfoxyl,
sulfonate, urea, -Si(le)3, -01ta,
-0C(0)-Ita, -N(Ita)2, -C(0)Ita, -C(0)0Ita, -0C(0)N(Ita)2, -C(0)N(Ita)2, -
N(Ita)C(0)01e, -N(Ita)C(0)Ita, -
N(Ita)C(0)N(Ita)2, N(Ita)C(Nle)N(Ra)2, -N(Ita)S(0),Ita (where t is 1 or 2), -
S(0),ORa (where t is 1 or 2),
-S(0)tN(Ita)2 (where t is 1 or 2), or ¨0-P(=0)(01e)2 where each Ita is
independently hydrogen, alkyl, haloalkyl,
carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl
and each of these moieties can be optionally substituted as defined herein.
[00154]
"Heteroaryl-alkyl" refers to a -(heteroaryl)alkyl radical where heteroaryl and
alkyl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for heteroaryl and alkyl respectively. The "heteroaryl-alkyl" is
bonded to the parent molecular structure
through any atom of the heteroaryl group.
[00155]
"Heteroaryl-heterocycloalkyl" refers to an -(heteroaryl)heterocycloalkyl
radical where
heteroaryl and heterocycloalkyl are as disclosed herein and which are
optionally substituted by one or more of the
substituents described as suitable substituents for heteroaryl and
heterocycloalkyl respectively. The "heteroaryl-
heterocycloalkyl" is bonded to the parent molecular structure through an atom
of the heteroaryl group.
[00156]
"Heteroaryl-cycloalkyl" refers to an -(heteroaryl)cycloalkyl radical where
heteroaryl and
cycloalkyl are as disclosed herein and which are optionally substituted by one
or more of the substituents described
as suitable substituents for heteroaryl and cycloalkyl respectively. The
"heteroaryl-cycloalkyl" is bonded to the
parent molecular structure through a carbon atom of the heteroaryl group.
41

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00157]
"Heterocyclyl", "heterocycloalkyl" or `heterocarbocycly1" each refer to any 3-
to
18-membered non-aromatic radical monocyclic or polycyclic moiety comprising at
least one heteroatom selected
from nitrogen, oxygen, phosphorous and sulfur. A heterocyclyl group can be a
monocyclic, bicyclic, tricyclic or
tetracyclic ring system, wherein the polycyclic ring systems can be a fused,
bridged or spiro ring system.
Heterocyclyl polycyclic ring systems can include one or more heteroatoms in
one or both rings. A heterocyclyl
group can be saturated or partially unsaturated. Partially unsaturated
heterocycloalkyl groups can be termed
"heterocycloalkenyl" if the heterocyclyl contains at least one double bond, or
"heterocycloalkynyl" if the
heterocyclyl contains at least one triple bond. Whenever it appears herein, a
numerical range such as "5 to 18"
refers to each integer in the given range; e.g., "5 to 18 ring atoms" means
that the heterocyclyl group can consist of 5
ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms. For
example, bivalent radicals derived from
univalent heterocyclyl radicals whose names end in "-y1" by removal of one
hydrogen atom from the atom with the
free valence are named by adding "-idene" to the name of the corresponding
univalent radical, e.g., a piperidine
group with two points of attachment is a piperidylidene.
[00158]
An N-containing heterocyclyl moiety refers to an non-aromatic group in which
at least one of
the ring atoms is a nitrogen atom. The heteroatom(s) in the heterocyclyl
radical can be optionally oxidized. One or
more nitrogen atoms, if present, can be optionally quaternized. Heterocyclyl
also includes ring systems substituted
with one or more nitrogen oxide (-0-) substituents, such as piperidinyl N-
oxides. The heterocyclyl is attached to the
parent molecular structure through any atom of any of the ring(s).
[00159]
"Heterocyclyl" also includes ring systems wherein the heterocycyl ring, as
defined above, is
fused with one or more carbocycyl groups wherein the point of attachment is
either on the carbocycyl or
heterocyclyl ring, or ring systems wherein the heterocyclyl ring, as defined
above, is fused with one or more aryl or
heteroaryl groups, wherein the point of attachment to the parent molecular
structure is on the heterocyclyl ring. In
some embodiments, a heterocyclyl group is a 3-10 membered non¨aromatic ring
system having ring carbon atoms
and 1-4 ring heteroatoms, wherein each heteroatom is independently selected
from nitrogen, oxygen, phosphorous
and sulfur ("3-10 membered heterocyclyl"). In some embodiments, a heterocyclyl
group is a 5-8 membered non¨
aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms,
wherein each heteroatom is independently
selected from nitrogen, oxygen, phosphorous and sulfur ("5-8 membered
heterocyclyl"). In some embodiments, a
heterocyclyl group is a 5-6 membered non¨aromatic ring system having ring
carbon atoms and 1-4 ring
heteroatoms, wherein each heteroatom is independently selected from nitrogen,
oxygen, phosphorous and sulfur
("5-6 membered heterocyclyl"). In some embodiments, the 5-6 membered
heterocyclyl has 1-3 ring heteroatoms
selected from nitrogen, oxygen phosphorous and sulfur. In some embodiments,
the 5-6 membered heterocyclyl has
1-2 ring heteroatoms selected from nitrogen, oxygen, phosphorous and sulfur.
In some embodiments, the 5-6
membered heterocyclyl has 1 ring heteroatom selected from nitrogen, oxygen,
phosphorous and sulfur.
[00160]
Exemplary 3¨membered heterocyclyls containing 1 heteroatom include, without
limitation,
azirdinyl, oxiranyl, thiorenyl. Exemplary 4¨membered heterocyclyls containing
1 heteroatom include, without
limitation, azetidinyl, oxetanyl and thietanyl. Exemplary 5¨membered
heterocyclyls containing 1 heteroatom
include, without limitation, tetrahydrofuranyl, dihydrofuranyl,
tetrahydrothiophenyl, dihydrothiophenyl,
42

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
pyrrolidinyl, dihydropyrrolyl and pyrroly1-2,5¨dione. Exemplary 5¨membered
heterocyclyls containing 2
heteroatoms include, without limitation, dioxolanyl, oxathiolanyl and
dithiolanyl. Exemplary 5¨membered
heterocyclyls containing 3 heteroatoms include, without limitation,
triazolinyl, oxadiazolinyl, and thiadiazolinyl.
Exemplary 6¨membered heterocyclyl groups containing 1 heteroatom include,
without limitation, piperidinyl,
tetrahydropyranyl, dihydropyridinyl, and thianyl. Exemplary 6¨membered
heterocyclyl groups containing 2
heteroatoms include, without limitation, piperazinyl, morpholinyl, dithianyl,
dioxanyl, and triazinanyl. Exemplary
7¨membered heterocyclyl groups containing 1 heteroatom include, without
limitation, azepanyl, oxepanyl and
thiepanyl. Exemplary 8¨membered heterocyclyl groups containing 1 heteroatom
include, without limitation,
azocanyl, oxecanyl and thiocanyl. Exemplary bicyclic heterocyclyl groups
include, without limitation, indolinyl,
isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl,
tetrahydrobenzothienyl, tetrahydrobenzofuranyl,
tetrahydroindolyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl,
decahydroquinolinyl, decahydroisoquinolinyl,
octahydrochromenyl, octahydroisochromenyl, decahydronaphthyridinyl, decahydro-
1,8¨naphthyridinyl,
octahydropyrrolo[3,2¨b]pyrrole, indolinyl, phthalimidyl, naphthalimidyl,
chromanyl, chromenyl, 1H¨
benzo[e] [1,4] diazepinyl,
1,4,5,7¨tetrahydropyrano [3 ,4¨b]pyrrolyl, 5,6¨dihydro-4H¨furo [3
,2¨b]pyrrolyl, 6,7¨
dihydro-5H¨furo[3 ,2¨b] pyranyl, 5,7¨dihydro-4H¨thieno [2,3¨c ]pyranyl,
2,3¨dihydro-1H¨pyrrolo[2,3¨b]pyridinyl,
2,3¨dihydrofuro[2,3¨b]pyridinyl,
4,5,6,7¨tetrahydro-1H¨pyrrolo[2,3¨b]pyridinyl, 4,5,6,7¨tetrahydrofuro [3
,2¨
c]pyridinyl, 4,5,6,7¨tetrahydrothieno[3,2¨b]pyridinyl, 1,2,3,4¨tetrahydro-
1,6¨naphthyridinyl, and the like.
[00161]
Unless stated otherwise, heterocyclyl moieties are optionally substituted by
one or more
substituents which independently include: acyl, alkyl, alkenyl, alkynyl,
alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl,
heteroalkyl, heteroaryl,
heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halo, haloalkoxy,
haloalkyl, ester, ether, mercapto, thio, alkylthio,
arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate,
silyl, sulfinyl, sulfonyl, sulfonamidyl,
sulfoxyl, sulfonate, urea, -Si(Ra)3-, -OR', -SRa, -0C(0)-Ra, -N(Ra)2, -C(0)Ra,
-C(0)01ta, -0C(0)N(Ra)2,
-C(0)N(Ra)2, -N(Ra)C(0)OR', -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2,
N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0),Ra (where t is 1
or 2), -S(0)tOlta (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or ¨0-
P(=0)(01ta)2 where each Ita is
independently hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl,
aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl and each of these
moieties can be optionally substituted as
defined herein.
[00162]
"Heterocyclyl-alkyl" refers to a -(heterocyclyl)alkyl radical where
heterocyclyl and alkyl are as
disclosed herein and which are optionally substituted by one or more of the
substituents described as suitable
substituents for heterocyclyl and alkyl respectively. The "heterocyclyl-alkyl"
is bonded to the parent molecular
structure through any atom of the heterocyclyl group. The terms "heterocyclyl-
alkenyl" and "heterocyclyl-alkynyl"
mirror the above description of "heterocyclyl-alkyl" wherein the term "alkyl"
is replaced with "alkenyl" or
"alkynyl" respectively, and "alkenyl" or "alkynyl" are as described herein.
[00163]
"Imino" refers to the "-(C=N)-R" radical where Rb is selected from hydrogen,
alkyl, alkenyl,
alkynyl, haloalkyl, heteroalkyl (bonded through a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl,
heterocycloalkyl (bonded through a ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon)
43

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
or heteroarylalkyl, unless stated otherwise in the specification, each of
which moiety can itself be optionally
substituted as described herein.
[00164] "Moiety" refers to a specific segment or functional group of a
molecule. Chemical moieties are
often recognized chemical entities embedded in or appended to a molecule.
[00165] "Nitro" refers to the ¨NO2 radical.
[00166] "Oxa" refers to the -0- radical.
[00167] "Oxo" refers to the =0 radical.
[00168] "Phosphate" refers to a ¨0-P(=0)(0Rb)2 radical, where each Rb is
independently selected from
hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through a
chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded through a ring
carbon), heterocycloalkylalkyl, heteroaryl
(bonded through a ring carbon) or heteroarylalkyl, unless stated otherwise in
the specification, each of which moiety
can itself be optionally substituted as described herein. In some embodiments,
when le is hydrogen and depending
on the pH, the hydrogen can be replaced by an appropriately charged counter
ion.
[00169] "Phosphonate" refers to a ¨0-P(=0)(Rb)(0Rb) radical, where each
Rb is independently selected
from hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through
a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded through a ring
carbon), heterocycloalkylalkyl, heteroaryl
(bonded through a ring carbon) or heteroarylalkyl, unless stated otherwise in
the specification, each of which moiety
can itself be optionally substituted as described herein. In some embodiments,
when le is hydrogen and depending
on the pH, the hydrogen can be replaced by an appropriately charged counter
ion.
[00170] "Phosphinate" refers to a ¨P(=0)(Rb)(0Rb) radical, where each Rb
is independently selected
from hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through
a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded through a ring
carbon), heterocycloalkylalkyl, heteroaryl
(bonded through a ring carbon) or heteroarylalkyl, unless stated otherwise in
the specification, each of which moiety
can itself be optionally substituted as described herein. In some embodiments,
when Ita is hydrogen and depending
on the pH, the hydrogen can be replaced by an appropriately charged counter
ion.
[00171] A "leaving group or atom" is any group or atom that will, under
the reaction conditions, cleave
from the starting material, thus promoting reaction at a specified site.
Suitable non-limiting examples of such
groups unless otherwise specified include halogen atoms, mesyloxy, p-
nitrobenzensulphonyloxy,
trifluoromethyloxy, and tosyloxy groups.
[00172] "Protecting group" has the meaning conventionally associated with
it in organic synthesis, i.e.,
a group that selectively blocks one or more reactive sites in a
multifunctional compound such that a chemical
reaction can be carried out selectively on another unprotected reactive site
and such that the group can readily be
removed after the selective reaction is complete. A variety of protecting
groups are disclosed, for example, in T.H.
Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, Third
Edition, John Wiley & Sons, New York
(1999), incorporated herein by reference in its entirety. For example, a
hydroxy protected form is where at least one
of the hydroxy groups present in a compound is protected with a hydroxy
protecting group. Likewise, amines and
other reactive groups can similarly be protected.
44

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00173] As used herein, the terms "substituted" or "substitution" mean
that at least one hydrogen
present on a group atom (e.g., a carbon or nitrogen atom) is replaced with a
permissible substituent, e.g., a
substituent which upon substitution for the hydrogen results in a stable
compound, e.g., a compound which does not
spontaneously undergo transformation such as by rearrangement, cyclization,
elimination, or other reaction. Unless
otherwise indicated, a "substituted" group can have a substituent at one or
more substitutable positions of the group,
and when more than one position in any given structure is substituted, the
substituent is either the same or different
at each position. Substituents include one or more group(s) individually and
independently selected from acyl,
alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, aralkyl, aryl,
aryloxy, amino, amido, azide, carbonate,
carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy,
cyano, halo, haloalkoxy, haloalkyl,
ester, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo,
phosphate, phosphonate, phosphinate, silyl,
sulfonyl, sulfonamidyl, sulfoxyl, sulfonate, urea, -Si(Ra)3, -OR', -SRa, -
0C(0)4Ra, -N(Ra)2, -C(0)Ra,
-C(0)0Ra, -0C(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)0Ra, -N(Ra)C(0)Ra, -
N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2,
-N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -
S(0),1\1(Ra)2 (where t is 1 or
where each Ra is independently hydrogen, alkyl, haloalkyl, carbocyclyl,
carbocyclylalkyl, aryl, aralkyl,
heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl and
each of these moieties can be optionally
substituted as defined herein. For example, a cycloalkyl substituent can have
a halide substituted at one ormore ring
carbons, and the like. The protecting groups that can form the protective
derivatives of the above substituents are
known to those of skill in the art and can be found in references such as
Greene and Wuts, above.
[00174] "Sily1" refers to a ¨Si(Rb)3 radical where each Rb is
independently selected from alkyl, alkenyl,
alkynyl, haloalkyl, heteroalkyl (bonded through a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl,
heterocycloalkyl (bonded through a ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon)
or heteroarylalkyl, unless stated otherwise in the specification, each of
which moiety can itself be optionally
substituted as described herein.
[00175] "Sulfanyl", "sulfide", and "thio" each refer to the radical -S-
Rb, wherein Rb is selected from
alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through a chain
carbon), cycloalkyl, cycloalkylalkyl, aryl,
aralkyl, heterocycloalkyl (bonded through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring
carbon) or heteroarylalkyl, unless stated otherwise in the specification, each
of which moiety can itself be optionally
substituted as described herein. For instance, an "alkylthio" refers to the
"alkyl-S-" radical, and "arylthio" refers to
the "aryl-S-" radical, each of which are bound to the parent molecular group
through the S atom. The terms
"sulfide", "thiol", "mercapto", and "mercaptan" can also each refer to the
group ¨RbSH.
[00176] "Sulfinyl" or "sulfoxide" refers to the -S(0)-R' radical,
wherein for "sulfinyl", Rb is H and for
"sulfoxide", Rb is selected from alkyl, alkenyl, alkynyl, haloalkyl,
heteroalkyl (bonded through a chain carbon),
cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded through a
ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon) or heteroarylalkyl, unless stated
otherwise in the specification, each of
which moiety can itself be optionally substituted as described herein.
[00177] "Sulfonyl" or "sulfone" refers to the -S(02)-R' radical, wherein
Rb is selected from hydrogen,
alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through a chain
carbon), cycloalkyl, cycloalkylalkyl, aryl,

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
aralkyl, heterocycloalkyl (bonded through a ring carbon),
heterocycloalkylalkyl, heteroaryl (bonded through a ring
carbon) or heteroarylalkyl, unless stated otherwise in the specification, each
of which moiety can itself be optionally
substituted as described herein.
[00178] "Sulfonamidyl" or "sulfonamido" refers to the following
radicals: ¨S(=0)2-N(Rb)2, -N(Rb)-
S(=0)2-Rb, ¨S(=0)2-N(Rb)-, or -N(Rb)-S(=0)2-, where each Rb is independently
selected from hydrogen, alkyl,
alkenyl, alkynyl, haloalkyl, heteroalkyl (bonded through a chain carbon),
cycloalkyl, cycloalkylalkyl, aryl, aralkyl,
heterocycloalkyl (bonded through a ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon)
or heteroarylalkyl, unless stated otherwise in the specification, each of
which moiety can itself be optionally
substituted as described herein. The Rb groups in ¨S(=0)2-N(Rb)2 can be taken
together with the nitrogen to which
they are attached to form a 4-, 5-, 6-, or 7-membered heterocyclyl ring. In
some embodiments, the term designates a
C1-C4 sulfonamido, wherein each Rb in the sulfonamido contains 1 carbon, 2
carbons, 3 carbons, or 4 carbons total.
[00179] "Sulfoxyl" or "sulfoxide" refers to a ¨S(=0)20H radical.
[00180] "Sulfonate" refers to a ¨S(=0)2-OR' radical, wherein Rb is
selected from alkyl, alkenyl,
alkynyl, haloalkyl, heteroalkyl (bonded through a chain carbon), cycloalkyl,
cycloalkylalkyl, aryl, aralkyl,
heterocycloalkyl (bonded through a ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon)
or heteroarylalkyl, unless stated otherwise in the specification, each of
which moiety can itself be optionally
substituted as described herein.
[00181] "Thiocarbonyl" refers to a ¨(C=S)- radical.
[00182] "Urea" refers to a ¨N(Rb)-(C=0)-N(Rb)2 or ¨N(Rb)-(C=0)-N(Rb)-
radical, where each Rb is
independently selected from alkyl, alkenyl, alkynyl, haloalkyl, heteroalkyl
(bonded through a chain carbon),
cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl (bonded through a
ring carbon), heterocycloalkylalkyl,
heteroaryl (bonded through a ring carbon) or heteroarylalkyl, unless stated
otherwise in the specification, each of
which moiety can itself be optionally substituted as described herein.
[00183] Where substituent groups are specified by their conventional
chemical formulae, written from left
to right, they equally encompass the chemically identical substituents that
would result from writing the structure
from right to left, e.g., -CH20- is equivalent to -OCH2-=
Compounds
[00184] In one aspect, provided herein are compounds of Formula (I):
0
=
Wb5 X
Wd
Formula (I)
46

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
Wb5 is CR8, CHR8, or N,
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea or carbonate;
X is absent or is ¨(CH(R9)),-;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-, -N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when Wb5 is N, no more than one of X or Y is absent;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino,
acyl, acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
R13 N
X
I )1
R12N R1
Wd iS R11
wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and K-17
are independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, carboxylic
acid, oxo, or NR'R" wherein R' and R" are taken together with nitrogen to form
a cyclic moiety.
[00185] In certain embodiments, Wb5 is Cle. In some embodiments, R8 is
hydrogen.
[00186] In certain embodiments, Cy is aryl or heteroaryl substituted with
0-1 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is aryl or heteroaryl substituted
with 0 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is aryl (e.g., phenyl) substituted
with 0-1 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is phenyl substituted with 0-1
occurrences of R3 and 0-3 occurrences
of R5. In some embodiments, Cy is phenyl substituted with 0 occurrences of R3
and 0 occurrences of R5. In some
47

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
embodiments, Cy is phenyl substituted with 1 occurrence of R3 and 0
occurrences of R5. In some embodiments, R3
is alkyl (e.g., methyl). In some embodiments, R3 is halo (e.g., chloro or
fluoro). In some embodiments, R3 is
heteroaryl (e.g., 1-methy1-4-pyrazolyl, 2-methyl-5-pyrimidyl, 2-methoxy-5-
pyrimidyl, 2-amino-5-pyrimidyl, 2-
amino-5-pyridyl, 2,3-dimethy1-5-pyridyl, 3-methoxy-5-pyridyl, 5-pyrimidyl, 5-
methyl-3-pyridyl, 2-methy1-4-
pyridyl, 2-methyl-4-pyridazinyl, 2-methoxy-5-pyrimidyl or 1,3-dimethy1-4-
pyrazoly1).
[00187] In some embodiments, In some embodiments, Cy is phenyl
substituted with 1 occurrence of R3
and 1 occurrence of R5. In some embodiments, R3 is halo (e.g., chloro or
fluoro) and R5 is halo (e.g., chloro or
fluoro).
[00188] In certain embodiments, Cy is heteroaryl substituted with 0-1
occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is 5-membered heteroaryl (e.g.,
thiophenyl or isothiazolyl) substituted
with 0-1 occurrences of R3 and 0-3 occurrences of R5. In some embodiments, Cy
is thiophenyl substituted with 0
occurrences of R3 and 1 occurrence of R5. In some embodiments, Cy is
thiophenyl substituted with 0-1 occurrence
of R3 and 0 occurrence of R5. In some embodiments, Cy is thiophenyl
substituted with 1 occurrence of R3 and 0
occurrence of R5. In some embodiments, Cy is isothiazolyl substituted with 0
occurrences of R3 and 1 occurrence of
R5. In some embodiments, Cy is isothiazolyl substituted with 0-1 occurrence of
R3 and 0 occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 1 occurrence of R3 and 0
occurrence of R5. In some embodiments,
Cy is 6-membered heteroaryl (e.g., pyridinyl).
[00189] In certain embodiments, B is hydrogen.
[00190] In certain embodiments, B is aryl (e.g., 6-membered aryl)
substituted with 0-4 occurrences of
R2. In some embodiments, B is phenyl substituted ith 0-4 occurrences of R2. In
some embodiments, B is phenyl
substituted with 0 occurrences of R2. In some embodiments, B is phenyl
substituted with 1 occurrence of R2. In
some embodiments, R2 is halo (e.g., fluoro).
[00191] In certain embodiments, B is cycloalkyl (e.g., cyclopropyl)
substituted with 0-4 occurrences of
R2. In some embodiments, B is cyclopropyl substituted with 0-4 occurrences of
R2. In some embodiments, B is
cyclopropyl substituted with 0 occurrences of R2.
[00192] In certain embodiments, B is alkyl (e.g., methyl) substituted
with 0-4 occurrences of R2. In
some embodiments, B is alkyl (e.g., methyl) substituted with 0 occurrences of
R2.
[00193] In some embodiments, X is ¨(CH(R9)),-. In some embodiments, z is
1. In some embodiments,
R9 is Ci_io alkyl (e.g., methyl).
[00194] In some embodiments, is absent, -0-, -NH(R9)-, or -S(=0)2-. In
certain embodiments, Y is
absent. In some embodiments, Y is ¨N(R9)-. In some embodiments, R9 is
hydrogen.
;s5Y
HN õss
[00195] In certain embodiments, X-Y is . In
some embodiments, X-Y is -CH2-N(CH3). In
some embodiments, X-Y is (S)-CH(CH3)-NH-. In some embodiments, X-Y is (R)-
CH(CH3)-NH-.
[00196] In certain embodiments, X1 is N. In some embodiments, X1 is CR14.
In some embodiments,
R14 is hydrogen. In some embodiments, R14 is cyano.
48

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00197] In certan embodiments, R1 is hydrogen. In some embodiments, R1
is halo (e.g., chloro). In
some embodiments, R1 is amino. Non-limiting examples include alkylamino
groups, such as methylamino,
ethylamino, propylamino, isopropylamino, butylamino, isobutylamino,
pentylamino, isopentylamino, and
hexylamino. In some embodiments, R1 is methylamino. In some embodiments, R1
is benzylamino, including
optionally substituted benzylamino. In some embodiments, R1 is hydroxyl.
[00198] In certain embodiments, R11 is hydrogen. In some embodiments, R12
is hydrogen. In some
embodiments, R13 is hydrogen. In some embodiments, R11, R12 and R13
are hydrogen.
JVVIJ JVVV
I
[00199] In certain embodiments, Wd is N .
In some embodiments Wd is CI.
JVVV
NH2
In some embodiments, Wd is . In some embodiments, Wd is
avvy
JVVV
NN
1.1 NLNI-1\11e
e= In some embodiments, Wd is . In some
embodiments,
JVVV
N CN
Wd is N . In some embodiments, Wd is N . In some embodiments, Wd is
JVVV JVVV
N
N OHIn some embodiments, Wd iS NOMe
[00200] In some embodiments, Cy is aryl substituted with 0-1 occurrences
of R3 and 0-3 occurrences of
R5. For example, Cy is phenyl substituted with 1 occurrence of R3 and 0
occurrences of R5.
[00201] In other embodiments, Cy is heteroaryl substituted with 0-1
occurrences of R3 and 0-3
occurrences of R5. For example, Cy is heteroaryl substituted with 0-1
occurrences of R3 and 0-3 occurrences of R5.
In some embodiments, Cy is 5-membered heteroaryl substituted with 0-1
occurrences of R3 and 0-3 occurrences of
R5. In some embodiments, Cy is thiophenyl substituted with 0 occurrences of R3
and 1 occurrence of R5. In some
embodiments, Cy is thiophenyl substituted with 0-1 occurrence of R3 and 0
occurrence of R5. In some
embodiments, Cy is thiophenyl substituted with 1 occurrence of R3 and 0
occurrence of R5. In some embodiments,
Cy is isothiazolyl substituted with 0 occurrences of R3 and 1 occurrence of
R5. In some embodiments, Cy is
isothiazolyl substituted with 0-1 occurrence of R3 and 0 occurrence of R5. In
some embodiments, Cy is isothiazolyl
substituted with 1 occurrence of R3 and 0 occurrence of R5. In other
embodiments, Cy is 6-membered heteroaryl.
49

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R11
R13 N
N
Ri2 N R1
[00202] In some embodiments of compounds of Formula (I), Wd is
[00203] In other embodiments of compounds of Formula (I), Xis ¨(CH(R9)),-
. For example, z is 1.
[00204]
JVUNI R11
R13 N
R13 N
N
I
R12 KI
Rl R12I N=:1-- Rio
[00205] In various embodiments, Wd is or %MN
wherein RI , R11, R12,
and R13 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, or NR'R" wherein R' and R"
are taken together with nitrogen to form a cyclic moiety.
[00206] In some embodiments of the compound of Formula (I), at least one
of RI , R11, R12 and R13 is
hydrogen, cyano, halo, unsubstituted or substituted alkyl, unsubstituted or
substituted alkynyl, or unsubstituted or
substituted alkenyl. In some embodiments, at least one of RI , R11, R12 and
R13
is unsubstituted or substituted aryl.
In some embodiments, at least one of RI , R11, R12 and R13
is unsubstituted or substituted heteroaryl, which includes
but is not limited to heteroaryl having a five membered ring, heteroaryl
having a six membered ring, heteroaryl with
at least one nitrogen ring atom, heteroaryl with two nitrogen ring atoms,
monocylic heteroaryl, and bicylic
heteroaryl. In some embodiments, at least one of RI , R11, R12 and R13
is unsubstituted or substituted heterocyclyl,
which includes but is not limited to heterocyclyl with one nitrogen ring atom,
heterocyclyl with one oxygen ring
atom, heterocyclyl with one sulfur ring atom, 5 membered heterocyclyl, 6
membered heterocyclyl, saturated
heterocyclyl, unsaturated heterocyclyl, heterocyclyl having an unsaturated
moiety connected to the heterocyclyl
ring, heterocyclyl substituted by oxo, and heterocyclyl substituted by two
oxo. In some embodiments, at least one of
R10, R11, R12 and R13
is unsubstituted or substituted cycloalkyl, including, but not limited to,
cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl, or cycloalkyl each of which can be substituted by one
oxo; or cycloalkyl having an
unsaturated moiety connected to the cycloalkyl ring. In some embodiments, at
least one of RI , R11, R12 and R13 is
unsubstituted or substituted amido, carboxylic acid, unsubstituted or
substituted acyloxy, unsubstituted or
substituted alkoxycarbonyl, unsubstituted or substituted acyl, or
unsubstituted or substituted sulfonamido. In some
embodiments, RI , R11, R12,
and R13 are independently selected from hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl,
alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano,
and hydroxyl. In some
embodiments, RI , R11, R12,
and R13 are independently selected from hydrogen, amino, and chloro. In some
embodiments, RI is selected from amino and chloro.
[00207] In some embodiments, when at least one of RI , R11, R12 and R13
is alkyl, alkynyl,
alkenyl, aryl,
heteroaryl, heterocyclyl, cycloalkyl, alkoxycarbonyl, amido, acyloxy, acyl, or
sulfonamido, it is substituted with one
or more of alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl,
aryl, heteroaryl, alkoxy, amido, amino, acyl,

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl or nitro, each of
which alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, or
sulfonamido can itself be substituted.
[00208] In some embodiments of compounds of Formula (I), Wd is:
avvy
RN, N
I _1_
R12NR1c1
R11
[00209] In some embodiments of compounds of Formula (I), Wd is a
structure selected from:
NAN
JUVII JVVV N JNAIV
N
NN
I j R13 N N/N
N
I I ) N I
*L ,,
N R,iu Rir- )"--N Rii
or N . In some embodiments, Wd
is
IIIIVV %/VW
N N
I I
N*L Rio
N Ri
and RI is hydrogen, amino or halo. For example, Wd is and RI is Cl-.
In
.vuv
N
'-, N
I
./-NR-io
another instance, Wd is and RI is hydrogen.
[00210] In some embodiments, Cy is aryl substituted with 0-1 occurrences
of R3 and 0-3 occurrences of
R5. For example, Cy is phenyl substituted with 1 occurrence of R3 and 0
occurrences of R5. In other embodiments,
Cy is heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences of
R5. Cy can be, for example, pyridinyl,
pyridazinyl, thiophenyl, furanyl, pyrrolyl, thiazolyl, or isothiazolyl. In
some embodiments, Cy is 5-membered
heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences of R5.
In other embodiments, Cy is 6-
membered heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences
of R5. In some embodiments, Cy is
aryl, thiophenyl, or isothiazolyl. In some embodiments, Cy is thiophenyl
substituted with 0 occurrences of R3 and 1
occurrence of R5. In some embodiments, Cy is thiophenyl substituted with 0-1
occurrence of R3 and 0 occurrence of
R5. In some embodiments, Cy is thiophenyl substituted with 1 occurrence of R3
and 0 occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 0 occurrences of R3 and 1
occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 0-1 occurrence of R3 and 0
occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 1 occurrence of R3 and 0
occurrence of R5.
[00211] In some embodiments, the compound of Formula (I) has a structure
of Formula (II):
51

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R3 0
0
, - -
x
1
R8 Y
Wd
Formula (II)
[00212] For example, the compound of Formula (II) can have a structure of
Formula (Ha) or (MI):
R3 0 R3 0
B B
N N
le le
/
x x
1 1
R8 Y R8 Y
Wd Wd
Formula (Ha) or Formula (MI)
[00213] In other embodiments, the compound of Formula (II) has the
structure of Formula (IIIa) or
(11th):
R3 0 R3 0
NB
NB
1. 1
R8 7 Rs 7
Wd, or \Aid
Formula (Ma) or Formula (IIIb)
[00214] In some embodiments, the compound of Formula (II) has a structure
of Formula (11th-1):
R3 0
N13
1
4111 ......õ..-- R9
R8 Y
R1.Z.,..........:,,,N,................., N
I
R12N."... Rlo
R11
Formula (IIIb-1).
52

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00215] In some embodiments of compounds of Formula (Ha), (lib), (Ma), (Mb),
or (IIIb-1), Wd is
R13 N
N
, I i
w2../. N R10
R11
. In some embodiments of compounds of Formula (Ha), (IIb), (IIIa), (IIIb), or
(IIIb-1), Wd is
N .v.v
1. N jj R13 N N
N N \
I *L I ) 1\1-
IN
N R1 R121\r w 1
a structure selected from : , or
. In some
.11.11JV
N r N
/N1R1 0
embodiments of compounds of Formula (Ha), (IIb), (Ma), (Mb), or (IIIb-1), Wd
is and RI is
Juw
N r N
N*L Rio
hydrogen, amino or halo. For example, Wd is
and RI is Cl-. In another instance of a compound
...
i. N
N*L Rio
of Formula (Ha), (MI), (Ma), (Mb), or (IIIb-1), Wd is and RI is hydrogen.
[00216]
In some embodiments of compounds of Formula (Ha), (MI), (IIIa), (Mb), or (IIIb-
1), B is aryl
substituted with 0-3 occurrences of R2. For example, B is phenyl substituted
with 0-3 occurrences of R2. In some
embodiments of compounds of Formula (Ha), (MI), (IIIa), (Mb), or (IIIb-1), B
is unsubstituted phenyl. In other
embodiments of compounds of Formula (Ha), (MI), (IIIa), (11th), or (IIIb-1), B
is phenyl substituted with 1
occurrence of R2. R2 is, in some instances, halo or alkyl. In other
embodiments of compounds of Formula (Ha), (MI),
(IIIa), (Mb), or (IIIb-1), B is cycloalkyl or heterocyclyl.
[00217] In still other embodiments, the compound of Formula (I) has a
structure selected from:
R3 0 R3 0 R3 0
H N N N
, B RA , B H , B
1 1 1
I H I I
õ........, ..*-..... õ5"..yi
H N H R5 N N X HNN H
1
Wd Wd Wd
, , or .
[00218]
In some embodiments, the compound of Formula (I) has the structure of Formula
(IVa) or
Formula (IVb):
53

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
3
,B
&N-
S wb5 s'.-Wb5 X
)/
Wd or Wd
Formula (IVa) Formula (IVb).
[00219] For example, the compound of Formula (IVa) or Formula (IVb) has
the structure of Formula
(Va) or Formula (Vb):
R3 0 0
SwbR9 Swb5--R9
Wd or Wd
Formula (Va) Formula (Vb).
[00220] In some embodiments, the compound of Formula (I) has a structure
of Formula (Va-1):
0
3
-DeL
N NI3
S wbR9
R13 NN
R12 N Rl
R11
Formula (Va-1).
[00221] In some embodiments of compounds of Formula (IVa), (IVb), (Va), (Vb),
or (Va-1), Wb5 is Cle. For
example, Wb5 is CH. In some embodiments, the compound is a compound of Formula
(IVa), (IVb), (Va), (Vb), or
(Va-1), where R5 is H. In other embodiments of a compound of Formula (IVa),
(IVb), (Va), (Vb), or (Va-1), R5 is
selected from hydrogen, alkyl, cycloalkyl, halo, aryl, and heteroaryl. For
example, R5 is selected from methyl,
chloro or pyrazolo.
[00222] In some embodiments of compounds of Formula (IVa), (IVb), (Va),
(Vb), or (Va-1), Wd is
avvv
R1 NN
I
R12N Rio
Ri
. In some embodiments of compounds of Formula (IVa), (IVb), (Va), (Vb), or (Va-
1), Wd is
a structure selected from:
54

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
~As
WU', 41.11/V N N JIJIIV
NN R13 N
N
----:õ..-.õ--"-- N%."1"- Ri o Rir=-..-..-^- N-- Ri 1
or N . In some embodiments of
JUVII
N
N
, I
N*L Ri o
compounds of Formula (IVa), (IVb), (Va), (Vb), or (Va-1), Wd is
and RI is hydrogen, amino or
N r N
N*L R1
halo. For example, Wd is and RI is Cl-. In another instance of a compound
of Formula (IVa),
, I
N*I R10
(IVb), (Va), (Vb), or (Va-1), Wd is
and RI is hydrogen. In some embodiments of compounds of
Formula (IVa), (IVb), (Va), (Vb), or (Va-1), B is aryl substituted with 0-3
occurrences of R2. For example, B is
phenyl substituted with 0-3 occurrences of R2. In some embodiments of
compounds of Formula (IVa), (IVb), ((Va),
(Vb), or (Va-1), B is unsubstituted phenyl. In other embodiments of compounds
of Formula (IVa), (IVb), (Va),
(Vb), or (Va-1), B is phenyl substituted with 1 occurrence of R2. R2 is, in
some instances, halo or alkyl. In other
embodiments of compounds of Formula (IVa), (IVb), (Va), (Vb), or (Va-1), B is
cycloalkyl or heterocyclyl.
[00223] In other embodiments, the compound of Formula (I) has the
structure of Formula (VIa) or
Formula (VIb):
, B
N - B
N
eL
Wb5 X S'Nwb's'Lx
1 1
Y, Y,
Wd or Wd
Formula (VIa) Formula (VIb).
[00224] In some embodiments of the compound of Formula (VIa) or (VIb),
Wd is
wuv
Ri3 N ji N
R12 N Rio
R11 .
[00225] In other embodiments, the compound of Formula (VIa) or (VIb) has
the structure of Formula
(VIIa) or (VIIb):

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
R3\ IL ,B
N
S'Nw \R9
yvb5 ¨b
Wd or Wd
Formula (Vila) Formula (VIIb).
[00226] In some embodiments, the compound of Formula (Vila) or (VIIb) has
the structure of Formula
(Villa) or (VIIIb):
0 0
R3\ it
B
I
S'XX
Y, Y\
Wd or Wd
Formula (Villa) Formula (VIIIb).
[00227] In some embodiments, the compound of Formula (Vila) or (VIIb) has
the structure of Formula
(IXa) or (IXb):
0
R3\ ji 0
,B
N
S-
JNB R9
Wd or Wd
Formula (IXa) Formula (IXb).
[00228] In some embodiments, the compound of Formula (I) has a structure
of Formula (IXa-1):
0
R3\ N,B
C'T
R1 N
Ri2 -^y^-=
N Rl
R11
Formula (IXa-1).
[00229] In some embodiments, R3 is selected from alkyl, cycloalkyl, halo,
aryl, and heteroaryl. For
example, R3 is selected from methyl, chloro, and pyrazolo.
[00230] In some embodiments, B is aryl substituted with 0-3 occurrences
of R2. For example, B is
phenyl substituted with 0-3 occurrences of R2. B is, e.g., unsubstituted
phenyl. Alternatively, B is phenyl substituted
with 1 occurrence of R2. R2 can be, for instance, halo or alkyl. In other
embodiments, B is cycloalkyl. In other
embodiments, B is heterocyclyl.
56

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00231]
For example, the compound of Formula (I) is a compound of Formula (Via),
(Vib), (Vila),
(Viib), (Villa), (Viiib), (IXa), (IXb), or (iXa-1), where R3 is H. In other
embodiments of a compound of Formula
(Via), (Vila), (Villa), or (IXa), R3 is selected from hydrogen, alkyl,
cycloalkyl, halo, aryl, and heteroaryl. For
example, R3 is selected from methyl, chloro or pyrazolo. In some embodiments
of compounds of Formula (Via),
R13 N
N
I
R121\r RI
Ri
(Vib), (Vila), (Villa), (IXa), (IXb), or (IXa-1), Wd is
. In some
embodiments of compounds of Formula (Via), (Vib), (Vila), (Viib), (Villa),
(VIM)), (IXa), (IXb), or (iXa-1), Wd is
a structure selected from :
OW/
JUVII N ONAIV
11 R13 N
N N
II )
R11I
, or
N . In some embodiments of
compounds of Formula (Via), (Vib), (Vila), (Viib), (Villa), (Viiib), (IXa),
(IXb), or (IXa-1), Wd is
N Nr N
Ri
and RI is hydrogen, amino or halo. For example, Wd is
and RI is Cl-. In
another instance of a compound of Formula (Via), (Vib), (Vila), (Viib),
(Villa), (VIM)), (IXa), (IXb), or (iXa-1),
duw
I
/N% Ri 0
Wd is
and RI is hydrogen. In some embodiments of compounds of Formula (Via), (Vib),
(Vila),
(Viib), (Villa), (Viiib), (IXa), (IXb), or (IXa-1), B is aryl substituted with
0-3 occurrences of R2. For example, B is
phenyl substituted with 0-3 occurrences of R2. In some embodiments of
compounds of Formula (VIa), (Vib), (Vila),
(Viib), (Villa), (Viiib), (IXa), (IXb), or (IXa-1), B is unsubstituted phenyl.
In other embodiments of compounds of
Formula (Via), (Vib), (Vila), (Viib), (Villa), (Viiib), (IXa), (IXb), or (IXa-
1), B is phenyl substituted with 1
occurrence of R2. R2 is, in some instances, halo or alkyl. In other
embodiments of compounds of Formula (Via),
(Vib), (Vila), (Viib), (Villa), (Viiib), (IXa), (IXb), or (IXa-1), B is
cycloalkyl or heterocyclyl.
[00232] In another aspect, provided herein are compounds of Formula (X)
or (XI):
RI
N R1
1101
Wb X Wb X
Wd or Wd
57

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Formula (X) Formula (XI),
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein:
Wb5 is N, CHR8, or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is ¨(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)2-, -C(=0)-, or ¨0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfonamido, halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above
substituents can be substituted with 0, 1, 2, or 3 R17;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea, carbonate, or
NR'R" wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is ¨(CH(R16)),¨;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R16)-, -C(=0)-(CHR16),-, -C(=0)-,
-N(R16)-C(=0)-, or
-N(R16)-C(=0)NH-,-N(R16)C(R16)2-, or -C(=0)-N(R16)-(CHR16),-;
each z is an integer of 1,2, 3, or 4;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl; and
JVV1/
R'IN jx
I 1
R12N R1
Wd iS R11 /
wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and R17
are independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, oxo, or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.
58

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00233] In some embodiments, the compound is a compound of Formula (X).
For example, the
compound has the Formula:
R3
N R1
Wb5.-X
I
Wd .
[00234] In some embodiments, Cy is a 5- or 6-membered ring. In some
embodiments, Cy is a 6-
membered ring, such as a 6-membered aryl ring. For instance, Cy is a 6-
membered ring, including e.g. phenyl. In
other embodiments, Wb5 is CH. In yet other embodiments, R11 is H. In still
other embodiments, R12 is H. In other
embodiments, X-Y is -CH2-N(CH3).
[00235] In one aspect, provided herein are compounds are provided of
Formula (I):
0
B
= N
-
Wb5 X
1
Wd
Formula (I)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
Wb5 is Cle, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea or carbonate;
X is absent or is -(CH(R9)),-;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-, -N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when Wb5 is N, no more than one of X or Y is absent;
59

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino,
acyl, acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
R11
R1.-3 N
-.....- ..,-...../Lx
1 ii
_,.....- ..1,-).
R12 N.., R1
Wd is 4Ø0,JV
/
wherein X1 is N or CR14;
wherein R10, R11, R12, R13, R14, and K-17
are independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, carboxylic
acid, oxo, or NR'R" wherein R' and R" are taken together with nitrogen to form
a cyclic moiety.
[00236] In one aspect, provided herein are compounds of Formula (XII):
0
B
410 N
-
Wb5 X
I
Wd
Formula (XII)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
Wb5 is CR8, CHR8, or N,
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl, aryl or
heteroaryl, each of
which is substituted with 0-4 R2;
R2 is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
arylalkyl, heteroaryl,
heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea or carbonate;
X is absent or is ¨(CH(R9)),-;

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, aryl, heteroaryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl,
alkoxy, amido, amino,
acyl, acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl; and
X4.1
R10
x' 50
- N R12
Wd is R11
wherein one of X5 and X2 is N and one of X5 and X2 is C;
X3 and X4 are each independently CR13 or N;
X2 and X3 are not both N; and
each R10, R11, R12, K-13,
and R17 is independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, oxo, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety.
[00237] In certain embodiments, Wb5 is Cle. In some embodiments, R8 is
hydrogen.
[00238] In certain embodiments, X3 is CR13. In some embodiments, R13 is
hydrogen. In some
embodiments, X3 is N.
[00239] In certain embodiments, X4 is CR13. In some embodiments, R13 is
hydrogen. In some
embodiments, X4 is N.
[00240] In some embodiments, Cy is aryl or heteroaryl substituted with 0-
1 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is aryl or heteroaryl substituted
with 0 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is aryl (e.g., phenyl) substituted
with 0-1 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is phenyl substituted with 1
occurrence of R3 and 0 occurrences of R5.
[00241] In some embodiments, Cy is heteroaryl substituted with 0-1
occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy is 5-membered heteroaryl (e.g.,
thiophenyl or isothiazolyl) substituted
with 0-1 occurrences of R3 and 0-3 occurrences of R5. In some embodiments, Cy
is thiophenyl substituted with 0
occurrences of R3 and 1 occurrence of R5. In some embodiments, Cy is
thiophenyl substituted with 0-1 occurrence
of R3 and 0 occurrence of R5. In some embodiments, Cy is thiophenyl
substituted with 1 occurrence of R3 and 0
occurrence of R5. In some embodiments, Cy is isothiazolyl substituted with 0
occurrences of R3 and 1 occurrence of
R5. In some embodiments, Cy is isothiazolyl substituted with 0-1 occurrence of
R3 and 0 occurrence of R5. In some
61

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
embodiments, Cy is isothiazolyl substituted with 1 occurrence of R3 and 0
occurrence of R5. In some embodiments,
Cy is 6-membered heteroaryl (e.g., pyridinyl).
[00242] In certain embodiments, R3 is halo (e.g., chloro). In some
embodiments, R3 is heteroaryl (e.g.,
1-methy1-4-pyrazoly1).
[00243] In some embodiments, X is ¨(CH(R9)),-. In some embodiments, z is
1. In some embodiments,
R9 is Ci_io alkyl (e.g., methyl).
[00244] In some embodiments, is absent, -0-, -NH(R9)-, or -S(=0)2-. In
certain embodiments, Y is
absent. In some embodiments, Y is ¨N(R9)-. In some embodiments, R9 is
hydrogen.
;s5Y
HN õss
[00245] In certain embodiments, X-Y is . In
some embodiments, X-Y is -CH2-N(CH3). In
some embodiments, X-Y is (S)-CH(CH3)-NH-. In some embodiments, X-Y is (R)-
CH(CH3)-NH-.
[00246] In certain embodiments, B is aryl or heteroaryl substituted with
0-4 occurrences of R2. In some
embodiments, B is aryl (e.g., 6-membered aryl) substituted with 0-4
occurrences of R2. In some embodiments, B is
phenyl substituted with 0-4 occurrences of R2. In some embodiments, B is
phenyl substituted with 0 occurrences of
R2. In some embodiments, B is phenyl substituted with 1 occurrence of R2. In
some embodiments, R2 is phenyl
substituted at the ortho position. In some embodiments, R2 is phenyl
substituted at the meta position. In some
embodiments, R2 is phenyl substituted at the para position. In some
embodiments, R2 is halo (e.g., fluoro).
[00247] In certain embodiments, B is cycloalkyl substituted with 0-4
occurrences of R2. In some
embodiments, B is cycloalkyl (e.g., cyclopropyl) substituted with 0
occurrences of R2.
[00248] In certain embodiments, B is alkyl substituted with 0-4
occurrences of R2. In some
embodiments, B is alkyl (e.g., methyl or ethyl) substituted with 0 occurrences
of R2. In some embodiments, B is
alkyl (e.g., methyl or ethyl) substituted with 1 occurrence of R2. In some
embodiments, R2 is heterocyclyl (e.g.,
pyrrolyl).
[00249] In certain embodiments, X1 is N and X2 is C. In some embodiments,
X1 is C and X2 is N.
[00250] In certain embodiments, R1 is hydrogen. In some embodiments, R1
is alkyl (e.g., methyl).
[00251] In certain embodiments, R11 is hydrogen.
[00252] In certain embodiments, R12 is hydrogen. In some embodiments, R12
is halo (e.g., chloro).
62

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
ivv Iv
NIMN\
[00253] In certain embodiments, Wd is N . In
some embodiments, Wd is N . In
snrirv .rtnry
some embodiments, Wd is ul N . In some embodiments, Wd
is N . In some embodiments,
MAP
.nniv
N N
,N N
Wd is N . In some embodiments, Wd is
[00254] In some embodiments, R3 is halo, alkyl, alkoxy, heteroaryl, or
cycloalkyl. For example, R3 is
CH3, CH2CH3, CF3, Cl, or F. In other embodiments, R3 is CH3, CF3, or Cl. In
other embodiments, each R3 and R5 is
independently selected from CH3, OCH3, CF3, and halo. In certain embodiments,
R3 is halo (e.g., chloro). In some
embodiments, R3 is heteroaryl (e.g., 1-methyl-4-pyrazoly1).
[00255] In some embodiments, B is cycloalkyl, heterocyclyl, aryl, or
heteroaryl, which is substituted
with 0-4 occurrences of R2, R3 is H, halo, alkyl, alkoxy, aryl, cycloalkyl, or
heteroaryl, each R5 is H, halo, alkyl,
alkoxy, aryl, cycloalkyl, or heteroaryl, and R9 is hydrogen or alkyl.
[00256] In some embodiments, R5 is selected from alkyl, cycloalkyl, halo,
aryl, and heteroaryl. For
example, R5 is selected from methyl, chloro, and pyrazolo.
JNINIV
N., iv
Rb0Q5Q
2N R12
[00257] In various embodiments, Wd is R11
[00258] In such embodiments, X3 is CR13 or N; each R10, R11, R12 and R13
is independently hydrogen,
alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido,
halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with
nitrogen to form a cyclic moiety;
and one of X1 and X2 is N and one of X1 and X2 is C.
[00259] For example, X1 is N and X2 is C. Alternatively, X1 is C and X2
is N. In some embodiments,
X3 is CR13 or N. In other embodiments, X4 is CR13 or N.
[00260] In some embodiments, X3 is CR13. For example, R13 is H. In other
embodiments, X3 is N.
[00261] In some embodiments, X4 is CR13. For example, R13 is H. In other
embodiments, X4 is N.
[00262] In some embodiments of the compound of Formula (I), at least one
of R10, R11, R12 and R13 is
hydrogen, cyano, halo, unsubstituted or substituted alkyl, unsubstituted or
substituted alkynyl, or unsubstituted or
substituted alkenyl. In some embodiments, at least one of R10, R11, R12 and
R13
is unsubstituted or substituted aryl.
In some embodiments, at least one of R10, R11, R12 and R13
is unsubstituted or substituted heteroaryl, which includes,
63

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
but is not limited to, heteroaryl having a five membered ring, heteroaryl
having a six membered ring, heteroaryl with
at least one nitrogen ring atom, heteroaryl with two nitrogen ring atoms,
monocylic heteroaryl, and bicylic
heteroaryl. In some embodiments, at least one of RI , R11, R12 and R13
is unsubstituted or substituted heterocyclyl,
which includes, but is not limited to, heterocyclyl with one nitrogen ring
atom, heterocyclyl with one oxygen ring
atom, heterocyclyl with one sulfur ring atom, 5 membered heterocyclyl, 6
membered heterocyclyl, saturated
heterocyclyl, unsaturated heterocyclyl, heterocyclyl having an unsaturated
moiety connected to the heterocyclyl
ring, heterocyclyl substituted by oxo, and heterocyclyl substituted by two
oxo. In some embodiments, at least one of
R10, R11, R12 and R13
is unsubstituted or substituted cycloalkyl, including, but not limited to,
cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl, cycloalkyl substituted by one oxo; or cycloalkyl
having an unsaturated moiety connected to
the cycloalkyl ring. In some embodiments, at least one of RI , R11, R12 and
R13
is unsubstituted or substituted amido,
unsubstituted or substituted acyloxy, unsubstituted or substituted
alkoxycarbonyl, unsubstituted or substituted acyl,
or unsubstituted or substituted sulfonamido. In some embodiments, RI , R11,
R12,
and R13 are independently selected
from hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, and hydroxyl. In some embodiments, RI , R11, R12,
and R13 are independently selected
from hydrogen, amino, and chloro. In some embodiments, RI is selected from
amino and chloro.
[00263] In some embodiments, when at least one of RI , R11, R12 and R13 is
alkyl, alkynyl,
alkenyl, aryl,
heteroaryl, heterocyclyl, cycloalkyl, alkoxycarbonyl, amido, acyloxy, acyl, or
sulfonamido, it is substituted with one
or more of alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl,
aryl, heteroaryl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy or nitro, each of
which alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, or
sulfonamido may itself be substituted.
[00264] In some embodiments, Wd is:
OVVII WV
N
X x3
Rio_c_L-N 3
N
Ri2 Rb0 R12
'N
R11 or R11
[00265] In some embodiments of compounds of Formula (I), Wd is a structure
selected from:
%/VW JIAIV ONAllf
R13 R13 N
-N N
Rio
\N
R12 'N R12
R11 R11 R11
, or
JN.INN
N
x3
X3
12
[00266] In some embodiments, Wd is NR or D 1 2
" and R12 is hydrogen or
halo.
64

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
JVVI/ JVVV
x3
N'*L 12 *L 12
N R
For example, Wd is and R12 is Cl. In another instance, Wd is N R
and R12 is
hydrogen.
..0111A,
N
wo X3 R1 0 _e/ Xi 3
[00267] In other embodiments, Wd is N
or Nand RI is hydrogen
or
halo.
asrusr
Nk
N
X3 X3
NJ
[00268] In still other embodiments, Wd is N
or N. For example, Wd
is
GAM =AA/V
N N
N N
µ.--N
or
[00269] In some embodiments, Cy is aryl substituted with 0-1 occurrences
of R3 and 0-3 occurrences of
R5. For example, Cy is phenyl substituted with 1 occurrence of R3 and 0
occurrences of R5. In other embodiments,
Cy is heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences of
R5. Cy may be, for example,
pyridinyl, pyridazinyl, thiophenyl, furanyl, pyrrolyl, thiazolyl, or
isothiazolyl. In some embodiments, Cy is 5-
membered heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences
of R5. In other embodiments, Cy is
6-membered heteroaryl substituted with 0-1 occurrences of R3 and 0-3
occurrences of R5. In some embodiments, Cy
is aryl, thiophenyl, or isothiazolyl. In some embodiments, Cy is thiophenyl
substituted with 0 occurrences of R3 and
1 occurrence of R5. In some embodiments, Cy is thiophenyl substituted with 0-1
occurrence of R3 and 0 occurrence
of R5. In some embodiments, Cy is thiophenyl substituted with 1 occurrence of
R3 and 0 occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 0 occurrences of R3 and 1
occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 0-1 occurrence of R3 and 0
occurrence of R5. In some
embodiments, Cy is isothiazolyl substituted with 1 occurrence of R3 and 0
occurrence of R5.
[00270] In some embodiments, the compound of Formula (XII) has the
structure of Formula (XIII):
R3 0
X
R8
Wd
(Formula XII)

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00271] For example, the compound of Formula (I) can have a structure of
Formula (XIIIa) or (XIIIb):
R3 0 R3 0
B B
N N
le 1 140
x x
1 I
R8 Y\ R8 Y\
Wd " lm
d .
Formula (XIIIa) or Formula (XIIIb)
[00272] In other embodiments, the compound of Formula (XIIIa) or (XIIIb)
has the structure of
Formula (XIVa) or (XIVb):
R3 0 R3 0
N13
N13
el 1
Wd , or Wd ,
Formula (XIVa) or Formula (XIVb)
[00273] In some embodiments, the compound of Formula (XIII) has a
structure of Formula (XIVb-1):
R3 0
, B
N
I. / R9
z
R8 Y
N...v v
., .,3
Rio_950)
*2
N R12
R11
Formula (XIVb-1).
[00274] In some embodiments of compounds of Formula (XIIIa), (XIIIb),
(XIVa), (XIVb), or (XIVb-
1), Wd 1S:
N ,L,
_N:-1/x3
Rio_s.: c 's3 R10
\ N
N R12 ' N R12
R11 or R11
[00275] In some embodiments of compounds of Formula (XIIIa), (XIIIb),
(XIVa), (XIVb), or (XIVb-
1), Wd is:
66

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
OVVII JIAIV ONAllf
R13 R13
N.- N -.----",,z,-- N..r N
Rio_c__L____ Rio 80.-N Rio_c_jr\ NI
...-- ..,-....L.,
N R12 , N- -R12 N R12
R11 , R11 R11
,or .
[00276] In some embodiments of compounds of Formula (XIIIa), (XIIIb),
(XIVa), (XIVb), or (XIVb-
IN a m v
3
..........1 /s
%....-N'L 12 ------ ..,:jL
1), Wd is or R12
N* R N and R12 is hydrogen or halo. For example,
Wd iS
N....,õ
'N R12

2and R12 is Cl- In another instance, Wd is 'N R12 and R12 is hydrogen.
[00277] In other embodiments of compounds of Formula (XIIIa), (XIIIb),
(XIVa), (XIVb), or (XIVb-
Jvw
N N ,,, 1 v---: X3 I/ --IN '`3
R1CIN R101 ---c-
1), Wd is 'N N
or and R1 is hydrogen or halo.
[00278] In still other embodiments of compounds of Formula (XIIIa),
(XIIIb), (XIVa), (XIVb), or
N..........<:,..õ )(3 / ....N -. )(3
....-N c-----L ,...- N /=.,..--
<_-:-."--c
(XIVb-1), Wd is 'N
or N . For example, Wd is 'N N
or N .
[00279] In some embodiments of compounds of Formula (XIIIa), (XIIIb),
(XIVa), (XIVb), or (XIVb-
1), B is aryl substituted with 0-3 occurrences of R2. For example, B is phenyl
substituted with 0-3 occurrences of R2.
In some embodiments of compounds of Formula (XIIIa), (XIIIb), (XIVa), (XIVb),
or (XIVb-1), B is unsubstituted
phenyl. In other embodiments of compounds of Formula (XIIIa), (XIIIb), (XIVa),
(XIVb), or (XIVb-1), B is phenyl
substituted with 1 occurrence of R2. R2 is, in some instances, halo or alkyl.
In other embodiments of compounds of
Formula ((XIIIa), (XIIIb), (XIVa), (XIVb), or (XIVb-1), B is cycloalkyl or
heterocyclyl.
[00280] In still other embodiments, the compound of Formula (XII) is a
compound which has a
structure selected from:
R3 0 R3 0 R3 0
H N , B R5=NL , B H N, B
1 1 1
I I I
H N H R5 NNX H N N H
1
Wd vvd , and Wd
, .
[00281] In some embodiments, the compound of Formula (XIV) has the
structure of Formula (XVa) or
Formula (XVb):
67

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
3
,B ,B
N
N\ I
S wb5 S---ThAti,5 X
Wd or Wd
Formula (XVa) Formula (XVb).
[00282] In other embodiments, the compound of Formula (XVa) or (XVb) is a
compound of Formula
(XVIa) or (XVIb):
R3 0 0
N'{'
wbR9 Sw1)5-- R9
Wd or Wd
Formula (XVIa) Formula (XVIb).
[00283] In some embodiments, the compound of Formula (XII) has a structure
of Formula (XVIa-1):
N,B
N
µS R9
R8 1(
N
y_
R1OVb0
R11
Formula (XVIa-1).
[00284] In some embodiments of compounds of Formula (XVa), (XVb), (XVIa),
(XVIb), or (XVIa-1),
Wb5 is Cle. For example, Wb5 is CH. In other embodiments of a compound of
Formula (XVa) or (XVb), R3 is
selected from alkyl, cycloalkyl, halo, aryl, and heteroaryl. For example, R3
is selected from methyl, chloro or
pyrazolo.
[00285] In some embodiments of compounds of Formula (XVa), (XVb), (XVIa),
(XVIb), or (XVIa-1),
Wd is:
.A111V
N
x3
N R12
R10
R12
R11 or R11
[00286] In some embodiments of of compounds of Formula (XVa), (XVb),
(XVIa), (XVIb), or (XVIa-
1), Wd is a structure selected from:
68

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
OVVIf s/VW ~IV
R13 R13 N
N ... ...,------ N
-
Rio_c N _L__ Rio 80õ ..N Rio, N N
.õ)....... L.,
N R12 , N- - R12 Nõ R12
R11 , R11 R11
,and .
[00287] In some embodiments of compounds of Formula (XVa), (XVb), (XVIa),
(XVIb), or (XVIa-1),
JINV
/N.,..... x3 N a...1 m v
...... /s3
%..--N .---- ..<,..1õ,
12
Wd is R12
N R or N and R12 is hydrogen or halo. For example, Wd
is
&NW
N...., õ N...., v
/ --. A3 / ---- A3
µ..-N 12 µ..õ,- N *L
'N R - and R12 is Cl-. In another instance, Wd is 'N R12 and R12
is hydrogen.
[00288] In other embodiments of compounds of Formula (XVa), (XVb),
(XVIa), (XVIb), or (XVIa-1),
nn
N..... x N ,,,J v
R10 3 R1O_U1 )'s3
-,--N
Wd is 'N
or N and R1 is hydrogen or halo.
[00289] In still other embodiments of compounds of Formula (XVa), (XVb),
(XVIa), (XVIb), or
N....<( NN,...... N¨j\ N
/ -- X3 i-N X3
µ...-N \.-.-.-----c÷ .....-N /-cr"-- \....------
(XVIa-1), Wd is ' N
or N . For example, Wd is 'N N
or N
.
[00290] In some embodiments of compounds of Formula (XVa), (XVb), (XVIa),
(XVIb), or (XVIa-1),
B is aryl substituted with 0-3 occurrences of R2. For example, B is phenyl
substituted with 0-3 occurrences of R2. In
some embodiments of compounds of Formula (XVa), (XVb), (XVIa), (XVIb), or
(XVIa-1), B is unsubstituted
phenyl. In other embodiments of compounds of Formula (XVa), (XVb), (XVIa),
(XVIb), or (XVIa-1), B is phenyl
substituted with 1 occurrence of R2. R2 is, in some instances, halo or alkyl.
In other embodiments of compounds of
Formula (XVa), (XVb), (XVIa), (XVIb), or (XVIa-1), B is cycloalkyl or
heterocyclyl.
[00291] In some embodiments, the compound of Formula (I) is a compound of
Formula (XVIIa) or
(XVIIb):
, B
N-B
eN
S'Nwb5 x S'Nwb's'Lx
1
Y, Y,
Wd or Wd
Formula (XVIIa) Formula (XVIIb).
69

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00292] In other embodiments, the compound of Formula (XVIIa) or (XVIIIb)
has the structure of
Formula (XVIIIa) or (XVIIIb):
0 0
R3\ ji ,B
(--rN
s-N 9
wb5 S'=,õ, b R9
"
Wd or Wd
Formula (XVIIIa) Formula (XVIIIb).
[00293] In some embodiments, the compound of Formula (XVIIIa) or (XVIIIb)
has the structure of
Formula (XIXa) or (XIXb):
0 0
R3\
( 1
Y, Y,
Wd or Wd
Formula (XIXa) Formula (XIXb).
[00294] In some embodiments, the compound of Formula (XVIIIa) or (XVIIIb)
has the structure of
Formula (XXa) or (XXl3):
0
R3L 0
,B
e.õNB S- NR9 S- R9
Wd or Wd
Formula (XXa) Formula (XXl3).
[00295] In some embodiments, the compound of Formula (XII) has a
structure of Formula (XXa-1):
R3
NB
R10_91`*520)^3
N R12
Ril
Formula (XXa-1).
[00296] In some embodiments of a compound of Formula (XVIIa), (XVIIb),
(XVIIIa), (XVIIIb),
(XIXa), (XIXb), (XXa), (XXl3), or (XXa-1), R3 is selected from alkyl,
cycloalkyl, halo, aryl, and heteroaryl. For

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
example, R3 is selected from methyl, chloro or pyrazolo. In some embodiments
of compounds of Formula (XVIIa),
(XVIIb), (XVIIIa), (XVIIIb), (XIXa), (XIXb), (X(a), (XXb), or (XXa-1), Wd is:
Jvw
N
_Nr x3
Rio_c__L--- 's3 R10 ,
\ N
N R12 'N R12
R11 R11
or .
[00297]
In some embodiments of of compounds of Formula (XVIIa), (XVIIb), (XVIIIa),
(XVIIIb),
(XIXa), (XIXb), (XXa), (XXb), or (XXa-1), Wd is a structure selected from:
wv
R13 R13 N
N....N ..-"*....,-- N,._1/
Rio Rio
Rio_y _c3 2I
N
Ns*, R12 \ 'N R12 N R12
R11 , R11 R11
,and .
[00298]
In some embodiments of compounds of Formula (XVIIa), (XVIIb), (XVIIIa),
(XVIIIb),
N_....1/1 x N
3 ,,,v
U., /s3
.....-N *L
(XIXa), (XIXb), (XXa), (XXb), or (XXa-1), Wd is 'N R12or
N R12 and R12 is hydrogen or
Juw
N...._
...2N 17 µ.--N *L
halo. For example, Wd is 'N R.-and R12 is
Cl-. In another instance, Wd is ' N R12 and R12 is
hydrogen.
[00299]
In other embodiments of compounds of Formula (XVIIa), (XVIIb), (XVIIIa),
(XVIIIb),
N
N_
c_-.1/ x3
¨N X3
R10 Rio_c_.
(XIXa), (XIXb), (XXa), (XXb), or (XXa-1), Wd is ' N or
N and R1 is hydrogen or
halo.
[00300]
In still other embodiments of compounds of Formula (XVIIa), (XVIIb), (XVIIIa),
(XVIIIb),
vw
N¨,1( N x3 / .i ¨N X3
,N'\J N
(XIXa), (XIXb), (XXa), (XXb), or (XXa-1), Wd is or
. For example, Wd is
dWV
or
N
/
N.......T. <1..x":.........
/ i¨N N IN
K....!--
.
[00301]
In some embodiments of compounds of Formula (XVIIa), (XVIIb), (XVIIIa),
(XVIIIb),
(XIXa), (XIXb), (XXa), (XXb), or (XXa-1), B is aryl substituted with 0-3
occurrences of R2. For example, B is
71

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
phenyl substituted with 0-3 occurrences of R2. In some embodiments of
compounds of Formula (XVIIa), (XVIIb),
(XVIIIa), (XVIIIb), (XIXa), (XIXb), (XXa), (XXb), or (XXa-1), B is
unsubstituted phenyl. In other embodiments of
compounds of Formula (XVIIa), (XVIIb), (XVIIIa), (XVIIIb), (XIXa), (XIXb),
(XXa), (XXI)), or (XXa-1), B is
phenyl substituted with 1 occurrence of R2. R2 is, in some instances, halo or
alkyl. In other embodiments of
compounds of Formula (XVIIa), (XVIIb), (XVIIIa), (XVIIIb), (XIXa), (XIXb),
(XXa), (XXI)), or (XXa-1), B is
cycloalkyl or heterocyclyl.
[003 02] In some embodiments of the compound of Formula (XII), B is aryl
or heteroaryl substituted
with 0 or 1 occurrences of R2 and Cy is a 5- or 6-membered aryl or heteroaryl
group. For example, B is aryl
substituted with 0 or 1 occurrences of R2 and Cy is a 5- or 6-membered aryl or
heteroaryl group. In another example,
B is aryl or heteroaryl substituted with 0 or 1 occurrence of R2 and Cy is a 6-
membered aryl group. Cy is, for
instance, phenyl substituted with alkyl, fluroalkyl, aryl, heteroaryl or halo.
In such embodiments, Wd can be, for
../VW GAM =AA/V
z N
example, or
[00303] In other embodiments of the compound of Formula (XII), B is aryl
or heteroaryl substituted
with 0 or 1 occurrences of R2, Cy is a 5- or 6-membered aryl or heteroaryl
group, X is ¨(CH(R9)),- and Y is -
NH(R9)-, wherein each R9 is chosen independently. For example, -X-Y- is -
CH2(CH3)-NH-. In some embodiments,
B is aryl substituted with 0 or 1 occurrences of R2 and Cy is a 5- or 6-
membered aryl or heteroaryl group. In another
example, B is aryl or heteroaryl substituted with 0 or 1 occurrence of R2 and
Cy is a 6-membered aryl group. Cy is,
for instance, phenyl substituted with alkyl, fluroalkyl, aryl, heteroaryl or
halo. In such embodiments, Wd can be, for
N
N
example, or
[00304] In some embodiments, the compound disclosed herein is a compound
of Formula (XIIIb)
where R3 is halo, alkyl, heteroalkyl, fluoroalkyl, alkenyl, alkynyl,
cycloalkyl, heteroaryl, or heterocyclyl; X is
.N N
11
CH(R9)-, where R9 is methyl or ethyl; le is H; Y is ¨NH-; and Wd is or
[003 05] In another aspect, provided herein is a compound of formula (XXI)
or (XXII):
RI
111
R1
1101 4101
Wb5 Wb5
Wd or Wd
Formula (XXI) Formula (XXII),
72

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein:
Wb5 is N, CHR8 or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is ¨(L)-R1';
L is a bond, -S-, -N(R15)-, -C(R15)2-, -C(=0)-, or ¨0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, suffinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocyclyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R"
wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is ¨(CH(R9)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is an integer of 1,2, 3, or 4;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl; and
.11JVNJ
X X
0 k50)
2N R12
Wd iS R11
wherein one of X5 and X2 is N and one of X5 and X2 is C;
wherein X3 and X4 are each independently selected from CR13 and N; and
each R10, R11, R12, R13,
and R17 is independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate,
urea, carbonate, oxo, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety.
73

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00306] In some embodiments, Cy is a 5- or 6-membered ring. For instance,
Cy is a 6-membered ring,
including e.g. phenyl. In other embodiments, Wb5 is CH. In yet other
embodiments, R10, R11, R12,
and R13 are H. In
some embodiments, X-Y is -CH2-N(CH3).
[00307] In some embodiments, Cy is a 6-membered ring, such as a 6-
membered aryl ring. In other
embodiments, Wb5 is CH. In still other embodiments, -X-Y- is -CH2-N(CH3).
[00308] In another aspect, provided herein is a compound of Formula
(XXIII):
R1
0 N
Wb5 Xi
1
y
Wd
Formula (XXIII)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Wb5 is N or CR8;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
Cy is aryl or heteroaryl substituted by 0-1 occurrences of R3 and 0-3
occurrences of R5;
R1 is ¨(L)-R1';
L is a bond, -S-, -N(R16)-, -C(R15)2-, -C(=0)-, or ¨0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, heterocyclylalkyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy,
amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea,
carbonate, substituted nitrogen,or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
R3 is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, haloalkyl,
heteroalkyl, alkoxy, amido,
amino, acyl, acyloxy, sulfinyl, sulfonyl, sulfoxide, sulfone, sulfonamido,
halo, cyano, heteroaryl, aryl, hydroxyl, or
nitro; wherein each of the above substituents can be substituted with 0, 1, 2,
or 3 R17;
each R5 isindependently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido,
amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R"
wherein R' and R" are taken
together with nitrogen to form a cyclic moiety;
X is absent or is ¨(CH(R14)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR14),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)0(R14)2_,
or
each z is an integer of 1,2, 3, or 4;
74

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
each R9 and R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, or
heteroaryl;
each R14 and R15 is independently hydrogen, alkyl, aryl, heteroaryl or halo;
and
N-NN
Rio_yN R12
Wd iS R11
wherein R10, R1i, R12,
and R17 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocycloalkyloxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro,
phosphate, urea, carbonate, oxo, or NR'R"
wherein R' and R" are taken together with nitrogen to form a cyclic moiety.
[00309] In some embodiments, R5 is halo, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heteroaryl, or
heterocyclyl. In some embodiments, R5 is alkyl (e.g., methyl) or halo (e.g.,
chloro). In some embodiments, R5 is
halo (e.g., chloro).
[00310] In certain embodiments, n is 1 and L is -C(=0)- or -0-. In some
embodiments, L is -C(=0)-.
[00311] In certain embodiments, R1' is alkyl or NR'R" wherein R' and R"
are taken together with
nitrogen to form a cyclic moiety. In some embodiments, R' and R" are taken
together with nitrogen to form a
heterocyclic moiety (e.g., morpholinyl).
[00312] In certain embodiments, X is -(CH(R9)),-. In some embodiments, z
is 1. In some
embodiments, X is -CH2- or -CH(CH3)-. In some embodiments, the carbon of the -
CH(CH3)- moiety has a (S)
stereochemical configuration. In some embodiments, the carbon of the -CH(CH3)-
moiety has- a (R) stereochemical
configuration.
[00313] In certain embodiments, Y is absent, -0-, -NH(R9)-, or -S(=0)2-.
In some embodiments, R9 is
methyl or hydrogen.
[00314] In certain embodiments, -X-Y- is -CH2-N(CH3). In some
embodiments, X-Y is
(S)-CH(CH3)-NH-. In some embodiments, -X-Y- is (R)-CH(CH3)-NH-.
[00315] In certain embodiments, R10, R11 and R12
are independently selected from hydrogen, amino,
and chloro. In some embodiments, R1 is amino or chloro.
[00316] In some embodiments, Cy is aryl substituted with 0-3 occurrences
of R5. For example, Cy is
phenyl substituted with 1 occurrence of R3.
[00317] In some embodiments, R3 is halo, alkyl, heteroalkyl, fluoroalkyl,
alkenyl, alkynyl, cycloalkyl,
heteroaryl, or heterocyclyl. For example, R3 is alkyl or halo.
[00318] In other embodiments, L is -(C(=0)- or -0-.
[00319] In some embodiments, R1' is substituted alkyl, substituted
nitrogen, or NR'R" wherein R' and
R" are taken together with nitrogen to form a cyclic moiety. For instance, the
cyclic moiety is a heterocyclic or
heteroaryl group such as a morpholino group. In other instances, R1' is
substituted alkyl, including alkyl which is
substituted with a heterocyclyl group.

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00320]
In some embodiments, X is absent or is ¨(CH(R9)),-. In some embodiments, R9 is
methyl or
hydrogen. For example, z is 1. In some instances, X is -CH2- or -CH(CH3)-. In
some embodiments, the carbon of the
-CH(CH3)- moiety has a (S) stereochemical configuration. Alternatively, the
carbon of the -CH(CH3)- moiety has a
(R) stereochemical configuration. In some embodiments, Y is absent, -0-, -
NH(R9)-, or -S(=0)2-. In some
embodiments, -X-Y- is -CH2-N(CH3). Alternatively, -X-Y- is (S)-CH(CH3)-NH-or
(R)-CH(CH3)-NH-.
[00321]
In some embodiments, R10, R11 and R12 are independently selected from
hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, and
hydroxyl. For example, R10, R11 and R12 are independently selected from
hydrogen, amino, and chloro. In one
instance, R1 is amino or chloro.
[00322]
In some embodiments, Cy is aryl or heteroaryl group substituted by 0-1
occurrences of R3 and
0-3 occurrences of R5. In some embodiments, Cy is aryl substituted with 0-1
occurrences or R3 and 0-3 occurrences
of R5. For example, Cy is phenyl substituted with 1 occurrence of R3 and 0
occurrences of R5. In other
embodiments, Cy is phenyl substituted with 0 occurrences of R3 and 0
occurrences of R5. In other embodiments, Cy
is heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences of
R5. Cy can be, for example, pyridinyl,
pyridazinyl, thiophenyl, furanyl, pyrrolyl, thiazolyl, or isothiazolyl. In
some embodiments, Cy is 5-membered
heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences of R5.
In other embodiments, Cy is 6-
membered heteroaryl substituted with 0-1 occurrences of R3 and 0-3 occurrences
of R5. In some embodiments, Cy is
aryl, thiophenyl, or isothiazolyl. For example, Cy is thiophenyl substituted
with 0 occurrences of R3 and 1
occurrence of R5. In another example, Cy is isothiazolyl substituted with 0
occurrences of R3 and 1 occurrence of
R5.
[00323]
In some embodiments or compounds of Formula (XXIII), Rr can be hydrogen,
alkyl, heteroalkyl,
alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl,
alkoxy, heterocyclyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl,
sulfonamido, halo, cyano, hydroxyl, nitro,
phosphate, urea, carbonate or NR'R", wherein R' and R" are taken together with
nitrogen to form a cyclic moiety.
[00324]
In some embodiments of compounds of Formula (XXIII), Rr can be hydrogen, or
unsubstituted or substituted alkyl (including, but not limited to, -CH3, -
CH2CH3, n-propyl, isopropyl, n- butyl, tert-
butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, Rr is
unsubstituted or substituted alkenyl
(including, but not limited to, unsubstituted or substituted C2-05alkenyl such
as, for example, vinyl, allyl, 1-methyl
propen- 1 -yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl
(including, but not limited to, unsubstituted
or substituted C2-05alkynyl such as acetylenyl, propargyl, butynyl, or
pentynyl). Alternatively, Rr is unsubstituted
or substituted aryl (including, but not limited to, monocyclic or bicyclic
aryl) or unsubstituted or substituted
arylalkyl (including, but not limited to, monocyclic or bicyclic aryl linked
to alkyl wherein alkyl includes, but is not
limited to, CH3, -CH2CH3, n-propyl, isopropyl, n- butyl, sec-butyl, and
pentyl). In some other embodiments, Rr is
unsubstituted or substituted heteroaryl, including, but not limited to,
monocyclic and bicyclic heteroaryl.
Monocyclic heteroaryl Rrincludes, but is not limited to, pyrrolyl, thienyl,
furyl, pyridinyl, pyranyl, pyrimidinyl,
pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl.
Bicyclic heteroaryl Rr includes, but is not
limited to, benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl,
benzimidazolyl, benzoxazolyl,
76

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl,
pyrrolo [1, 2-b]pyridazinyl,
pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl,
and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
[00325] Also provided herein are compounds of Formula (XXIII) wherein R1'
is unsubstituted or
substituted heteroarylalkyl, including, but not limited to, monocyclic and
bicyclic heteroaryl as described above, that
are linked to alkyl, which in turn includes, but is not limited to, CH3, -
CH2CH3, n-propyl, isopropyl, n- butyl, sec-
butyl, and pentyl. In some embodiments, R1' is unsubstituted or substituted
cycloalkyl (including, but not limited to,
cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted
heteroalkyl (non-limiting examples include
ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further
embodiments, R1' is unsubstituted or
substituted heterocyclyl which includes, but is not limited to, pyrrolidinyl,
tetrahydrofuranyl, piperidinyl,
tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and
piperazinyl. In yet other embodiments of the
compounds of Formula (XXIII), R1' is unsubstituted or substituted alkoxy
including, but not limited to, C1-C4alkoxy
such as methoxy, ethoxy, propoxy or butoxy. R1' can also be unsubstituted or
substituted heterocyclyloxy, including
but not limited to, 4-NH piperidin- 1 -yl-oxy, 4-methyl piperidin-1 -yl-oxy, 4-
ethyl piperidin- 1 -yl-oxy, 4-isopropyl-
piperidin- 1 -yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R1' is
unsubstituted or substituted amino,
wherein the substituted amino includes, but is not limited to, dimethylamino,
diethylamino, di-isopropyl amino, N-
methyl N-ethyl amino, and dibutylamino. In some embodiments, R1' is
unsubstituted or substituted acyl,
unsubstituted or substituted acyloxy, unsubstituted or substituted C1-
C4acyloxy, unsubstituted or substituted
alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or
substituted sulfonamido. In other
embodiments, R1' is halo, selected from ¨I, -F, -Cl, and -Br. In some
embodiments, R1' is selected from cyano,
hydroxyl, nitro, phosphate, urea, or carbonate. In some embodiments, R1' can
be -CH3, -CH2CH3, n-propyl,
isopropyl, n- butyl, tert- butyl, sec-butyl, pentyl, hexyl, heptyl, -OCH3, -
OCH2CH3, or -CF3.
[00326] In some embodiments, R1' of the compounds of Formula (XXIII) can
also be NR'R" wherein
R' and R" are taken together with the nitrogen to form a cyclic moiety having
from 3 to 8 ring atoms. The cyclic
moiety so formed may further include one or more heteroatoms which are
selected from S, 0, and N. The cyclic
moiety so formed is unsubstituted or substituted, including, but not limited
to, morpholinyl, azetidinyl, pyrrolidinyl,
piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl.
Further non-limiting exemplary cyclic
moieites are the following:
CH 3
rN
N
rS
1¨NO ¨N/ __ \ ,\--N "222,--N)
,3),N ¨1 \ /0
, -2-
0 0
,.,,,,,0C 0 ,0 H
....\---- I A,". /
,JS Nji---- ri\I-S\-\ CH3
¨N \;.I\1) 0
0 , , µ1' .
[00327] Also provided herein are compounds of Formula (XXIII), wherein
when R1' is alkyl, alkenyl,
alkynyl, cycloalkyl, heteroalkyl, heterocyclyl, heterocyclyloxy, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, acyl,
alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, or NR'R", (wherein
R' and R" are taken together with
77

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
nitrogen to form a cyclic moiety), wherein R1' is optionally substituted with
one or more of the following
substituents: alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocyclyl,
heterocyclyloxy, aryl, arylalkyl,
heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy,
alkoxycarbonyl, halo, cyano,
hydroxyl, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are
taken together with nitrogen to form a
cyclic moiety. Each of the above substituents can be further substituted with
one or more substituents chosen from
alkyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo,
cyano, hydroxyl, nitro, oxo, phosphate,
urea, and carbonate.
[00328] For example, provided herein are compounds wherein when R1' is
alkyl, the alkyl is substituted
with NR'R" wherein R' and R" are taken together with the nitrogen to form a
cyclic moiety. The cyclic moiety so
formed can be unsubstituted or substituted. Non-limiting exemplary cyclic
moieties include, but are not limited to,
morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, and
thiomorpholinyl. In other examples of the
compounds of Formula (XXIII), when R1' is alkyl, the alkyl is substituted with
heterocyclyl, which includes, but is
not limited to, oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolyl,
tetrahydropyranyl, piperidinyl, morpholinyl, and
piperazinyl. All of the above listed heterocyclyl substituents can be
unsubstituted or substituted.
[00329] In yet other examples of the compounds of Formula (XXIII), when
R1' is alkyl, the alkyl is
substituted with a 5, 6, 7, 8, 9, or 10 membered monocyclic or bicyclic
heteroaryl, which is unsubstituted or
substituted. In some embodiments, the monocyclic heteroaryl includes, but is
not limited to, pyrrolyl, thienyl, furyl,
pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl,
thiazolyl, pyrazolyl, and oxazolyl. The bicyclic
heteroaryl includes, but is not limited to, benzothiophenyl, benzofuryl,
indolyl, quinolinyl, isoquinolinyl,
benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl,
pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-
b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-
a]pyridinyl, and pyrrolo[1, 2-f][1, 2,
4]triazinyl.
[00330] In some embodiments of the compound of Formula (XXIII), L is -
N(R2)-, wherein R2 is
hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not
limited to -CH3, -CH2CH3, n-propyl,
isopropyl, n- butyl, tert- butyl, sec-butyl, pentyl, hexyl, and heptyl), or
unsubstituted or substituted C3-C7cycloalkyl
(which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl,
and cyclohexyl). In other embodiments
of the compound of Formula (XXIII), R2 is unsubstituted or substituted
heterocyclyl (which includes but is not
limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl,
piperidinyl, and piperazinyl), or
unsubstituted or substituted C2-C1oheteroalkyl (which includes but is not
limited to methoxyethoxy, methoxymethyl,
and diethylaminoethyl). Alternatively, R2 is unsubstituted or substituted
monocyclic heteroaryl (which includes but
is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl,
pyrazinyl, pyridazinyl, imidazolyl, thiazolyl,
pyrazolyl, and oxazoly1) or unsubstituted or substituted monocyclic aryl.
[00331] In some embodiments of the compound of Formula (XXIII), R1 is
¨0R1', wherein R1' is
hydrogen or alkyl. In another example of the compound of Formula (XXIII), R1
is ¨0-alkyl, where alkyl is
isopropyl.
[00332] In other embodiments of the compound of Formula (XXIII), R1 is -
NHR1', -N(CH3)R1', -
N(CH2CH3)R1', -N(CH(CH3)2)R1', or ¨0R1', wherein R1' is unsubstituted or
substituted heterocyclyl (nonlimiting
78

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
examples thereof include 4-NH piperidin-l-yl, 4-methyl piperidin- 1 -yl, 4-
ethyl piperidin- 1 -yl, 4-isopropyl-
piperidin- 1 -yl, and pyrrolidin-3-y1), unsubstituted or substituted
monocyclic aryl, or unsubstituted or substituted
monocyclic heteroaryl (including, but not limited to, pyrrolyl, thienyl,
furyl, pyridinyl, pyranyl, pyrimidinyl,
pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl.). In
one example, RI is ¨0-aryl, i.e. phenoxy.
In another example, RI is ¨0-(4-methyl)piperidin- 1-y1 or ¨0-(4-
isopropyl)piperidin- 1 -yl.
[00333] In some embodiments of compounds of Formula (XXIII), RI' is an
amido group of the
0 , R1,,
./.. =R1"
Formula
, where RI" and RI" are each independently hydrogen, alkyl, heteroalkyl,
alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, or RI"
and RI" are taken together with
nitrogen to form a cyclic moiety.
[00334]
Also provided herein are compounds of Formula (XXIII), wherein when RI' is
alkyl, alkenyl,
alkynyl, cycloalkyl, heteroalkyl, heterocyclyl, heterocyclyloxy, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, acyl,
alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, or NR'R" (wherein
R' and R" are taken together with
nitrogen to form a cyclic moiety), then RI' is optionally substituted with an
amido group of the Formula:
0 , R1,,
4-
'Rh' , where RI" and RI" are each independently hydrogen, alkyl, heteroalkyl,
alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, or RI"
and RI" are taken together with
nitrogen to form a cyclic moiety.
[00335]
When RI" and RI" are taken together with the nitrogen to form a cyclic moiety,
the cyclic
moiety can have, for example, from 3 to 8 ring atoms. The cyclic moiety so
formed can further include one or more
heteroatoms which are selected from S, 0, and N. In some embodiments, the
cyclic moiety so formed is
unsubstituted or further substituted with a group which is hydrogen, alkyl,
heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, thio, sulfoxide, sulfone, halo, cyano, hydroxyl,
nitro, phosphate, urea, or carbonate.
[00336]
In some embodiments of the compound of Formula (XXIII), RI is one of the
following
moieties:
CH3
/N¨CH3 I
/N1.-CH NH
,_,
0 \,,
. 0----) 0"---\/ lor13 0........======== _¨NO 0 __ <
v
i1/1-1,.
v.," ../VV I I I
1 1 1 1 1 '
"CH3
)
NCF13
101 --N' --N >. 1¨N\
N .1.,,õ
CH3
-CH(CH3)2 , H ,
, ,
79

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
CH3
I
1-N/ CH3
0 H3 N
C ,....
H3C
N 0
\
, I
CH, I. --
/ CH3 j HN--- IN ,,/
A
k--....õ.õ,,,N,....,.... 2
H3C0
, --
CI 0 I. CN
.....õ..--....... ..õõCH, H C
N H3C\
N -1-N( )0 3\t\
0 N \()
;-2N / \ __ /
`341, `Txn,
0
N Me0 N
I ;N S N).1----- H3C
\ 0 I ;IV rS
0 0 . r N A.--
N,)
1 ,
'>,_ N 1 1
, , ,
cH2cH3
N rN\ _ ,CN
.......,.. , 0
II /N N CN
N
- Nj1"--
--
J,
\? )
\c. 9)
1,')
, , , ,
o
OS 2Me
'N _ ,S02Me õ,.....N.-------/
0)
- N
0)
1
,I
_ OH
_...-.. ,.,
' N
I , --S
/\) I N I N --
S\
0---N I /2
1.-44--
-2;61'1' 'Lt,..Z-N )
'"2/,,--- N
, -I. , or
,-S\ /N
I /2¨N\._ j
N
0
/r\j N-Ac .--S02Me 0 N-So m
2 e
0---\) 0----) 7----\/
=AlV
I , I
0 - / _____________________________ 0
\ N
,CH3 N,Ac
NCH3
N
r-J
N i rp
r) ri r __
,3zco 22.-0 :24-0
, c-?2,- -c
, 4

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
,S02Me ,Ac
N\ ) ,S02Me
N\
/N--CH3
N __ /
rf r..1 N __ /
HN"¨\)
:%--o
WI,
/ 'rift
/ ' / /
r01---CF13 r.01-.'Ac
0 N ---*AC
HN"\) Hir\) (1--NH V¨NH
'r. , , 2. =
(:) ro (---,N_cH3 (N-AC
r) J.¨N,) r N ,) r.N
(.2c¨NH HN
i H N"--j
i HN---j
/ HN
i
O\, ? H I H
N
H ¨N,3
01 101 II\I 0
N,/s( szt.,N ,/s(
la, S ''2a '/"S \
0 A= 1.1
01 \ 0 0" 01
CN g
,12( N,/ \ * (:) ,N-\_ N¨µ
(:) / \ /--\
;$ N 0
0 N
0 , v \
0 CH3
CH3
[00337] In some embodiments of compounds of Formula (XXIII), the
compound is selected from:
R1 R1 R1
N
N N
(R5), (R5), ¨õ 1 (R5), ¨õ 1
I I I
Y, Y, Y,
Wd , Wd,
Wd,
R1 R1
i 1\1
(R ) N (R5),,
N X
I I
111 111
Wd, and Wd .
[00338] In some embodiments, the compound of Formula (XXIII) has the
Formula (XXIV):
R3 R1
. 1\1
X
I
111
Wd
81

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Formula (XXIV).
[00339] Alternatively, the compound has the Formula:
R3 R1 R1 R1 R3 R1
R3
0 N 40 N 0 N N
I
, ..--
õ,.......õ:;,), ... ..
X X R3
I I I
111 `1( `1( 111
Wd Wd Wd, or Wd
, , .
[00340] For example, the compound has the Formula (XXIIa):
li
01 N ==õ,..,.,p
-%
Wb5 Xi
I
Y
"d
(Formula XXIIa).
[00341] In some embodiments of compounds of Formula (XXI), (XXII or
(XXIIa), Wd
Nj, N N-,* y
X3 ,s3
is 'N R12
12 =
'N R12or --- Ri2 and R12 is hydrogen or halo. For example, Wd is
and R is
N-,1( x3
c=N
Cl-. In another instance, Wd is ' N R12 and R12 is hydrogen.
[00342] In other embodiments of compounds of Formula (XXI), (XXII or
(XXIIa), Wd is
aVVV
i,
1\1....N v N m
R10 , 's3 R10_C L,i...' 73
'N N
or and R1 is hydrogen or halo.
[00343] In still other embodiments of compounds of Formula (XXI), (XXII
or (XXIIa), Wd is
N...., N m y N..... N-N
/.3 "".., ,s3
_-.1\1
c--N (.--;-L- tr--1
'N N 'N N N
or . For example, Wd is or =
82

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00344] In some embodiments, the compound described herein is a compound
of Formula (XXI),
(XXII or (XXIIa), where Wb5 is N; X is -CH(R9)-, where R9 is methyl or ethyl;
Y is ¨NH-; and Wd is
N¨r\J N N m
or
../VV ../VV
RI
In some embodiments, Wd is N . In other embodiments, Wd is N .
In yet other
aVV
N
N
embodiments, Wd is N . In some embodiments, Wd is N
[00345] In one aspect, provided herein are compounds of Formula (XXV):
C F3 0
PO
Wb
Wb 1 Wb5 X
Wd
Formula (XXV)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or its
pharmaceutically acceptable forms thereof, wherein
Wbi and Wb2 are each independently CR3, S, 0, N or NR13, wherein at least one
of Wbi and Wb2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
Wb5 is Cle, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
B is hydrogen, alkyl, amino, heteroalkyl, cycloalkyl, heterocyclyl,
heterocyclylalkyl, aryl or
heteroaryl, each of which is substituted with 0-4 occurrences of R2;
X is absent or is ¨(CH(R9)),-;
83

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R9)-, -C(=0)-(CHR9),-, -C(=0)-, -
N(R9)-C(=0)-, or -
N(R9)-C(=0)NH-,-N(R9)C(R9)2-, or
each z is independently an integer of 1, 2, 3, or 4;
wherein when Wb5 is N, no more than one of X or Y is absent;
each R2 is independently alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl,
arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo,
cyano, hydroxyl, nitro, phosphate, urea or carbonate;
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R9 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
Wd is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more
Rb, R11 or R12;
wherein each Rb is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl,
alkynyl, cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate, or
NR'R", wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.
[00346] In certain embodiments, Wb5 is Cle. In some embodiments, R8 is
hydrogen.
[00347] In certain embodiments, Wb1 is CR3. In some embodiments, Wb1 is N.
In some embodiments,
Wb1 is S. In some embodiments, Wb1 is 0.
[00348] In certain embodiments, Wb2 is CR3. In some embodiments, Wb2 is N.
In some embodiments,
Wb1 is S. In some embodiments, Wb1 is 0.
[00349] In some embodiments, Wb1 and Wb2 are CR3. In some embodiments, Wb1
is S and Wb2 is CR3. In
some embodiments, Wb1 is S and Wb2 is N.
[00350] In certain embodiments, each R3 is independently hydrogen, alkyl,
alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl, heteroalkyl, alkoxy, amido, amino, acyl, acyloxy,
sulfinyl, sulfonyl, sulfoxide, sulfone,
sulfonamido, halo, cyano, heteroaryl, aryl, hydroxyl, or nitro. In some
embodiments, each R3 is independently
hydrogen, alkyl, cycloalkyl, heterocyclyl, fluoroalkyl, alkoxy, halo, cyano,
heteroaryl or aryl. In some
embodiments, R3 is hydrogen, alkyl, fluoroalkyl, alkoxy or aryl. In some
embodiments, each R3 is hydrogen.
[00351] In certain embodiments, p is 0. In some embodiments, p is 1.
[00352] In certain embodiments, B is aryl (e.g., 6-membered aryl)
substituted with 0-4 occurrences of R2.
In some embodiments, B is phenyl substituted with 0-4 occurrences of R2. In
some embodiments, B is phenyl
substituted with 0 occurrences of R2. In some embodiments, B is phenyl
substituted with 1 occurrence of R2. In
some embodiments, R2 is halo (e.g., fluoro).
84

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00353] In some embodiments, Xis -(CH(R9)),-. In some embodiments, z is 1.
In some embodiments, R9
is Ci_io alkyl (e.g., methyl).
[00354] In some embodiments, is absent, -0-, -NH(R9)-, or -S(=0)2-. In
certain embodiments, Y is absent.
In some embodiments, Y is -N(R9)-. In some embodiments, R9 is hydrogen.
;Ir
HN ,õss
[00355] In certain embodiments, X-Y is 5" ' . In some embodiments, X-Y
is -CH2-N(CH3). In some
embodiments, X-Y is (S)-CH(CH3)-NH-. In some embodiments, X-Y is (R)-CH(CH3)-
NH-.
[00356] In some embodiments, Wd is aryl. In some embodiments, Wd is
heteroaryl (e.g., a bicyclic
heteroaryl). In some embodiments, Wd is selected from:
IN jv N-----:\ -^hivi
Rb N b :IrN N b ;s5SL(NH A N
NKR 1
---N
/.=....
R12 --- N
N¨ NN
R11 R11 R12 R11
R12 R12
N=----\
cs.ssi\l Rb
,
N¨ ---- N1 N¨ ---- Nil
NN
,.
I R12 N
R11 R11 R12 R11
vuv R12
N \ / \ -, I N
N I N N i
R12 ----N 101 CN-N I. )¨NH
H2N R12
1\1 NH2 ,csss,N ,csss N ,css',1\( ,css',1\L NH2
01
A\1N 1\1 1\1
Ri2¨er S R12 C
r F 3 ....--.....N
µ S \ NI-I
JIM/
N .---- N NN NLN-N Nr:::-;\"N
,-I ,--.1/ N
R12 s R _ - -**.-N/'...- 12N "...-... R121" 1R, -
,N"*".....L.)
N N N-1\1.//
%/VW
Njr----N\ csss., ,N NH2
¨ ::-T-
N ' N.--N
--:-.2'rN_R12 m
1,,J ,N N)'/ N, N -I )..,..,..../.... N
R ,_ N Ri2''
N

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
JVV,/ ./VVV OW/
Ri 2 R1 N
N \i<,-N 11
N \--N
\
õ):,-...
N S N R1 2 N - Ni R1 L N
=ftn.", R12
JVIIV %NV,/
N'iN)_R12
I
1.:õ.
N ,õ..-.._, R I 4 ,,,..-N
õ,..1...¨./ N N
and H .
N-:---- \
1
N N
1 ,N-
...1
[00357] In some embodiments, Wd is R12
. In other embodiments, Wd is R12 N
.11AILI ..A1111/
-,,
In some embodiments, Wd is R1 2 N
1 2 'L- -**--'
. In some embodiments, Wd 1S R N S . In certain
WON uw
N N,_ R12 NN
1 j 1
,... ..õ--õ, ...._..;,..,--
. o1 2
N
embodiments, Wd is N S . In some
embodiments, Wd is . In certain embodiments,
JIM/
R1 N
1
N
Wd is N . In certain embodiments, Wd is . In certain embodiments,
Wd is
"wi R12
cSSS N Rb
-.....õ... ........T...
1 R12 N
------/- ----- N
CA-T..--
R12Th.:1-N 1 ,
N N
.....:_µ.. .õ,..,c.,
R11 N H
. In certain embodiments, Wd is . In some embodiments, Wd 1S .
41./NAI
/L
N Ni \--N
R12 NN N
,õ.1... .õõ)......> ,N--.1/
In some embodiments, Wd is RI G N
. In some embodiments, Wd is . In some
41.01"/
Ni."-'...%N
,N--
12N
embodiments, Wd is R .
[00358] In certain embodiments, R12 is hydrogen, alkyl, alkoxy,
hydroxyl, haloalkyl, halo, cyano,
amino or amido. In some embodiments, R12 is hydrogen. In some embodiments, R12
is halo (e.g., chloro or fluoro).
In some embodiments, R12 is cyano. In some embodiments, R12 is amino. In some
embodiments, R12 is amido (i.e.,
86

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
-C(=0)NH2). In some embodiments, R12 is alkyl (e.g., methyl). In some
embodiments, R12 is haloalkyl (e.g.,
trifluoromethyl). In some embodiments, R12 is alkoxy (e.g., methoxy). In some
embodiments, R12 is hydroxyl.
[00359] In certain embodiments, R11 is hydrogen or amino. In some
embodiments, R11 is amino.
[00360] In
certain embodiments, Rb is hydrogen or amino. In some embodiments, Rb is
hydrogen. In
some embodiments, Rb is amino.
N--r-- A
NH
.csssLIr
N:::----\
I
;ssLrLr N H N N
I I
N
[00361] In some embodiments, Wd is N \./ . In some
embodiments, Wd is NH2 .
N:-,---- \
-cssr NH
VIA",
I
N N .---:N
I zN.f
In some embodiments, Wd is OH . In some
embodiments, Wd is N . In some embodiments,
J1Art/
aVV,/
.---:"----Cr¨_, \
õ, N
Wd is N . In some embodiments, Wd is N . In some
embodiments, Wd is U
=
Jvvv
.1V1"/
N N
I ,
..õ1:-..... ,.....,
1.., õ.õ--....s
H2N N s
In some embodiments, Wd is N . In some
embodiments, Wd is . In some
N N
S S
embodiments, Wd is CI N . In some embodiments, Wd is
N . In some embodiments, Wd
.A.A."/Juvv
N
N N
N
I I
l:,... õ.....¨,...õ7-
is N . In some embodiments, Wd iS CI N . In some
embodiments, Wd is
VVVV
JVVV
N
N N N
..õ.1..;:z. õõ--.....õ_,7-- ...,,I....... ,.....:,7-1
H2N N . In some embodiments, Wd is Me N . In some embodiments,
Wd is
%Ann/
`.SS N1 -.s'
I\1
2
NN &)
N NC N
I
-... ..õ.....õ:õ...7 F \ \
HO N . In some embodiments, Wd is NH
. In some embodiments, Wd is NH =
87

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
.ss
0
NH 2
N
N
H 2 N ¨NH F3C
In some embodiments, Wd is . In some embodiments, Wd is . In some
N
N H2
N
NNf
embodiments, Wd is . In some embodiments, Wd is N .
In some embodiments, Wd is
JVVV JVVV
N
NN
. In some embodiments, Wd is N . In some embodiments, Wd is N .
In some
JVVV N
NC
NC N
embodiments, Wd is N . In some embodiments, Wd is
NH2 . In some embodiments, Wd is
JVVV
I
[00362] In some embodiments of the compound of Formula (XXV), at least
one of R11 and R12 is
hydrogen, cyano, halo, unsubstituted or substituted alkyl, unsubstituted or
substituted alkynyl, or unsubstituted or
substituted alkenyl. In some embodiments, at least one of R11 and R12 is
unsubstituted or substituted aryl. In some
embodiments, at least one of R11 and R12 is unsubstituted or substituted
heteroaryl, which includes, but is not limited
to, heteroaryl having a 5 membered ring, heteroaryl having a six membered
ring, heteroaryl with at least one
nitrogen ring atom, heteroaryl with two nitrogen ring atoms, monocylic
heteroaryl, and bicylic heteroaryl. In some
embodiments, at least one of R11 and R12 is unsubstituted or substituted
heterocyclyl, which includes, but is not
limited to, heterocyclyl with one nitrogen ring atom, heterocyclyl with one
oxygen ring atom, heterocyclyl with one
sulfur ring atom, 5 membered heterocyclyl, 6 membered heterocyclyl, saturated
heterocyclyl, unsaturated
heterocyclyl, heterocyclyl having an unsaturated moiety connected to the
heterocyclyl ring, heterocyclyl substituted
by oxo, and heterocyclyl substituted by two oxo. In some embodiments, at least
one of R11 and R12 is unsubstituted
or substituted cycloalkyl, including, but not limited to, cyclopropyl,
cyclobutyl, cyclopentyl, cyclohexyl, cycloalkyl,
each of which can be substituted by one oxo, and cycloalkyl having an
unsaturated moiety connected to the
cycloalkyl ring. In some embodiments, at least one of R11 and R12 is
unsubstituted or substituted amido,
unsubstituted or substituted acyloxy, unsubstituted or substituted
alkoxycarbonyl, unsubstituted or substituted acyl,
or unsubstituted or substituted sulfonamido.
[00363] In some embodiments, when at least one of R11 and R12 is alkyl,
alkynyl, alkenyl, aryl,
heteroaryl, heterocyclyl, cycloalkyl, alkoxycarbonyl, amido, acyloxy, acyl, or
sulfonamido, it is substituted with one
or more of alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl,
aryl, heteroaryl, alkoxy, amido, amino, acyl,
88

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl or nitro, each of
which alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, or
sulfonamido can itself be substituted.
[00364] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula (XXVI):
cF3 0
0
--
x
1
R8 Y
Wd
Formula (XXVI). .
[00365] In some embodiments, the compound of Formula (XXVI) has a
structure of Formula (XXVIa) or
(XXVIb):
CF3 0 CF3 0
B B
N N
le 1 le
x x
1 1
R8 Y R8 Y
Wd or Wd
Formula (XXVIa) Formula (XXVIb).
[00366] In some embodiments, the compound of Formula (XXVI) has a
structure of Formula (XXVIIa) or
(XXVIIb):
CF3 0 CF3 0
,
eB , B
N N
l R9 10 1
R9
_
_ z
R8 `? R8 )1
Wd or Wd
Formula (XXVIIa) Formula (XXVIIb).
[00367] In some embodiments of compounds of Formula (XXVIa), (XXVIb),
(XXVIIa), or (XXVIIb),
B is aryl substituted with 0-3 occurrences of R2. For example, B is phenyl
substituted with 0-3 occurrences of R2. In
some embodiments of compounds of Formula (XXVIa), (XXVIb), (XXVIIa), or
(XXVIIb), B is unsubstituted
phenyl. In other embodiments of compounds of Formula (XXVIa), (XXVIb),
(XXVIIa), or (XXVIIb), B is phenyl
substituted with 1 occurrence of R2. R2 is, in some instances, halo or alkyl.
In other embodiments of compounds of
Formula (XXVIa), (XXVIb), (XXVIIa), or (XXVIIb), B is cycloalkyl or
heterocyclyl.
[00368] In still other embodiments, the compound of Formula (I) has a
structure selected from:
89

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
CFO C F3 0 CFO3
H N ,B R6 ,B H N ,B
N
I N.\.yH R6N/1 NX
HNN H
Wd Wd , or Wd
[00369] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula (XXVIII):
0
F3C
,B
Swb5 x
Wd
Formula (XXVIII).
[00370] In some embodiments, the compound of Formula (XXVIII) has a
structure of Formula (XXIX):
0
F3C
1\(B
R9
Wd
Formula (XXIX).
[00371] In certain embodiments, the compound of Formula (XXV) has a
structure of Formula (XXX):
0
F3C
,B
tf,Nj
Swb5 x
Wd
Formula (XXX).
[00372] In certain embodiments, the compound of Formula (XXX) has a
structure of Formula (XXXI):

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
F3C 0
N,B
wb5--\, R9
Y,
Wd
Formula (XXXI).
[00373] In certain embodiments, the compound of Formula (XXX) has a
structure of Formula (XXXII):
F3C 0
N B
/ I
Y,
Wd
Formula (XXXII).
[00374] In some embodiments, the compound of Formula (XXXII) has a
structure of Formula (XXXIII):
0
F3C
N, B
R9
Y,
Wd
Formula (XXXIII).
[00375] In another aspect, provided herein is a compound of Formula
(XXXIV) or (XXXV):
CF3 CF3
R3
N/ R1 N
PO PO
Wb2
W1,1 wo y w wo
Wd or Wd
Formula (XXXIV) Formula (XXXV)
or an enantiomer, a mixture of enantiomers, or a mixture of two or more
diastereomers thereof, or their
pharmaceutically acceptable forms thereof, wherein:
Wbi and Wb2 are each independently CR3, S, 0, N or NR13, wherein at least one
of Wbi and Wb2 is
CR3, N or NR13;
p is 0, 1, 2 or 3;
91

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Wb5 is CR8, CHR8, or N;
R8 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, alkoxy,
amido, amino, acyl,
acyloxy, sulfonamido, halo, cyano, hydroxyl, or nitro;
R1 is ¨(L)-R1';
L is a bond, -S-, -N(R15)-, -C(=0)-, or ¨0-;
R1' is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
cycloalkylalkyl, heterocyclyl,
heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl, nitro, phosphate, urea,
carbonate, substituted nitrogen, or
NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic
moiety;
each R15 is independently hydrogen, alkyl, cycloalkyl, or heteroalkyl;
X is absent or is ¨(CH(R16)),;
Y is absent, -0-, -S-, -S(=0)-, -S(=0)2-, -N(R16)-, -C(=0)-(CHR16),-, -C(=0)-,
-N(R16)-C(=0)-, or
-N(R16)-C(=0)NH-,-N(R16)C(R16)2-, or -C(=0)-N(R16)-(CHR16)z-;
each z is an integer of 1,2, 3, or 4;
each R3 is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, fluoroalkyl,
heteroalkyl, alkoxy, amido, amino, acyl, acyloxy, sulfinyl, sulfonyl,
sulfoxide, sulfone, sulfonamido, halo, cyano,
heteroaryl, aryl, hydroxyl, or nitro;
each R13 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl or
heteroalkyl;
each R16 is independently hydrogen, alkyl, cycloalkyl, heterocyclyl,
heteroalkyl, aryl, halo or
heteroaryl;
Wd is heterocyclyl, aryl, cycloalkyl, or heteroaryl which is optionally
substituted with one or more
Rb, R11 or R12;
wherein each Rb is independently hydrogen, halo, phosphate, urea, carbonate,
alkyl, alkenyl,
alkynyl, cycloalkyl, amino, heteroalkyl, or heterocyclyl; and
each R11 and R12 is independently hydrogen, alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl,
heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy,
heterocyclyloxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, sulfonamido, halo, haloalkyl, cyano, hydroxyl, nitro,
phosphate, urea, carboxylic acid, carbonate, or
NR'R", wherein R' and R" are taken together with nitrogen to form a cyclic
moiety.
[00376] In certain embodiments, Wbi is CR3 and Wb2 is CR3.
[00377] In certain embodiments, Wb5 is CH.
[00378] In certain embodiments, L is a bond, -N(R15)- or
[00379] For all structures disclosed herein, the following
embodimentsapply to their corresponding
variable positions.
[00380] In some embodiments of the compounds disclosed herein, B is aryl
or heteroaryl substituted
with 0 or 1 occurrences of R2 and Cy is a 5- or 6-membered aryl or heteroaryl
group. For example, B is aryl
substituted with 0 or 1 occurrences of R2 and Cy is a 5- or 6-membered aryl or
heteroaryl group. In another example,
92

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
B is aryl or heteroaryl substituted with 0 or 1 occurrence of R2 and Cy is a 6-
membered aryl group. Cy is, for
instance, phenyl substituted with alkyl, fluoroalkyl, aryl, heteroaryl or
halo.
[00381] In other embodiments of the the compounds disclosed herein, B is
aryl or heteroaryl
substituted with 0 or 1 occurrences of R2, Cy is a 5- or 6-membered aryl or
heteroaryl group, X is ¨(CH(R9)),- and Y
is -NH(R9)-, wherein each R9 is chosen independently from hydrogen, alkyl,
cycloalkyl, and heteroalkyl. For
example, X-Y is -CH2(CH3)-NH-. In some embodiments, B is aryl substituted with
0 or 1 occurrences of R2 and Cy
is a 5- or 6-membered aryl or heteroaryl group. In another example, B is aryl
or heteroaryl substituted with 0 or 1
occurrence of R2 and Cy is a 6-membered aryl group. Cy is, for instance,
phenyl substituted with alkyl, fluoroalkyl,
aryl, heteroaryl or halo.
[00382] In some embodiments, B is cycloalkyl, heterocyclyl, aryl, or
heteroaryl, which is substituted
with 0-4 occurrences of R2, R3 is H, halo, alkyl, alkoxy, aryl, cycloalkyl, or
heteroaryl, each R5 is H, halo, alkyl,
alkoxy, aryl, cycloalkyl, or heteroaryl, and R9 is hydrogen or alkyl.
[00383] In some embodiments, B is unsubstituted or substituted alkyl,
including, but not limited to, ¨
(CH2)2-NRalta, wherein each Ra is independently hydrogen, alkyl, fluoroalkyl,
cycloalkyl, cycloalkylalkyl, aryl,
aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, or
NRalta are combined together to form a
cyclic moiety, which includes but is not limited to piperidinyl, piperazinyl,
and morpholinyl. In some embodiments,
B is unsubstituted or substituted amino. In some embodiments, B is
unsubstituted or substituted heteroalkyl.
[00384] In some embodiments, B is selected from unsubstituted or
substituted aryl, including, but not
limited to, unsubstituted or substituted phenyl; unsubstituted or substituted
heteroaryl including, but not limited to,
pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyrimidin-4-yl, pyrimidin-2-yl,
pyrimidin-5-yl, or pyrazin-2-yl,
unsubstituted or substituted monocyclic heteroaryl, unsubstituted or
substituted bicyclic heteroaryl, heteroaryl
having two heteroatoms as ring atoms, unsubstituted or substituted heteroaryl
comprising a nitrogen ring atom,
heteroaryl comprising two nitrogen ring atoms, heteroaryl having a nitrogen
and a sulfur as ring atoms,
unsubstituted or substituted heterocyclyl including, but not limited to,
morpholinyl, tetrahydropyranyl, piperazinyl,
and piperidinyl, unsubstituted or substituted cycloalkyl including, but not
limited to, cyclopentyl and cyclohexyl.
[00385] In some embodiments, B is unsubstituted or substituted with one
or more R2 substituents. In
some embodiments, R2 is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, heteroaryl alkoxy,
amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano,
hydroxyl or nitro, each of which alkyl,
heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl,
alkoxy, amido, amino, acyl, acyloxy, or
sulfonamido, can itself be substituted.
[00386] In some embodiments, R2 is unsubstituted or substituted alkyl,
unsubstituted or substituted
heteroalkyl, unsubstituted or substituted alkenyl, unsubstituted or
substituted alkynyl, unsubstituted or substituted
cycloalkyl, or unsubstituted or substituted heterocyclyl. In some embodiments,
R2 is unsubstituted or substituted
aryl, unsubstituted or substituted arylalkyl, unsubstituted or substituted
heteroaryl, or unsubstituted or substituted
heteroarylalkyl. In some embodiments, R2 is unsubstituted or substituted
alkoxy, unsubstituted or substituted amido,
unsubstituted or substituted amino. In some embodiments, R2 is unsubstituted
or substituted acyl, unsubstituted or
substituted acyloxy, unsubstituted or substituted alkoxycarbonyl, or
unsubstituted or substituted sulfonamido. In
93

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
some embodiments, R2 is halo, selected from ¨I, -F, -Cl, or -Br. In some
embodiments, R2 is selected from cyano,
hydroxyl, nitro and a carbonate. In some embodiments, R2 is unsubstituted or
substituted phosphate. In some
embodiments, R2 is unsubstituted or substituted urea. In some embodiments,
when R2 is alkyl, R2 is methyl, ethyl,
propyl, isopropyl, n- butyl, tert- butyl, sec-butyl, pentyl, hexyl or heptyl.
[00387] In some embodiments, when R2 is alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, or
hydroxyl, it is substituted by phosphate, substituted by urea, or substituted
by carbonate.
[00388] In some embodiments, when R2 is alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl,
aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, or sulfonamido, it
is substituted by one or more of alkyl, heteroalkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aryl, heteroaryl,
alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano,
hydroxyl or nitro, each of which
alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
heteroaryl, alkoxy, amido, amino, acyl, acyloxy,
alkoxycarbonyl, or sulfonamido can itself be substituted.
[00389] In some embodiments, there are no occurrences of R2. In other
embodiments, there is one
occurrence of R2. In still other embodiments, there are two occurrences of R2.
In yet other embodiments, there are
three occurrences of R2. In yet other embodiments, there are four occurrences
of R2. For example, in some
embodiments B is aryl or heteroaryl and there there are no occurrences of R2.
In other instances, B is aryl or
heteroaryl and there there is one occurrence of R2 where R2 is alkyl or halo.
[00390] In some embodiments, R3 is halo, alkyl, alkoxy, heteroaryl, or
cycloalkyl. For example, R3 is
H, CH3, CH2CH3, CF3, Cl, or F. In other embodiments, R3 is CH3, CF3, or Cl. In
other embodiments, each R3 and R8
is independently selected from H, CH3, OCH3, CF3, and halo.
[00391] In some embodiments, R3 is unsubstituted or substituted alkyl,
unsubstituted or substituted
alkenyl, or unsubstituted or substituted alkynyl. In some embodiments, R3 is
unsubstituted or substituted aryl,
unsubstituted or substituted heteroaryl, unsubstituted or substituted
cycloalkyl, or unsubstituted or substituted
heterocyclyl. In some embodiments, R3 is unsubstituted or substituted alkoxy,
unsubstituted or substituted amido,
unsubstituted or substituted amino. In some embodiments, R3 is unsubstituted
or substituted acyl, unsubstituted or
substituted acyloxy, unsubstituted or substituted alkoxycarbonyl, or
unsubstituted or substituted sulfonamido. In
some embodiments, R3 is halo, selected from ¨I, -F, -Cl, or -Br. In some
embodiments, R3 is halo, alkyl, alkoxy,
heteroaryl, or cycloalkyl. For example, R3 is CH3, CH2CH3, CF3, Cl, or F. In
some instances, R3 is CH3, CF3, or Cl.
[00392] In some embodiments, R3 is selected from cyano, hydroxyl, and
nitro. In some embodiments,
when R3 is alkyl, R3 is methyl, ethyl, propyl, isopropyl, n- butyl, tert-
butyl, sec-butyl, pentyl, hexyl or heptyl. In
some embodiments, R3 is -CF3, -CH2F or ¨CHF2.
[00393] In some embodiments, when R3 is alkyl, alkenyl, alkynyl, aryl,
heteroaryl, cycloalkyl,
heterocyclyl, alkoxy, amido, amino, acyl, acyloxy, alkoxycarbonyl,or
sulfonamido, it is substituted with one or more
of alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
heteroaryl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl or nitro, each of
which alkyl, heteroalkyl, alkenyl,
94

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, or
sulfonamido can itself be substituted.
[00394] In some embodiments, R3 is a 5-membered heteroaryl group. Such
groups include, for
example, pyrrole, furan, thiophene, triazole, oxazole, pyrazole, and
isoxazole. In other embodiments, R3 is a 5-
membered nonaromatic heterocycle, including, but not limited to, oxazoline and
oxazolidinone. In still other
embodiments, R3 is a 6-membered heteroaryl group including pyridine, pyrazine,
pyrimidine and pyridazine.
Alternatively, R3 is a 6-membered nonaromatic heterocycle, including moieties
such as morpholino or piperidino. In
other embodiments, R3 is a fused 5/6-bicyclic heteroaryl, for example
benzothiazole, benzoxazole, benzisoxazole,
indazole, benzimidazole, benzothiophene, indole, isoindole, purine, or
pyrazolopyrimidine. In yet other
embodiments, R3 is a fused 5/6-bicyclic nonaromatic heterocycle.
[00395] In some embodiments, R3 is a C1-C6 alkyl group substituted with a
5-membered heteroaryl, a
5-membered heterocycle, a 6-membered heteroaryl, a 6-membered nonaromatic
heterocycle, a fused 5/6-bicyclic
heteroaryl, or a fused 5/6-bicyclic nonaromatic heterocycle. Alternatively, R3
is amino, sulfinyl, sulfonyl, sulfoxide,
sulfone, or alkoxy wherein the N, S, and 0 heteroatom has a covalent bond
either directly or through a C1-C6 alkyl
group to a 5-membered heteroaryl, a 5-membered nonaromatic heterocycle, a 6-
membered heteroaryl, a 6-
membered heterocycle, a fused 5/6-bicyclic heteroaryl, or a fused 5/6-bicyclic
heterocycle.
[00396] In other embodiments, R3 is a C1-C6 alkyl group substituted with
a fused polycyclic group,
wherein the polycyclic group has greater than two rings and is carbocyclic or
heterocyclic; C1-C6 alkyl group
substituted with a bridged cycloalkyl or bridged heterocyclic group; C1-C6
alkyl group substituted with a spirocyclic
cycloalkyl or spirocyclic heterocyclic group; or branched C4-C12 alkyl group,
wherein said branched alkyl group
contains at least one terminal t-butyl group.
[00397] Each of the embodiments named above for R3 is unsubstituted or
optionally additionally
substituted with an alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl,
heterocyclyl, aryl, heteroaryl, alkoxy, amido,
amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl or
nitro group.
[00398] In certain embodiments, R3 is a substituted or unsubstituted
heterocyclyl or heteroaryl group
selected from pyridine, pyrazole, piperazine, and pyrrolidine, wherein the
substituent can be a C1-C6 alkyl group or a
halogen.
[00399] In some embodiments, a compound is provided wherein R3 is
selected from a 5-membered
heteroaryl such as pyrrole, a furan, and a thiophene group; 5-membered
heterocycle such as pyrrolidine, a
tetrahydrofuran, or a tetrahydrothiophene group; 6-membered heteroaryl such as
pyridine, pyrazine, pyrimidine, and
pyridazine; 6-membered heterocycle such as piperidine, tetrahydropyran, and
thiane; and fused 5/6-bicyclic
heteroaryl such as indole, isoindole, benzofuran, isobenzofuran,
benzothiophene, benzimidazole, indazole,
benzoxazole, benzisoxazole, and purine. In certain embodiments, R3 is a
substituted or unsubstituted group selected
from pyridine, pyrazole, piperazine, and pyrrolidine. By way of non-limiting
example, the R3 group can be
substituted with a C1-C6 alkyl group or a halogen. For example, the R3 group
can be substituted with a methyl
group.
[00400] In some embodiments, a compound is provided wherein R3 is
selected from

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
N
r N rNN

R RN RN
RI R N Nyi\I
JVW . JVVV JVVV JVVV JVVV
ri N NN R R
R RNI RI R ii N
H
)R'
JVVV JVVV JVVV all At JVVV
R
NR N¨NR N¨NR INH N R C j

r N ce N
,,...õ)
N
I
JUN/ JVVV JVNIV JVW
0
R¨\)R IIµN R r¨\\/\1 N=/R
NII N- N'- X
N
\=1=1 X = NR, S, 0
I I I I
wherein R is H, C1-C6 alkyl, C1-C6 alkoxy, halo and haloalkyl. In certain
embodiments, R is methyl. In other
embodiments, a compound is provided wherein le is selected from:
HN
/
N¨N N =--, N
:,..) 0 0
0 0, 0 I\
JNAA1 .AINI ~AI JVV1/ ~jv WV,/ JVVLf
JVVV
F----- .---
N/
N¨N N N¨NH N¨N
N¨N N¨N
U U y
alIV1/ JVVV avvy alrLIV11 JYtAl
JVVV JVVV JNINA1
oo
F
F NF NN N F3CN i\IL Nk
i\ILN
y
JVVV 0.1%1V JUlAl ~IV JVVV JVVV NW JIIIIII
NC)
.
96

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00401] In some embodiments, each R5 is independently unsubstituted or
substituted alkyl (including,
but not limited to, unsubstituted or substituted C1-C4alkyl). In some
embodiments, each R5 is independently
unsubstituted or substituted alkenyl including, but not limited to,
unsubstituted or substituted C2-05alkenyl. In some
embodiments, each R5 is independently unsubstituted or substituted alkynyl
including, but not limited to,
unsubstituted or substituted C2-05alkynyl. In some embodiments, each R5 is
independently unsubstituted or
substituted cycloalkyl including, but not limited to, unsubstituted or
substituted C3-05cycloalkyl. In some
embodiments, each R5 is independently unsubstituted or substituted
heterocyclyl. In some embodiments, each R5 is
independently unsubstituted or substituted heteroalkyl including, but not
limited to, unsubstituted or substituted C1-
C4heteroalkyl. In some embodiments, each R5 is independently unsubstituted or
substituted alkoxy including, but
not limited to, unsubstituted or substituted C1-C4alkoxy. In some embodiments,
each R5 is independently
unsubstituted or substituted amido including, but not limited to,
unsubstituted or substituted C1-C4amido. In some
embodiments, each R5 is independently unsubstituted or substituted amino. In
some embodiments, each R5 is
independently unsubstituted or substituted acyl, unsubstituted or substituted
acyloxy, unsubstituted or substituted
C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or
substituted sulfonamido, or
unsubstituted or substituted C1-C4sulfonamido. In some embodiments, each R5 is
independently halo, selected from
¨I, -F, -Cl, and -Br. In some embodiments, each R5 is independently selected
from cyano, hydroxyl, and nitro. In
some other embodiments, each R5 is independently -CH3, -CH2CH3, n-propyl,
isopropyl, -OCH3, -OCH2CH3, or -
CF3.
[00402] In some embodiments, when R5 is alkyl, alkenyl, alkynyl,
cycloalkyl, heteroalkyl, acyl, alkoxy,
amido, amino, acyloxy, alkoxycarbonyl, or sulfonamido, R5 is independently
optionally substituted with one or more
of alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl,
heteroaryl, alkoxy, amido, amino, acyl,
acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxyl or nitro, each of
which alkyl, heteroalkyl, alkenyl,
alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy, amido, amino,
acyl, acyloxy, alkoxycarbonyl, or
sulfonamido can itself be substituted.
[00403] In some embodiments, no R5 moieties are present.
[00404] In some embodiments, X is absent. In some embodiments, X is
¨(CH(R9)),-, and z is an integer
of 1, 2, 3 or 4.
[00405] In some embodiments, R9 is unsubstituted or substituted alkyl
including, but not limited to,
unsubstituted or substituted C1-C1oalkyl. In some embodiments, R9 is
unsubstituted or substituted cycloalkyl
including, but not limited to, unsubstituted or substituted C3-C7cycloalkyl.
In some embodiments, R9 is ethyl,
methyl or hydrogen. In some embodiments, R9 is unsubstituted or substituted
heterocyclyl including, but not limited
to, unsubstituted or substituted C2-C10heteroalkyl. In some embodiments, R9 is
unsubstituted or substituted
heteroalkyl including, but not limited to, unsubstituted or substituted C2-
C1oheteroalkyl.
[00406] Also provided herein are compounds wherein R9 is hydrogen, and X
is -CH2-, -CH2CH2-, -
CH2CH2CH2-, -CH(CH3)-, or -CH(CH2CH3)-. In other embodiments, X is ¨(CH(R9)),-
, R9 is not hydrogen, and z is
an integer of 1. When X is-CH(R9)- and R9 is not hydrogen, then the compound
can adopt either an (S)- or (R)-
stereochemical configuration with respect to the CH carbon. In some
embodiments, the compound is a racemic
97

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
mixture of (S)- and (R) isomers with respect to the CH carbon. In other
embodiments, provided herein is a mixture
of a given disclosed compound wherein individual compounds of the mixture
exist predominately in an (S)- or (R)-
isomeric configuration. For example, the compound mixture has an (S)-
enantiomeric excess of greater than about
55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about
90%, about 95%, about 96%,
about 97%, about 98%, about 99%, about 99.5%, or more at the X carbon. In
other embodiments, the compound
mixture has an (S)-enantiomeric excess of greater than about 55% to about
99.5%, greater than about about 60% to
about 99.5%, greater than about 65% to about 99.5%, greater than about 70% to
about 99.5%, greater than about
75% to about 99.5%, greater than about 80% to about 99.5%, greater than about
85% to about 99.5%, greater than
about 90% to about 99.5%, greater than about 95% to about 99.5%, greater than
about 96% to about 99.5%, greater
than about 97% to about 99.5%, greater than about 98% to greater than about
99.5%, greater than about 99% to
about 99.5%, or more.
[00407] In other embodiments, the compound mixture has an (R)-
enantiomeric excess of greater than
about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%,
about 90%, about 95%, about
96%, about 97%, about 98%, about 99%, about 99.5%, or more at the X carbon. In
some other embodiments, the
compound mixture has an (R)-enantiomeric excess of greater than about 55% to
about 99.5%, greater than about
about 60% to about 99.5%, greater than about 65% to about 99.5%, greater than
about 70% to about 99.5%, greater
than about 75% to about 99.5%, greater than about 80% to about 99.5%, greater
than about 85% to about 99.5%,
greater than about 90% to about 99.5%, greater than about 95% to about 99.5%,
greater than about 96% to about
99.5%, greater than about 97% to about 99.5%, greater than about 98% to
greater than about 99.5%, greater than
about 99% to about 99.5%, or more.
[00408] In some embodiments of the compounds disclosed herein, X is -
CH(R9)-, R9 is methyl or ethyl,
and the compound is the (S)- isomer.
[00409] In some embodiments of the compounds disclosed herein, Y is
absent.
[00410] In some embodiments, Y is -0-, -S-, -S(=0)-, -S(=0)2-, -C(=0)-, -
N(R9)(C=0)-, -
N(R9)(C=0)NH-, -N(R9)C(R9)2- (such as-N(R9)CH2-, including, but not limited
to, -N(CH3)CH2-, N(CH(CH3)2)CH2
- or N(CH2CH3)CH2-), -N(R9)-, -N(CH3)-, -N(CH2CH3)-, or -N(CH(CH3)2)-. In
some embodiments, Y is -C(=0)-
(CHR9),- and z is an integer of 1, 2, 3, or 4.
[00411] In some embodiments, at least one of X and Y is present. In some
embodiments of the the compounds
disclosed herein, -XY- is -CH2-, -CH2-N(CH3), -CH2-N(CH2CH3), -CH(CH3)-NH-,
(S) -CH(CH3)-NH-, or (R)
-CH(CH3)-NH-. In other embodiments, X-Y is -N(CH3)_CH2-, N(CH2CH3) CH2-, -
N(CH(CH3)2)CH2-, or -NHCH2-.
[00412] In some embodiments, B is one of the following moieties:
CI
1 1-0 1- -K>
-C H 3 -CH2CH3 -CH(CH3)2 \N
0 .......õ,N....,.CH,
H3C
...k."..,...........õõN,....õ..õ...-
...k.----.,õ....õ.õN .õ...,.... caez 40 1
......(222õ,... ......
N
98

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
H3C op cl H3c oCH3
'4%, A "A NO2
N
0
A 0 A 0 C
µ el A 0 _,õ IN ,,,c0 = \\NI ----
CN
0
CN F
0 F3
1 , 1
1
`%_N--0
-
`%,N `%.1\r µe I
1
I O01 N\N el \ N
/
1\1/
0
..,N I-12
1 I 1
µe µ1\1N H2 µz2z.1\r CN
0
X-)Lc)\Z" Ar' d,
`2k. N \ - N H 2 CF 3 µ 0
N N ---" =-==== 7,0H
-CN `zzz. \ hl 0 \
I I r r-N-
ir,,,C, frN,y0,, irõ...õNH2 (N, (N,),N., r.N7.N,
_esc..,, N,, (NyN,....) rN,y0
1 , 1 , 1 ,
\----CN `tcN) `t,a, \'N `t(CN 2' N V-.1' N V--..1.'
N
/ \
1-N\ I
N 0ro
N N N Na.-. N Nl D N 0
\ N
jN -(
\ N
N[) \ \l----- N\
/( NX ( D' N.., N.,/, N r-J,,, N Z-D/ N
N-7¶-
il T T
'µt, N
\ \
99

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0).,....
N
\ j
j
NjNY---,./ Nj N .,.)-- 3L r
,,( N C N \ N O
1
S S 0,
S
..õ1 ¨NI, ri N rE _NI ) õ..0_Nr
N \¨
.--= .--,... 1\1 --- 4,=,,, I I , I 1
)
x\
1 0 '(NCI 'zr'N y \\N
L---./
N
I I I\1 --S --S --; /
`zzt. 0 I I I ,) __
`2t(NN).1 I
A N 'Ltt.ZN \Z--N
F
A-GN-I-. ....-----N-CH3 Abh ..A. I. 'F )222L*
;\.-) A VI A el [ F )2Z- I1
HO CH3
F Me0 I. p
xis
A. xi. cH, F
CH2CH3
CN\ 0
H3c ilo F OH
H3C--,,,,,...-,
rN---,/
1 N -------- N-----
./
,2zz,,N
cH3 >1.-0 F
CN
SO2Me N/-------./
.,,-----./ N'El
kON
=
[00413] In one aspect, B is selected from the moieties presented in Table
1.
Table 1. Illustrative B moieties of the compounds described herein.
Sub- B Sub- B Sub- B
class class # class
# #
100

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
B-1 B-2 B-3 -CH(CH3)2
N
1-0
A-)
B-4 F3C 40 B-5 B-6
\. Si
+<1
)24 CI
B-7 H3C . B-8 H3C B-9
A. 0
N
B-10 B-11
....-"-= N -CH3 B-12
---µ el
F
B-13 Me0 B-14 B-15 HO .
)2z. F A
B-16 B-17 B-18 . ON
--:??2, I. 4. I.
ON 4.
CN
B-19 0 B-20 B-21 H3C OCH3
;22a,N A el
)24
B-22 B-23 B-24 '14
1
NO2 40
. --c% s- -.'===................-- N A I.
IN-Th
._.-o
B-25 0 B-26 VcH3 B-27
I
'az2_ ).zz_N c22(1\1
B-28 B-29 B-30
I I I
`zzz NCI `zz,,N `Zzte
B-31 B-32 /. B-33 N
I
CF3
`atte I
101

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
B-34 N B-35 B-36 NH2
01 /sN al \ N I
N `zzLe
B-37 B-38 B-39 0
I I
`2NNH 2' 2 t ( NCN i e
1
`zzz_e
B-40 NCI B-41 1 B-42 N
I I 1 µ.rI 01
'azt.e `zzz.N-(C)
0 0
B-43 N B-44 IN B-45
f
µ CN
\ NO
H
B-46 (-OH B-47 F B-48 N
I I
\ N `2z2N- µ NH2
B-49 f NC:) B-50 NNH2 B-51 N
I I
B-52
N N B-53 I B-54
r
I\IN
N N
I I I
`2zz.N- `azte µe
B-55B-56 B-57
N-
r ) N
N N I\
I-ID .N.N
I
I
I µe
\e µN-
B-58 ro B-59 B-60
N N) N.N NN
j1 I I
'222. N '22z.e µe
B-61B-62 _N B-63
1\1 Ni--- N. N NN
I I I )
µe\
41t.e N
102

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
B-64 B-65 ril¨D B-66
I I
1 \N-
`22(e (zrz.1\r
B-67 B-68 0 B-69 N
I I 'z=zz-NCN
B-70 1\1 B-71 N
B-72 1\1
I I I
j
`zza.NO
`zzz. N N `zz2i NN
I I )
B-73 N B-74 N B-75
I\I
I I I
`2z2.NN `22z.NCI µ.NN
0
B-76 jN B-77 B-78 N
0 1\1
1 1
Ãal )
`z22.NN).1 `?zz.NNO -i. N
B-79 ,-S B-80 ....--S B-81 ...--S /
I I,>¨ I,)'
/
N `111.7--N `1/7,,--- N
B-82 B-83B-84 %
S\ ,-Sµ S 7
I /2¨N\ I /1¨N I ¨NI\ )
N
N 'EILZ---N ) '17--N '¨

B-85 ....-SrN B-86 B-87 -CH3
'="--
B-88 -CH2CH3 B-89 B-90 H3C---.....
B-91 B-92 cH3 B-93 F 10
1111::::;;')11..
,zza. :11.40 cH3 F
103

CA 02842187 2014 - 01 -16
WO 2013/012915 PCT/US2012/047186
B-94 H30 B-95 F B-96 /CH2CH3
(1\1\
CH3 XII 1 F
B-97 i.j B-98 0 B-99
OH
N
/4722)
B- SO2Me B-101 ON B-102
100
[00414]
In some embodiments, one or more compounds described herein bind to a PI3
kinase (e.g.,
bind selectively). In some embodiments, one or more compounds described herein
bind selectively to
a y or 6 subtype of a PI3 kinase.
[00415]
In some embodiments, the IC50 of a subject compound for p110a, p110f3, p1107,
or p1106 is
less than about 1 1.IM, less than about 100 nM, less than about 50 nM, less
than about 10 nM, less than 1 nM or even
less than about 0.5 nM. In some embodiments, the IC50 of a subject compound
for mTor is less than about 1 1.IM,
less than about 100 nM, less than about 50 nM, less than about 10 nM, less
than 1 nM or even less than about 0.5
nM. In some other embodiments, one or more subject compounds exhibit dual
binding specificity and are capable
of inhibiting a PI3 kinase (e.g., a class I PI3 kinase) as well as a protein
kinase (e.g., mTor) with an IC50 value less
than about 1 1.IM, less than about 100 nM, less than about 50 nM, less than
about 10 nM, less than 1 nM or even less
than about 0.5 nM. One or more subject compounds are capable of inhibiting
tyrosine kinases including, for
example, DNA-dependent protein kinase (Pubmed protein accession number (PPAN)
AAA79184), Abl tyrosine
kinase (CAA52387), Bcr-Abl, hemopoietic cell kinase (PPAN CAI19695), Src (PPAN
CAA24495), vascular
endothelial growth factor receptor 2 (PPAN ABB82619), vascular endothelial
growth factor receptor-2 (PPAN
ABB82619), epidermal growth factor receptor (PPAN AG43241), EPH receptor B4
(PPAN EAL23820), stem cell
factor receptor (PPAN AAF22141), Tyrosine-protein kinase receptor TIE-2 (PPAN
Q02858), fms-related tyrosine
kinase 3 (PPAN NP_004110), platelet-derived growth factor receptor alpha (PPAN
NP 990080), RET (PPAN
CAA73131), and functional mutants thereof. In some embodiments, the tyrosine
kinase is Abl, Bcr-Abl, EGFR, or
Flt-3, and any other kinases listed in the Tables herein.
[00416]
In some embodiments, non-limiting exemplary compounds exhibit one or more
functional
characteristics disclosed herein. For example, one or more subject compounds
bind specifically to a PI3 kinase. In
some embodiments, the IC50 of a subject compound for p110a, p110f3, p1107, or
p1106 is less than about 11.IM, less
than about 100 nM, less than about 50 nM, less than about 10 nM, less than
about 1 nM, less than about 0.5 nM, less
than about 100 pM, or less than about 50 pM.
104

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00417] In some embodiments, one or more of the subject compounds can
selectively inhibit one or
more members of type I or class I phosphatidylinositol 3-kinases (P13-kinase)
with an IC50 value of about 100 nM,
50 nM, 10 nM, 5 nM, 100 pM, 10 pM or 1 pM, or less as measured in an in vitro
kinase assay.
[00418] In some embodiments, one or more of the subject compounds can
selectively inhibit one or
two members of type I or class I phosphatidylinositol 3-kinases (PI3-kinase)
such as PI3-kinase a, PI3-kinase p, PI3-
kinase 7, and P13-kinase 6. In some aspects, some of the subject compounds
selectively inhibit P13-kinase 6 as
compared to all other type I P13-kinases. In other aspects, some of the
subject compounds selectively inhibit PI3-
kinase 6 and P13-kinase 7 as compared to the rest of the type I PI3-kinases.
In yet other aspects, some of the subject
compounds selectively inhibit P13-kinase a and P13-kinase p as compared to the
rest of the type I P13-kinases. In
still yet some other aspects, some of the subject compounds selectively
inhibit P13-kinase 6 and P13-kinase a as
compared to the rest of the type I P13-kinases. In still yet some other
aspects, some of the subject compounds
selectively inhibit P13-kinase 6 and P13-kinase 3 as compared to the rest of
the type I P13-kinases, or selectively
inhibit P13-kinase 6 and P13-kinase a as compared to the rest of the type I
P13-kinases, or selectively inhibit PI3-
kinase a and P13-kinase y as compared to the rest of the type I P13-kinases,
or selectively inhibit P13-kinase y and
P13-kinase 3 as compared to the rest of the type I P13-kinases.
[00419] In yet another aspect, an inhibitor that selectively inhibits
one or more members of type I PI3-
kinases, or an inhibitor that selectively inhibits one or more type I P13-
kinase mediated signaling pathways,
alternatively can be understood to refer to a compound that exhibits a 50%
inhibitory concentration (IC50) with
respect to a given type I P13-kinase, that is at least about 10-fold, at least
about 20-fold, at least about 50-fold, at
least about 100-fold, at least about 1000-fold, at least about 10,000-fold, or
lower, than the inhibitor's IC50 with
respect to the rest of the other type I P13-kinases. In one embodiment, an
inhibitor selectively inhibits P13-kinase 6
as compared to PI3-kinase 3 with at least about 10-fold lower IC50 for PI3-
kinase 6. In certain embodiments, the
IC50 for PI3-kinase (5 is below about 100 nM, while the IC50 for P13-kinase 3
is above about 1000 nM. In certain
embodiments, the IC50 for P13-kinase 6 is below about 50 nM, while the IC50
for P13-kinase 3 is above about 5000
nM. In certain embodiments, the IC50 for PI3-kinase 6 is below about 10 nM,
while the IC50 for PI3-kinase 3 is
above about 1000 nM, above about 5,000 nM, or above about 10,000 nM.
Pharmaceutical Compositions
[00420] In some embodiments, provided herein are pharmaceutical
compositions comprising one or
more compounds as disclosed herein, or a pharmaceutically acceptable form
thereof (e.g., pharmaceutically
acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically
labeled derivatives), and one or more
pharmaceutically acceptable excipients carriers, including inert solid
diluents and fillers, diluents, including sterile
aqueous solution and various organic solvents, permeation enhancers,
solubilizers and adjuvants. In some
embodiments, a pharmaceutical composition described herein includes a second
active agent such as an additional
therapeutic agent, (e.g., a chemotherapeutic).
1. Formulations
105

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00421]
Pharmaceutical compositions can be specially formulated for administration in
solid or liquid
form, including those adapted for the following: oral administration, for
example, drenches (aqueous or non-aqueous
solutions or suspensions), tablets (e.g., those targeted for buccal,
sublingual, and systemic absorption), capsules,
boluses, powders, granules, pastes for application to the tongue, and
intraduodenal routes; parenteral administration,
including intravenous, intraarterial, subcutaneous, intramuscular,
intravascular, intraperitoneal or infusion as, for
example, a sterile solution or suspension, or sustained-release formulation;
topical application, for example, as a
cream, ointment, or a controlled-release patch or spray applied to the skin;
intravaginally or intrarectally, for
example, as a pessary, cream, stent or foam; sublingually; ocularly;
pulmonarily; local delivery by catheter or stent;
intrathecally, or nasally.
[00422]
Examples of suitable aqueous and nonaqueous carriers which can be employed in
pharmaceutical compositions include water, ethanol, polyols (such as glycerol,
propylene glycol, polyethylene
glycol, and the like), and suitable mixtures thereof, vegetable oils, such as
olive oil, and injectable organic esters,
such as ethyl oleate. Proper fluidity can be maintained, for example, by the
use of coating materials, such as
lecithin, by the maintenance of the required particle size in the case of
dispersions, and by the use of surfactants.
[00423]
These compositions can also contain adjuvants such as preservatives, wetting
agents,
emulsifying agents, dispersing agents, lubricants, and/or antioxidants.
Prevention of the action of microorganisms
upon the compounds described herein can be ensured by the inclusion of various
antibacterial and antifungal agents,
for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It can
also be desirable to include isotonic
agents, such as sugars, sodium chloride, and the like into the compositions.
In addition, prolonged absorption of the
injectable pharmaceutical form can be brought about by the inclusion of agents
which delay absorption such as
aluminum monostearate and gelatin.
[00424]
Methods of preparing these formulations or compositions include the step of
bringing into
association a compound described herein and/or the chemotherapeutic with the
carrier and, optionally, one or more
accessory ingredients. In general, the formulations are prepared by uniformly
and intimately bringing into
association a compound as disclosed herein with liquid carriers, or finely
divided solid carriers, or both, and then, if
necessary, shaping the product.
[00425]
Preparations for such pharmaceutical compositions are well-known in the art.
See, e.g., Anderson,
Philip O.; Knoben, James E.; Troutman, William G, eds., Handbook of Clinical
Drug Data, Tenth Edition,
McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third
Edition, Churchill Livingston, New
York, 1990; Katzung, ed., Basic and Clinical Pharmacology, Ninth Edition,
McGraw Hill, 20037ybg; Goodman and
Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw
Hill, 2001; Remingtons
Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000;
Martindale, The Extra Pharmacopoeia,
Thirty-Second Edition (The Pharmaceutical Press, London, 1999); all of which
are incorporated by reference herein
in their entirety. Except insofar as any conventional excipient medium is
incompatible with the compounds
provided herein, such as by producing any undesirable biological effect or
otherwise interacting in a deleterious
manner with any other component(s) of the pharmaceutically acceptable
composition, the excipient's use is
contemplated to be within the scope of this disclosure.
106

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00426]
In some embodiments, the concentration of one or more of the compounds
provided in the
disclosed pharmaceutical compositions is less than about 100%, about 90%,
about 80%, about 70%, about 60%,
about 50%, about 40%, about 30%, about 20%, about 19%, about 18%, about 17%,
about 16%, about 15%, about
14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%,
about 6%, about 5%, about
4%, about 3%, about 2%, about 1%, about 0.5%, about 0.4%, about 0.3%, about
0.2%, about 0.1%, about 0.09%,
about 0.08%, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%,
about 0.02%, about 0.01%, about
0.009%, about 0.008%, about 0.007%, about 0.006%, about 0.005%, about 0.004%,
about 0.003%, about 0.002%,
about 0.001%, about 0.0009%, about 0.0008%, about 0.0007%, about 0.0006%,
about 0.0005%, about 0.0004%,
about 0.0003%, about 0.0002%, or about 0.0001% w/w, w/v or v/v.
[00427]
In some embodiments, the concentration of one or more of the compounds as
disclosed herein
is greater than about 90%, about 80%, about 70%, about 60%, about 50%, about
40%, about 30%, about 20%, about
19.75%, about 19.50%, about 19.25% about 19%, about 18.75%, about 18.50%,
about 18.25%, about 18%, about
17.75%, about 17.50%, about 17.25%, about 17%, about 16.75%, about 16.50%,
about 16.25%, about 16%, about
15.75%, about 15.50%, about 15.25%, about 15%, about 14.75%, about 14.50%,
about 14.25%, about 14%, about
13.75%, about 13.50%, about 13.25%, about 13%, about 12.75%, about 12.50%,
about 12.25%, about 12%, about
11.75%, about 11.50%, about 11.25%, about 11%, about 10.75%, about 10.50%,
about 10.25%, about 10%, about
9.75%, about 9.50%, about 9.25%, about 9%, about 8.75%, about 8.50%, about
8.25%, about 8%, about 7.75%,
about 7.50%, about 7.25%, about 7%, about 6.75%, about 6.50%, about 6.25%,
about 6%, about 5.75%, about
5.50%, about 5.25%, about 5%, about 4.75%, about 4.50%, about 4.25%, about 4%,
about 3.75%, about 3.50%,
about 3.25%, about 3%, about 2.75%, about 2.50%, about 2.25%, about 2%, about
1.75%, about 1.50%, about
1.25%, about 1%, about 0.5%, about 0.4%, about 0.3%, about 0.2%, about 0.1%,
about 0.09%, about 0.08%, about
0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, about
0.01%, about 0.009%, about
0.008%, about 0.007%, about 0.006%, about 0.005%, about 0.004%, about 0.003%,
about 0.002%, about 0.001%,
about 0.0009%, about 0.0008%, about 0.0007%, about 0.0006%, about 0.0005%,
about 0.0004%, about 0.0003%,
about 0.0002%, or about 0.0001% w/w, w/v, or v/v.
[00428]
In some embodiments, the concentration of one or more of the compounds as
disclosed herein
is in the range from approximately 0.0001% to approximately 50%, approximately
0.001% to approximately 40 %,
approximately 0.01% to approximately 30%, approximately 0.02% to approximately
29%, approximately 0.03% to
approximately 28%, approximately 0.04% to approximately 27%, approximately
0.05% to approximately 26%,
approximately 0.06% to approximately 25%, approximately 0.07% to approximately
24%, approximately 0.08% to
approximately 23%, approximately 0.09% to approximately 22%, approximately
0.1% to approximately 21%,
approximately 0.2% to approximately 20%, approximately 0.3% to approximately
19%, approximately 0.4% to
approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6%
to approximately 16%,
approximately 0.7% to approximately 15%, approximately 0.8% to approximately
14%, approximately 0.9% to
approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. v/v.
[00429]
In some embodiments, the concentration of one or more of the compounds as
disclosed herein
is in the range from approximately 0.001% to approximately 10%, approximately
0.01% to approximately 5%,
107

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
approximately 0.02% to approximately 4.5%, approximately 0.03% to
approximately 4%, approximately 0.04% to
approximately 3.5%, approximately 0.05% to approximately 3%, approximately
0.06% to approximately 2.5%,
approximately 0.07% to approximately 2%, approximately 0.08% to approximately
1.5%, approximately 0.09% to
approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
[00430]
In some embodiments, the amount of one or more of the compounds as disclosed
herein is
equal to or less than about 10 g, about 9.5 g, about 9.0 g, about 8.5 g, about
8.0 g, 7.5 g, about 7.0 g, about 6.5 g,
about 6.0 g, about 5.5 g, about 5.0 g, about 4.5 g, about 4.0 g, about 3.5 g,
about 3.0 g, about 2.5 g, about 2.0 g,
about 1.5 g, about 1.0 g, about 0.95 g, about 0.9 g, about 0.85 g, about 0.8
g, about 0.75 g, about 0.7 g, about 0.65 g,
about 0.6 g, about 0.55 g, about 0.5 g, about 0.45 g, about 0.4 g, about 0.35
g, about 0.3 g, about 0.25 g, about 0.2 g,
about 0.15 g, about 0.1 g, about 0.09 g, about 0.08 g, about 0.07 g, about
0.06 g, about 0.05 g, about 0.04 g, about
0.03 g, about 0.02 g, about 0.01 g, about 0.009 g, about 0.008 g, about 0.007
g, about 0.006 g, about 0.005 g, about
0.004 g, about 0.003 g, about 0.002 g, about 0.001 g, about 0.0009 g, about
0.0008 g, about 0.0007 g, about 0.0006
g, about 0.0005 g, about 0.0004 g, about 0.0003 g, about 0.0002 g, or about
0.0001 g.
[00431]
In some embodiments, the amount of one or more of the compounds as disclosed
herein is
more than about about 0.0001 g, about 0.0002 g, about 0.0003 g, about 0.0004
g, about 0.0005 g, about 0.0006 g,
about 0.0007 g, about 0.0008 g, about 0.0009 g, about 0.001 g, about 0.0015 g,
about 0.002 g, about 0.0025 g, about
0.003 g, about 0.0035 g, about 0.004 g, about 0.0045 g, about 0.005 g, about
0.0055 g, about 0.006 g, about 0.0065
g, about 0.007 g, about 0.0075 g, about 0.008 g, about 0.0085 g, about 0.009
g, about 0.0095 g, about 0.01 g, about
0.015 g, about 0.02 g, about 0.025 g, about 0.03 g, about 0.035 g, about 0.04
g, about 0.045 g, about 0.05 g, about
0.055 g, about 0.06 g, about 0.065 g, about 0.07 g, about 0.075 g, about 0.08
g, about 0.085 g, about 0.09 g, about
0.095 g, about 0.1 g, about 0.15 g, about 0.2 g, about 0.25 g, about 0.3 g,
about 0.35 g, about 0.4 g, about 0.45 g,
about 0.5 g, about 0.55 g, about 0.6 g, about 0.65 g, about 0.7 g, about 0.75
g, about 0.8 g, about 0.85 g, about 0.9 g,
about 0.95 g, about 1 g, about 1.5 g, about 2 g, about 2.5, about 3 g, about
3.5, about 4 g, about 4.5 g, about 5 g,
about 5.5 g, about 6 g, about 6.5g, about 7 g, about 7.5g, about 8 g, about
8.5 g, about 9 g, about 9.5 g, or about 10 g.
[00432]
In some embodiments, the amount of one or more of the compounds as disclosed
herein is in
the range of about 0.0001 - about 10 g, about 0.0005 - about 9 g, about 0.001 -
about 8 g, about 0.005 - about 7 g,
about 0.01 - about 6 g, about 0.05 - about 5 g, about 0.1 - about 4 g, about
0.5 - about 4 g, or about 1 - about 3 g.
1A. Formulations for oral administration
[00433]
In some embodiments, provided herein are pharmaceutical compositions for oral
administration containing a compound as disclosed herein, and a pharmaceutical
excipient suitable for oral
administration. In some embodiments, provided herein are pharmaceutical
compositions for oral administration
containing: (i) an effective amount of a disclosed compound; optionally (ii)
an effective amount of one or more
second agents; and (iii) one or more pharmaceutical excipients suitable for
oral administration. In some
embodiments, the pharmaceutical composition further contains: (iv) an
effective amount of a third agent.
[00434]
In some embodiments, the pharmaceutical composition can be a liquid
pharmaceutical
composition suitable for oral consumption. Pharmaceutical compositions
suitable for oral administration can be
108

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
presented as discrete dosage forms, such as capsules, cachets, or tablets, or
liquids or aerosol sprays each containing
a predetermined amount of an active ingredient as a powder or in granules, a
solution, or a suspension in an aqueous
or non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid
emulsion. Such dosage forms can be
prepared by any of the methods of pharmacy, but all methods include the step
of bringing the active ingredient into
association with the carrier, which constitutes one or more ingredients. In
general, the pharmaceutical compositions
are prepared by uniformly and intimately admixing the active ingredient with
liquid carriers or finely divided solid
carriers or both, and then, if necessary, shaping the product into the desired
presentation. For example, a tablet can
be prepared by compression or molding, optionally with one or more accessory
ingredients. Compressed tablets can
be prepared by compressing in a suitable machine the active ingredient in a
free-flowing form such as powder or
granules, optionally mixed with an excipient such as, but not limited to, a
binder, a lubricant, an inert diluent, and/or
a surface active or dispersing agent. Molded tablets can be made by molding in
a suitable machine a mixture of the
powdered compound moistened with an inert liquid diluent.
[00435]
The present disclosure further encompasses anhydrous pharmaceutical
compositions and
dosage forms comprising an active ingredient, since water can facilitate the
degradation of some compounds. For
example, water can be added (e.g., about 5%) in the pharmaceutical arts as a
means of simulating long-term storage
in order to determine characteristics such as shelf-life or the stability of
formulations over time. Anhydrous
pharmaceutical compositions and dosage forms can be prepared using anhydrous
or low moisture containing
ingredients and low moisture or low humidity conditions. For example,
pharmaceutical compositions and dosage
forms which contain lactose can be made anhydrous if substantial contact with
moisture and/or humidity during
manufacturing, packaging, and/or storage is expected. An anhydrous
pharmaceutical composition can be prepared
and stored such that its anhydrous nature is maintained. Accordingly,
anhydrous pharmaceutical compositions can
be packaged using materials known to prevent exposure to water such that they
can be included in suitable
formulary kits. Examples of suitable packaging include, but are not limited
to, hermetically sealed foils, plastic or
the like, unit dose containers, blister packs, and strip packs.
[00436]
An active ingredient can be combined in an intimate admixture with a
pharmaceutical carrier
according to conventional pharmaceutical compounding techniques. The carrier
can take a wide variety of forms
depending on the form of preparation desired for administration. In preparing
the pharmaceutical compositions for
an oral dosage form, any of the usual pharmaceutical media can be employed as
carriers, such as, for example,
water, glycols, oils, alcohols, flavoring agents, preservatives, coloring
agents, and the like in the case of oral liquid
preparations (such as suspensions, solutions, and elixirs) or aerosols; or
carriers such as starches, sugars, micro-
crystalline cellulose, diluents, granulating agents, lubricants, binders, and
disintegrating agents can be used in the
case of oral solid preparations, in some embodiments without employing the use
of lactose. For example, suitable
carriers include powders, capsules, and tablets, with the solid oral
preparations. In some embodiments, tablets can be
coated by standard aqueous or nonaqueous techniques.
[00437]
Binders suitable for use in pharmaceutical compositions and dosage forms
include, but are not
limited to, corn starch, potato starch, or other starches, gelatin, natural
and synthetic gums such as acacia, sodium
alginate, alginic acid, other alginates, powdered tragacanth, guar gum,
cellulose and its derivatives (e.g., ethyl
109

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium
carboxymethyl cellulose), polyvinyl
pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl
cellulose, microcrystalline cellulose,
and mixtures thereof.
[00438] Examples of suitable fillers for use in the pharmaceutical
compositions and dosage forms
disclosed herein include, but are not limited to, talc, calcium carbonate
(e.g., granules or powder), microcrystalline
cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid,
sorbitol, starch, pre-gelatinized starch, and
mixtures thereof.
[00439] Disintegrants can be used in the pharmaceutical compositions as
provided herein to provide
tablets that disintegrate when exposed to an aqueous environment. Too much of
a disintegrant can produce tablets
which can disintegrate in the bottle. Too little can be insufficient for
disintegration to occur and can thus alter the
rate and extent of release of the active ingredient(s) from the dosage form.
Thus, a sufficient amount of disintegrant
that is neither too little nor too much to detrimentally alter the release of
the active ingredient(s) can be used to form
the dosage forms of the compounds disclosed herein. The amount of disintegrant
used can vary based upon the type
of formulation and mode of administration, and can be readily discernible to
those of ordinary skill in the art. About
0.5 to about 15 weight percent of disintegrant, or about 1 to about 5 weight
percent of disintegrant, can be used in
the pharmaceutical composition. Disintegrants that can be used to form
pharmaceutical compositions and dosage
forms include, but are not limited to, agar-agar, alginic acid, calcium
carbonate, microcrystalline cellulose,
croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch
glycolate, potato or tapioca starch, other
starches, pre-gelatinized starch, other starches, clays, other algins, other
celluloses, gums or mixtures thereof.
[00440] Lubricants which can be used to form pharmaceutical
compositions and dosage forms
include, but are not limited to, calcium stearate, magnesium stearate, mineral
oil, light mineral oil, glycerin, sorbitol,
mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl
sulfate, talc, hydrogenated vegetable oil
(e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn
oil, and soybean oil), zinc stearate, ethyl
oleate, ethylaureate, agar, or mixtures thereof. Additional lubricants
include, for example, a syloid silica gel, a
coagulated aerosol of synthetic silica, or mixtures thereof. A lubricant can
optionally be added, in an amount of less
than about 1 weight percent of the pharmaceutical composition.
[00441] When aqueous suspensions and/or elixirs are desired for oral
administration, the active
ingredient therein can be combined with various sweetening or flavoring
agents, coloring matter or dyes and, for
example, emulsifying and/or suspending agents, together with such diluents as
water, ethanol, propylene glycol,
glycerin and various combinations thereof.
[00442] The tablets can be uncoated or coated by known techniques to
delay disintegration and
absorption in the gastrointestinal tract and thereby provide a sustained
action over a longer period. For example, a
time delay material such as glyceryl monostearate or glyceryl distearate can
be employed. Formulations for oral use
can also be presented as hard gelatin capsules wherein the active ingredient
is mixed with an inert solid diluent, for
example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin
capsules wherein the active ingredient is
mixed with water or an oil medium, for example, peanut oil, liquid paraffin or
olive oil.
110

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00443] Surfactant which can be used to form pharmaceutical
compositions and dosage forms include,
but are not limited to, hydrophilic surfactants, lipophilic surfactants, and
mixtures thereof. That is, a mixture of
hydrophilic surfactants can be employed, a mixture of lipophilic surfactants
can be employed, or a mixture of at
least one hydrophilic surfactant and at least one lipophilic surfactant can be
employed.
[00444] A suitable hydrophilic surfactant can generally have an HLB
value of at least about 10, while
suitable lipophilic surfactants can generally have an HLB value of or less
than about 10. An empirical parameter
used to characterize the relative hydrophilicity and hydrophobicity of non-
ionic amphiphilic compounds is the
hydrophilic-lipophilic balance ("HLB" value). Surfactants with lower HLB
values are more lipophilic or
hydrophobic, and have greater solubility in oils, while surfactants with
higher HLB values are more hydrophilic, and
have greater solubility in aqueous solutions. Hydrophilic surfactants are
generally considered to be those compounds
having an HLB value greater than about 10, as well as anionic, cationic, or
zwitterionic compounds for which the
HLB scale is not generally applicable. Similarly, lipophilic (i.e.,
hydrophobic) surfactants are compounds having an
HLB value equal to or less than about 10. However, HLB value of a surfactant
is merely a rough guide generally
used to enable formulation of industrial, pharmaceutical and cosmetic
emulsions.
[00445] Hydrophilic surfactants can be either ionic or non-ionic.
Suitable ionic surfactants include,
but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid
derivatives of amino acids, oligopeptides,
and polypeptides; glyceride derivatives of amino acids, oligopeptides, and
polypeptides; lecithins and hydrogenated
lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and
derivatives thereof; lysophospholipids and
derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates;
fatty acid salts; sodium docusate;
acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-
glycerides; succinylated mono- and di-
glycerides; citric acid esters of mono- and di-glycerides; and mixtures
thereof.
[00446] Within the aforementioned group, ionic surfactants include, by
way of example: lecithins,
lysolecithin, phospholipids, lysophospholipids and derivatives thereof;
carnitine fatty acid ester salts; salts of
alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-
acetylated tartaric acid esters of mono-
and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of
mono- and di-glycerides; and mixtures
thereof.
[00447] Ionic surfactants can be the ionized forms of lecithin,
lysolecithin, phosphatidylcholine,
phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid,
phosphatidylserine, lysophosphatidylcholine,
lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid,
lysophosphatidylserine, PEG-
phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of
fatty acids, stearoy1-2-lactylate,
stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric
acid esters of mono/diglycerides, citric
acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate,
caprate, laurate, myristate, palmitate, oleate,
ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl
sulfate, docusate, lauroyl carnitines, palmitoyl
carnitines, myristoyl carnitines, and salts and mixtures thereof.
[00448] Hydrophilic non-ionic surfactants can include, but are not
limited to, alkylglucosides;
alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides;
polyoxyalkylene alkyl ethers such as polyethylene
glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol
alkyl phenols; polyoxyalkylene alkyl
111

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
phenol fatty acid esters such as polyethylene glycol fatty acids monoesters
and polyethylene glycol fatty acids
diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty
acid esters; polyoxyalkylene sorbitan fatty
acid esters such as polyethylene glycol sorbitan fatty acid esters;
hydrophilic transesterification products of a polyol
with at least one member of glycerides, vegetable oils, hydrogenated vegetable
oils, fatty acids, and sterols;
polyoxyethylene sterols, derivatives, and analogues thereof; polyoxyethylated
vitamins and derivatives thereof;
polyoxyethylene-polyoxypropylene block copolymers; and mixtures thereof;
polyethylene glycol sorbitan fatty acid
esters and hydrophilic transesterification products of a polyol with at least
one member of triglycerides, vegetable
oils, and hydrogenated vegetable oils. The polyol can be glycerol, ethylene
glycol, polyethylene glycol, sorbitol,
propylene glycol, pentaerythritol, or a saccharide.
[00449] Other hydrophilic-non-ionic surfactants include, without
limitation, PEG-10 laurate, PEG-12
laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-
15 oleate, PEG-20 oleate, PEG-20
dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG-15 stearate, PEG-
32 distearate, PEG-40 stearate,
PEG-100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32
dioleate, PEG-20 glyceryl laurate, PEG-30
glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30
glyceryl oleate, PEG-30 glyceryl
laurate, PEG-40 glyceryl laurate, PEG-40 palm kernel oil, PEG-50 hydrogenated
castor oil, PEG-40 castor oil, PEG-
35 castor oil, PEG-60 castor oil, PEG-40 hydrogenated castor oil, PEG-60
hydrogenated castor oil, PEG-60 corn oil,
PEG-6 caprate/caprylate glycerides, PEG-8 caprate/caprylate glycerides,
polyglyceryl-10 laurate, PEG-30
cholesterol, PEG-25 phyto sterol, PEG-30 soya sterol, PEG-20 trioleate, PEG-40
sorbitan oleate, PEG-80 sorbitan
laurate, polysorbate 20, polysorbate 80, POE-9 lauryl ether, POE-23 lauryl
ether, POE-10 oleyl ether, POE-20 oleyl
ether, POE-20 stearyl ether, tocopheryl PEG-100 succinate, PEG-24 cholesterol,
polyglycery1-10oleate, Tween 40,
Tween 60, sucrose monostearate, sucrose monolaurate, sucrose monopalmitate,
PEG 10-100 nonyl phenol series,
PEG 15-100 octyl phenol series, and poloxamers.
[00450] Suitable lipophilic surfactants include, by way of example
only: fatty alcohols; glycerol fatty
acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids
esters; propylene glycol fatty acid esters;
sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters;
sterols and sterol derivatives;
polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl
ethers; sugar esters; sugar ethers; lactic
acid derivatives of mono- and di-glycerides; hydrophobic transesterification
products of a polyol with at least one
member of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids
and sterols; oil-soluble
vitamins/vitamin derivatives; and mixtures thereof. Within this group, non-
limiting examples of lipophilic
surfactants include glycerol fatty acid esters, propylene glycol fatty acid
esters, and mixtures thereof, or are
hydrophobic transesterification products of a polyol with at least one member
of vegetable oils, hydrogenated
vegetable oils, and triglycerides.
[00451] In one embodiment, the pharmaceutical composition can include a
solubilizer to ensure good
solubilization and/or dissolution of a compound as provided herein and to
minimize precipitation of the compound.
This can be especially important for pharmaceutical compositions for non-oral
use, e.g., pharmaceutical
compositions for injection. A solubilizer can also be added to increase the
solubility of the hydrophilic drug and/or
112

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
other components, such as surfactants, or to maintain the pharmaceutical
composition as a stable or homogeneous
solution or dispersion.
[00452]
Examples of suitable solubilizers include, but are not limited to, the
following: alcohols and
polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene
glycol, propylene glycol, butanediols and
isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol,
dimethyl isosorbide, polyethylene glycol,
polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and
other cellulose derivatives,
cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols
having an average molecular weight of
about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether
(glycofurol) or methoxy PEG; amides and
other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone, 8-
caprolactam, N-alkylpyrrolidone, N-
hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam,
dimethylacetamide and polyvinylpyrrolidone;
esters such as ethyl propionate, tributylcitrate, acetyl triethylcitrate,
acetyl tributyl citrate, triethylcitrate, ethyl oleate,
ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate,
propylene glycol diacetate, 8-caprolactone
and isomers thereof, 6-valerolactone and isomers thereof, 13-butyrolactone and
isomers thereof; and other
solubilizers known in the art, such as dimethyl acetamide, dimethyl
isosorbide, N-methyl pyrrolidones,
monooctanoin, diethylene glycol monoethyl ether, and water.
[00453]
Mixtures of solubilizers can also be used. Examples include, but not limited
to, triacetin,
triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-
methylpyrrolidone, N-hydroxyethylpyrrolidone,
polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl
cyclodextrins, ethanol, polyethylene glycol
200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. In
some embodiments, solubilizers
include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and
propylene glycol.
[00454]
The amount of solubilizer that can be included is not particularly limited.
The amount of a
given solubilizer can be limited to a bioacceptable amount, which can be
readily determined by one of skill in the
art. In some circumstances, it can be advantageous to include amounts of
solubilizers far in excess of bioacceptable
amounts, for example to maximize the concentration of the drug, with excess
solubilizer removed prior to providing
the pharmaceutical composition to a subject using conventional techniques,
such as distillation or evaporation. Thus,
if present, the solubilizer can be in a weight ratio of about 10%, 25%, 50%,
100%, or up to about 200% by weight,
based on the combined weight of the drug, and other excipients. If desired,
very small amounts of solubilizer can
also be used, such as about 5%, 2%, 1% or even less. Typically, the
solubilizer can be present in an amount of about
1% to about 100%, more typically about 5% to about 25% by weight.
[00455]
The pharmaceutical composition can further include one or more
pharmaceutically acceptable
additives and excipients. Such additives and excipients include, without
limitation, detackifiers, anti-foaming agents,
buffering agents, polymers, antioxidants, preservatives, chelating agents,
viscomodulators, tonicifiers, flavorants,
colorants, oils, odorants, opacifiers, suspending agents, binders, fillers,
plasticizers, lubricants, and mixtures thereof.
[00456]
Exemplary preservatives can include antioxidants, chelating agents,
antimicrobial
preservatives, antifungal preservatives, alcohol preservatives, acidic
preservatives, and other preservatives.
Exemplary antioxidants include, but are not limited to, alpha tocopherol,
ascorbic acid, acorbyl palmitate, butylated
hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium
metabisulfite, propionic acid, propyl
113

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and sodium
sulfite. Exemplary chelating agents
include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate,
disodium edetate, dipotassium edetate,
edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate,
tartaric acid, and trisodium edetate.
Exemplary antimicrobial preservatives include, but are not limited to,
benzalkonium chloride, benzethonium
chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride,
chlorhexidine, chlorobutanol, chlorocresol,
chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol,
phenoxyethanol, phenylethyl alcohol,
phenylmercuric nitrate, propylene glycol, and thimerosal. Exemplary antifungal
preservatives include, but are not
limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben,
benzoic acid, hydroxybenzoic acid,
potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and
sorbic acid. Exemplary alcohol
preservatives include, but are not limited to, ethanol, polyethylene glycol,
phenol, phenolic compounds, bisphenol,
chlorobutanol, hydroxybenzoate, and phenylethyl alcohol. Exemplary acidic
preservatives include, but are not
limited to, vitamin A, vitamin C, vitamin E, beta¨carotene, citric acid,
acetic acid, dehydroacetic acid, ascorbic acid,
sorbic acid, and phytic acid. Other preservatives include, but are not limited
to, tocopherol, tocopherol acetate,
deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated
hydroxytoluened (BHT),
ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate
(SLES), sodium bisulfite, sodium
metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus,
Phenonip, methylparaben, Germall 115,
Germaben II, Neolone, Kathon, and Euxyl. In certain embodiments, the
preservative is an anti¨oxidant. In other
embodiments, the preservative is a chelating agent.
[00457]
Exemplary oils include, but are not limited to, almond, apricot kernel,
avocado, babassu,
bergamot, black current seed, borage, cade, camomile, canola, caraway,
carnauba, castor, cinnamon, cocoa butter,
coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening
primrose, fish, flaxseed, geraniol, gourd,
grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut,
lavandin, lavender, lemon, litsea cubeba,
macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive,
orange, orange roughy, palm, palm
kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran,
rosemary, safflower, sandalwood,
sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean,
sunflower, tea tree, thistle, tsubaki, vetiver,
walnut, and wheat germ oils. Exemplary oils include, but are not limited to,
butyl stearate, caprylic triglyceride,
capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360,
isopropyl myristate, mineral oil,
octyldodecanol, oleyl alcohol, silicone oil, and combinations thereof.
[00458]
In addition, an acid or a base can be incorporated into the pharmaceutical
composition to
facilitate processing, to enhance stability, or for other reasons. Examples of
pharmaceutically acceptable bases
include amino acids, amino acid esters, ammonium hydroxide, potassium
hydroxide, sodium hydroxide, sodium
hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium
hydroxide, magnesium aluminum silicate,
synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum
hydroxide, diisopropylethylamine,
ethanolamine, ethylenediamine, triethanolamine, triethylamine,
triisopropanolamine, trimethylamine,
tris(hydroxymethyl)aminomethane (TRIS) and the like. Also suitable are bases
that are salts of a pharmaceutically
acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid,
alkanesulfonic acid, amino acids, ascorbic
acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid,
fatty acids, formic acid, fumaric acid, gluconic
114

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid,
oxalic acid, para-bromophenylsulfonic acid,
propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic
acid, tannic acid, tartaric acid,
thioglycolic acid, toluenesulfonic acid, uric acid, and the like. Salts of
polyprotic acids, such as sodium phosphate,
disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used.
When the base is a salt, the
cation can be any convenient and pharmaceutically acceptable cation, such as
ammonium, alkali metals, alkaline
earth metals, and the like. Examples can include, but not limited to, sodium,
potassium, lithium, magnesium,
calcium and ammonium.
[00459]
Suitable acids are pharmaceutically acceptable organic or inorganic acids.
Examples of
suitable inorganic acids include hydrochloric acid, hydrobromic acid,
hydriodic acid, sulfuric acid, nitric acid, boric
acid, phosphoric acid, and the like. Examples of suitable organic acids
include acetic acid, acrylic acid, adipic acid,
alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid,
boric acid, butyric acid, carbonic acid,
citric acid, fatty acids, formic acid, fumaric acid, gluconic acid,
hydroquinosulfonic acid, isoascorbic acid, lactic
acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic
acid, propionic acid, p-
toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic
acid, tartaric acid, thioglycolic acid,
toluenesulfonic acid, uric acid and the like.
1B. Formulations for Parenteral Administration
[00460]
In some embodiments, provided herein are pharmaceutical compositions for
parenteral
administration containing a compound as disclosed herein, and a pharmaceutical
excipient suitable for parenteral
administration. In some embodiments, provided herein are pharmaceutical
compositions for parenteral
administration containing: (i) an effective amount of a disclosed compound;
optionally (ii) an effective amount of
one or more second agents; and (iii) one or more pharmaceutical excipients
suitable for parenteral administration. In
some embodiments, the pharmaceutical composition further contains: (iv) an
effective amount of a third agent.
[00461]
The forms in which the disclosed pharmaceutical compositions can be
incorporated for
administration by injection include aqueous or oil suspensions, or emulsions,
with sesame oil, corn oil, cottonseed
oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile
aqueous solution, and similar pharmaceutical
vehicles.
Aqueous solutions in saline are also conventionally used for injection.
Ethanol, glycerol, propylene glycol, liquid
polyethylene glycol, and the like (and suitable mixtures thereof),
cyclodextrin derivatives, and vegetable oils can
also be employed.
[00462] Aqueous solutions in saline are also conventionally used for
injection. Ethanol, glycerol,
propylene glycol, liquid polyethylene glycol, and the like (and suitable
mixtures thereof), cyclodextrin derivatives,
and vegetable oils can also be employed. The proper fluidity can be
maintained, for example, by the use of a
coating, such as lecithin, for the maintenance of the required particle size
in the case of dispersion and by the use of
surfactants. The prevention of the action of microorganisms can be brought
about by various antibacterial and
antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid,
thimerosal, and the like.
115

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00463]
Sterile injectable solutions are prepared by incorporating a compound as
disclosed herein in the
required amount in the appropriate solvent with various other ingredients as
enumerated above, as appropriate,
followed by filtered sterilization. Generally, dispersions are prepared by
incorporating the various sterilized active
ingredients into a sterile vehicle which contains the basic dispersion medium
and the appropriate other ingredients
from those enumerated above. In the case of sterile powders for the
preparation of sterile injectable solutions, certain
methods of preparation are vacuum-drying and freeze-drying techniques which
yield a powder of the active
ingredient plus any additional ingredient from a previously sterile-filtered
solution thereof.
[00464]
The injectable formulations can be sterilized, for example, by filtration
through a bacterial¨
retaining filter, or by incorporating sterilizing agents in the form of
sterile solid compositions which can be
dissolved or dispersed in sterile water or other sterile injectable medium
prior to use. Injectable compositions can
contain from about 0.1 to about 5% w/w of a compound as disclosed herein.
1C. Formulations for Topical Administration
[00465]
In some embodiments, provided herein are pharmaceutical compositions for
topical (e.g.,
transdermal) administration containing a compound as disclosed herein, and a
pharmaceutical excipient suitable for
topical administration. In some embodiments, provided herein are
pharmaceutical compositions for topical
administration containing: (i) an effective amount of a disclosed compound;
optionally (ii) an effective amount of
one or more second agents; and (iii) one or more pharmaceutical excipients
suitable for topical administration. In
some embodiments, the pharmaceutical composition further contains: (iv) an
effective amount of a third agent.
[00466]
Pharmaceutical compositions provided herein can be formulated into
preparations in solid,
semi-solid, or liquid forms suitable for local or topical administration, such
as gels, water soluble jellies, creams,
lotions, suspensions, foams, powders, slurries, ointments, solutions, oils,
pastes, suppositories, sprays, emulsions,
saline solutions, dimethylsulfoxide (DMS0)-based solutions. In general,
carriers with higher densities are capable of
providing an area with a prolonged exposure to the active ingredients. In
contrast, a solution formulation can provide
more immediate exposure of the active ingredient to the chosen area.
[00467]
The pharmaceutical compositions also can comprise suitable solid or gel phase
carriers or
excipients, which are compounds that allow increased penetration of, or assist
in the delivery of, therapeutic
molecules across the stratum corneum permeability barrier of the skin. There
are many of these penetration-
enhancing molecules known to those trained in the art of topical formulation.
Examples of such carriers and
excipients include, but are not limited to, humectants (e.g., urea), glycols
(e.g., propylene glycol), alcohols (e.g.,
ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl
myristate and sodium lauryl sulfate), pyrrolidones,
glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides,
alkanes, alkanols, water, calcium
carbonate, calcium phosphate, various sugars, starches, cellulose derivatives,
gelatin, and polymers such as
polyethylene glycols.
[00468]
Another exemplary formulation for use in the disclosed methods employs
transdermal
delivery devices ("patches"). Such transdermal patches can be used to provide
continuous or discontinuous infusion
of a compound as provided herein in controlled amounts, either with or without
another agent.
116

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00469]
The construction and use of transdermal patches for the delivery of
pharmaceutical agents is
well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and
5,001,139. Such patches can be constructed
for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
[00470]
Suitable devices for use in delivering intradermal pharmaceutically acceptable
compositions
described herein include short needle devices such as those described in U.S.
Patents 4,886,499; 5,190,521;
5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662.
Intradermal compositions can be
administered by devices which limit the effective penetration length of a
needle into the skin, such as those
described in PCT publication WO 99/34850 and functional equivalents thereof.
Jet injection devices which deliver
liquid vaccines to the dermis via a liquid jet injector and/or via a needle
which pierces the stratum corneum and
produces a jet which reaches the dermis are suitable. Jet injection devices
are described, for example, in U.S.
Patents 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189;
5,704,911; 5,383,851; 5,893,397;
5,466,220; 5,339,163; 5,312,335; 5,503,627; 5,064,413; 5,520,639; 4,596,556;
4,790,824; 4,941,880; 4,940,460; and
PCT publications WO 97/37705 and WO 97/13537. Ballistic powder/particle
delivery devices which use
compressed gas to accelerate vaccine in powder form through the outer layers
of the skin to the dermis are suitable.
Alternatively or additionally, conventional syringes can be used in the
classical mantoux method of intradermal
administration.
[00471]
Topically¨administrable formulations can, for example, comprise from about 1%
to about 10%
(w/w) compound of formula (I), although the concentration of the compound of
formula (I) can be as high as the
solubility limit of the compound of formula (I) in the solvent. In some
embodiments, topically¨administrable
formulations can, for example, comprise from about 1% to about 9% (w/w)
compound of formula (I), such as from
about 1% to about 8% (w/w), further such as from about 1% to about 7% (w/w),
further such as from about 1% to
about 6% (w/w), further such as from about 1% to about 5% (w/w), further such
as from about 1% to about 4%
(w/w), further such as from about 1% to about 3% (w/w), and further such as
from about 1% to about 2% (w/w)
compound of formula (I). Formulations for topical administration can further
comprise one or more of the
additional pharmaceutically acceptable excipients described herein.
1D. Formulations for Inhalation Administration
[00472]
In some embodiments, provided herein are pharmaceutical compositions for
inhalation
administration containing a compound as disclosed herein, and a pharmaceutical
excipient suitable for topical
administration. In some embodiments, provided herein are pharmaceutical
compositions for inhalation
administration containing: (i) an effective amount of a disclosed compound;
optionally (ii) an effective amount of
one or more second agents; and (iii) one or more pharmaceutical excipients
suitable for inhalation administration. In
some embodiments, the pharmaceutical composition further contains: (iv) an
effective amount of a third agent.
[00473]
Pharmaceutical compositions for inhalation or insufflation include solutions
and suspensions in
pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof,
and powders. The liquid or solid
pharmaceutical compositions can contain suitable pharmaceutically acceptable
excipients as described herein. In
some embodiments, the pharmaceutical compositions are administered by the oral
or nasal respiratory route for local
117

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
or systemic effect. Pharmaceutical compositions in pharmaceutically acceptable
solvents can be nebulized by use of
inert gases. Nebulized solutions can be inhaled directly from the nebulizing
device or the nebulizing device can be
attached to a face mask tent, or intermittent positive pressure breathing
machine. Solution, suspension, or powder
pharmaceutical compositions can be administered, e.g., orally or nasally, from
devices that deliver the formulation
in an appropriate manner.
1E. Formulations for Ocular Administration
[00474] In some embodiments, the disclosure provides a pharmaceutical
composition for treating
ophthalmic disorders. The pharmaceutical composition can contain an effective
amount of a compound as disclosed
herein and a pharmaceutical excipient suitable for ocular administration.
Pharmaceutical compositions suitable for
ocular administration can be presented as discrete dosage forms, such as drops
or sprays each containing a
predetermined amount of an active ingredient a solution, or a suspension in an
aqueous or non-aqueous liquid, an
oil-in-water emulsion, or a water-in-oil liquid emulsion. Other administration
foms include intraocular injection,
intravitreal injection, topically, or through the use of a drug eluting
device, microcapsule, implant, or microfluidic
device. In some cases, the compounds as disclosed herein are administered with
a carrier or excipient that increases
the intraocular penetrance of the compound such as an oil and water emulsion
with colloid particles having an oily
core surrounded by an interfacial film. It is contemplated that all local
routes to the eye can be used including
topical, subconjunctival, periocular, retrobulbar, subtenon, intracameral,
intravitreal, intraocular, subretinal,
juxtascleral and suprachoroidal administration. Systemic or parenteral
administration can be feasible including, but
not limited to, intravenous, subcutaneous, and oral delivery. An exemplary
method of administration will be
intravitreal or subtenon injection of solutions or suspensions, or
intravitreal or subtenon placement of bioerodible or
non-bioerodible devices, or by topical ocular administration of solutions or
suspensions, or posterior juxtascleral
administration of a gel or cream formulation.
[00475] Eye drops can be prepared by dissolving the active ingredient in
a sterile aqueous solution such
as physiological saline, buffering solution, etc., or by combining powder
compositions to be dissolved before use.
Other vehicles can be chosen, as is known in the art, including, but not
limited to: balance salt solution, saline
solution, water soluble polyethers such as polyethyene glycol, polyvinyls,
such as polyvinyl alcohol and povidone,
cellulose derivatives such as methylcellulose and hydroxypropyl
methylcellulose, petroleum derivatives such as
mineral oil and white petrolatum, animal fats such as lanolin, polymers of
acrylic acid such as
carboxypolymethylene gel, vegetable fats such as peanut oil and
polysaccharides such as dextrans, and
glycosaminoglycans such as sodium hyaluronate. In some embodiments, additives
ordinarily used in the eye drops
can be added. Such additives include isotonizing agents (e.g., sodium
chloride, etc.), buffer agent (e.g., boric acid,
sodium monohydrogen phosphate, sodium dihydrogen phosphate, etc.),
preservatives (e.g., benzalkonium chloride,
benzethonium chloride, chlorobutanol, etc.), thickeners (e.g., saccharide such
as lactose, mannitol, maltose, etc.;
e.g., hyaluronic acid or its salt such as sodium hyaluronate, potassium
hyaluronate, etc.; e.g., mucopolysaccharide
such as chondroitin sulfate, etc.; e.g., sodium polyacrylate, carboxyvinyl
polymer, crosslinked polyacrylate,
polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl
methylcellulose, hydroxyethyl cellulose,
carboxymethyl cellulose, hydroxy propyl cellulose or other agents known to
those skilled in the art).
118

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00476]
In some cases, the colloid particles include at least one cationic agent and
at least one non-ionic
sufactant such as a poloxamer, tyloxapol, a polysorbate, a polyoxyethylene
castor oil derivative, a sorbitan ester, or a
polyoxyl stearate. In some cases, the cationic agent is an alkylamine, a
tertiary alkyl amine, a quarternary
ammonium compound, a cationic lipid, an amino alcohol, a biguanidine salt, a
cationic compound or a mixture
thereof. In some cases, the cationic agent is a biguanidine salt such as
chlorhexidine, polyaminopropyl biguanidine,
phenformin, alkylbiguanidine, or a mixture thereof. In some cases, the
quaternary ammonium compound is a
benzalkonium halide, lauralkonium halide, cetrimide,
hexadecyltrimethylammonium halide,
tetradecyltrimethylammonium halide, dodecyltrimethylammonium halide,
cetrimonium halide, benzethonium
halide, behenalkonium halide, cetalkonium halide, cetethyldimonium halide,
cetylpyridinium halide,
benzododecinium halide, chlorallyl methenamine halide, rnyristylalkonium
halide, stearalkonium halide or a
mixture of two or more thereof. In some cases, cationic agent is a
benzalkonium chloride, lauralkonium chloride,
benzododecinium bromide, benzethenium
chloride, hexadecyltrimethylammonium bromide,
tetradecyltrimethylammonium bromide, dodecyltrimethylammonium bromide or a
mixture of two or more thereof.
In some cases, the oil phase is mineral oil and light mineral oil, medium
chain triglycerides (MCT), coconut oil;
hydrogenated oils comprising hydrogenated cottonseed oil, hydrogenated palm
oil, hydrogenate castor oil or
hydrogenated soybean oil; polyoxyethylene hydrogenated castor oil derivatives
comprising poluoxy1-40
hydrogenated castor oil, polyoxy1-60 hydrogenated castor oil or polyoxyl-100
hydrogenated castor oil.
1F. Formulations for Controlled Release Administration
[00477]
In some embodiments, provided herein are pharmaceutical compositions for
controlled release
administration containing a compound as disclosed herein, and a pharmaceutical
excipient suitable for controlled
release administration. In some embodiments, provided herein are
pharmaceutical compositions for controlled
release administration containing: (i) an effective amount of a disclosed
compound; optionally (ii) an effective
amount of one or more second agents; and (iii) one or more pharmaceutical
excipients suitable for controlled release
administration. In some embodiments, the pharmaceutical composition further
contains: (iv) an effective amount of
a third agent.
[00478]
Active agents such as the compounds provided herein can be administered by
controlled
release means or by delivery devices that are well known to those of ordinary
skill in the art. Examples include, but
are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899;
3,536,809; 3,598,123; and 4,008,719;
5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556;
5,639,480; 5,733,566; 5,739,108;
5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324;
6,113,943; 6,197,350; 6,248,363;
6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; 6,699,500
each of which is incorporated herein
by reference. Such dosage forms can be used to provide slow or controlled
release of one or more active agents
using, for example, hydropropylmethyl cellulose, other polymer matrices, gels,
permeable membranes, osmotic
systems, multilayer coatings, microparticles, liposomes, microspheres, or a
combination thereof to provide the
desired release profile in varying proportions. Suitable controlled release
formulations known to those of ordinary
skill in the art, including those described herein, can be readily selected
for use with the active agents provided
119

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
herein. Thus, the pharmaceutical compositions provided encompass single unit
dosage forms suitable for oral
administration such as, but not limited to, tablets, capsules, gelcaps, and
caplets that are adapted for controlled
release.
[00479] All controlled release pharmaceutical products have a common goal
of improving drug therapy
over that achieved by their non controlled counterparts. In some embodiments,
the use of a controlled release
preparation in medical treatment is characterized by a minimum of drug
substance being employed to cure or control
the disease, disorder, or condition in a minimum amount of time. Advantages of
controlled release formulations
include extended activity of the drug, reduced dosage frequency, and increased
subject compliance. In addition,
controlled release formulations can be used to affect the time of onset of
action or other characteristics, such as
blood levels of the drug, and can thus affect the occurrence of side (e.g.,
adverse) effects.
[00480] In some embodiments, controlled release formulations are designed
to initially release an
amount of a compound as disclosed herein that promptly produces the desired
therapeutic effect, and gradually and
continually release other amounts of the compound to maintain this level of
therapeutic or prophylactic effect over
an extended period of time. In order to maintain this constant level of the
compound in the body, the compound
should be released from the dosage form at a rate that will replace the amount
of drug being metabolized and
excreted from the body. Controlled release of an active agent can be
stimulated by various conditions including, but
not limited to, pH, temperature, enzymes, water, or other physiological
conditions or compounds.
[00481] In certain embodiments, the pharmaceutical composition can be
administered using
intravenous infusion, an implantable osmotic pump, a transdermal patch,
liposomes, or other modes of
administration. In one embodiment, a pump can be used (see, Sefton, CRC Crit.
Ref Biomed. Eng. 14:201(1987);
Buchwald et al., Surgery 88:507 (1980); Saudek et al., N Engl. J. Med. 321:574
(1989)). In another embodiment,
polymeric materials can be used. In yet another embodiment, a controlled
release system can be placed in a subject
at an appropriate site determined by a practitioner of skill, i.e., thus
requiring only a fraction of the systemic dose
(see, e.g., Goodson, Medical Applications of Controlled Release, 115-138 (vol.
2, 1984). Other controlled release
systems are discussed in the review by Langer, Science 249:1527-1533 (1990).
The one or more active agents can
be dispersed in a solid inner matrix, e.g., polymethylmethacrylate,
polybutylmethacrylate, plasticized or
unplasticized polyvinylchloride, plasticized nylon, plasticized
polyethyleneterephthalate, natural rubber,
polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-
vinylacetate copolymers, silicone rubbers,
polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers
such as hydrogels of esters of acrylic
and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked
partially hydrolyzed polyvinyl
acetate, that is surrounded by an outer polymeric membrane, e.g.,
polyethylene, polypropylene, ethylene/propylene
copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate
copolymers, silicone rubbers, polydimethyl
siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride,
vinylchloride copolymers with vinyl
acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene
terephthalate, butyl rubber
epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl
acetate/vinyl alcohol terpolymer, and
ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The one
or more active agents then diffuse
120

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
through the outer polymeric membrane in a release rate controlling step. The
percentage of active agent in such
parenteral compositions is highly dependent on the specific nature thereof, as
well as the needs of the subject.
2. Dosage
[00482]
A compound described herein can be delivered in the form of pharmaceutically
acceptable
compositions which comprise a therapeutically effective amount of one or more
compounds described herein and/or
one or more additional therapeutic agents such as a chemotherapeutic,
formulated together with one or more
pharmaceutically acceptable excipients. In some instances, the compound
described herein and the additional
therapeutic agent are administered in separate pharmaceutical compositions and
can (e.g., because of different
physical and/or chemical characteristics) be administered by different routes
(e.g., one therapeutic is administered
orally, while the other is administered intravenously). In other instances,
the compound described herein and the
additional therapeutic agent can be administered separately, but via the same
route (e.g., both orally or both
intravenously). In still other instances, the compound described herein and
the additional therapeutic agent can be
administered in the same pharmaceutical composition.
[00483]
The selected dosage level will depend upon a variety of factors including, for
example, the activity
of the particular compound employed, the route of administration, the time of
administration, the rate of excretion or
metabolism of the particular compound being employed, the rate and extent of
absorption, the duration of the
treatment, other drugs, compounds and/or materials used in combination with
the particular compound employed,
the age, sex, weight, condition, general health and prior medical history of
the patient being treated, and like factors
well known in the medical arts.
[00484]
In general, a suitable daily dose of a compound described herein and/or a
chemotherapeutic
will be that amount of the compound which, in some embodiments, can be the
lowest dose effective to produce a
therapeutic effect. Such an effective dose will generally depend upon the
factors described above. Generally, doses
of the compounds described herein for a patient, when used for the indicated
effects, will range from about 0.0001
mg to about 100 mg per day, or about 0.001 mg to about 100 mg per day, or
about 0.01 mg to about 100 mg per day,
or about 0.1 mg to about 100 mg per day, or about 0.0001 mg to about 500 mg
per day, or about 0.001 mg to about
500 mg per day, or about 0.01 mg to 1000 mg, or about 0.01 mg to about 500 mg
per day, or about 0.1 mg to about
500 mg per day, or about about 1 mg to 50 mg per day, or about 5 mg to 40 mg.
An exemplary dosage is about 10
to 30 mg per day. In some embodiments, for a 70 kg human, a suitable dose
would be about 0.05 to about 7 g/day,
such as about 0.05 to about 2.5 g/day. Actual dosage levels of the active
ingredients in the pharmaceutical
compositions described herein can be varied so as to obtain an amount of the
active ingredient which is effective to
achieve the desired therapeutic response for a particular patient,
composition, and mode of administration, without
being toxic to the patient. In some instances, dosage levels below the lower
limit of the aforesaid range can be more
than adequate, while in other cases still larger doses can be employed without
causing any harmful side effect, e.g.,
by dividing such larger doses into several small doses for administration
throughout the day.
[00485]
In some embodiments, the compounds can be administered daily, every other day,
three times a
week, twice a week, weekly, or bi-weekly. The dosing schedule can include a
"drug holiday," i.e., the drug can be
121

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
administered for two weeks on, one week off, or three weeks on, one week off,
or four weeks on, one week off, etc.,
or continuously, without a drug holiday.
The compounds can be administered orally, intravenously,
intraperitoneally, topically, transdermally, intramuscularly, subcutaneously,
intranasally, sublingually, or by any
other route.
[00486]
In some embodiments, a compound as provided herein is administered in multiple
doses.
Dosing can be about once, twice, three times, four times, five times, six
times, or more than six times per day.
Dosing can be about once a month, about once every two weeks, about once a
week, or about once every other day.
In another embodiment, a compound as disclosed herein and another agent are
administered together about once per
day to about 6 times per day. In another embodiment, the administration of a
compound as provided herein and an
agent continues for less than about 7 days. In yet another embodiment, the
administration continues for more than
about 6, about 10, about 14, about 28 days, about two months, about six
months, or about one year. In some cases,
continuous dosing is achieved and maintained as long as necessary.
[00487]
Administration of the pharmaceutical compositions as disclosed herein can
continue as long
as necessary. In some embodiments, an agent as disclosed herein is
administered for more than about 1, about 2,
about 3, about 4, about 5, about 6, about 7, about 14, or about 28 days. In
some embodiments, an agent as disclosed
herein is administered for less than about 28, about 14, about 7, about 6,
about 5, about 4, about 3, about 2, or about
1 day. In some embodiments, an agent as disclosed herein is administered
chronically on an ongoing basis, e.g., for
the treatment of chronic effects.
[00488]
Since the compounds described herein can be administered in combination with
other
treatments (such as additional chemotherapeutics, radiation or surgery), the
doses of each agent or therapy can be
lower than the corresponding dose for single-agent therapy. The dose for
single-agent therapy can range from, for
example, about 0.0001 to about 200 mg, or about 0.001 to about 100 mg, or
about 0.01 to about 100 mg, or about
0.1 to about 100 mg, or about 1 to about 50 mg per kilogram of body weight per
day.
[00489]
When a compound provided herein, is administered in a pharmaceutical
composition that
comprises one or more agents, and the agent has a shorter half-life than the
compound provided herein unit dose
forms of the agent and the compound provided herein can be adjusted
accordingly.
3. Kits
[00490]
In some embodiments, provided herein are kits. The kits can include a compound
or
pharmaceutical composition as described herein, in suitable packaging, and
written material that can include
instructions for use, discussion of clinical studies, listing of side effects,
and the like. Such kits can also include
information, such as scientific literature references, package insert
materials, clinical trial results, and/or summaries
of these and the like, which indicate or establish the activities and/or
advantages of the pharmaceutical composition,
and/or which describe dosing, administration, side effects, drug interactions,
or other information useful to the health
care provider. Such information can be based on the results of various
studies, for example, studies using
experimental animals involving in vivo models and studies based on human
clinical trials.
122

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00491] In some embodiments, a memory aid is provided with the kit, e.g.,
in the form of numbers next
to the tablets or capsules whereby the numbers correspond with the days of the
regimen which the tablets or capsules
so specified should be ingested. Another example of such a memory aid is a
calendar printed on the card, e.g., as
follows "First Week, Monday, Tuesday, . . . etc. . . . Second Week, Monday,
Tuesday, . . . "etc. Other variations of
memory aids will be readily apparent. A "daily dose" can be a single tablet or
capsule or several tablets or capsules
to be taken on a given day.
[00492] The kit can further contain another agent. In some embodiments,
the compound as disclosed
herein and the agent are provided as separate pharmaceutical compositions in
separate containers within the kit. In
some embodiments, the compound as disclosed herein and the agent are provided
as a single pharmaceutical
composition within a container in the kit. Suitable packaging and additional
articles for use (e.g., measuring cup for
liquid preparations, foil wrapping to minimize exposure to air, and the like)
are known in the art and can be included
in the kit. In other embodiments, kits can further comprise devices that are
used to administer the active agents.
Examples of such devices include, but are not limited to, syringes, drip bags,
patches, and inhalers. Kits described
herein can be provided, marketed and/or promoted to health providers,
including physicians, nurses, pharmacists,
formulary officials, and the like. Kits can also, in some embodiments, be
marketed directly to the consumer.
[00493] An example of such a kit is a so-called blister pack. Blister
packs are well known in the
packaging industry and are being widely used for the packaging of
pharmaceutical unit dosage forms (tablets,
capsules, and the like). Blister packs generally consist of a sheet of
relatively stiff material covered with a foil of a
preferably transparent plastic material. During the packaging process,
recesses are formed in the plastic foil. The
recesses have the size and shape of the tablets or capsules to be packed.
Next, the tablets or capsules are placed in
the recesses and the sheet of relatively stiff material is sealed against the
plastic foil at the face of the foil which is
opposite from the direction in which the recesses were formed. As a result,
the tablets or capsules are sealed in the
recesses between the plastic foil and the sheet. The strength of the sheet is
such that the tablets or capsules can be
removed from the blister pack by manually applying pressure on the recesses
whereby an opening is formed in the
sheet at the place of the recess. The tablet or capsule can then be removed
via said opening.
[00494] Kits can further comprise pharmaceutically acceptable vehicles
that can be used to administer
one or more active agents. For example, if an active agent is provided in a
solid form that must be reconstituted for
parenteral administration, the kit can comprise a sealed container of a
suitable vehicle in which the active agent can
be dissolved to form a particulate-free sterile solution that is suitable for
parenteral administration. Examples of
pharmaceutically acceptable vehicles include, but are not limited to: Water
for Injection USP; aqueous vehicles
such as, but not limited to, Sodium Chloride Injection, Ringer's Injection,
Dextrose Injection, Dextrose and Sodium
Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles
such as, but not limited to, ethyl
alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous
vehicles such as, but not limited to, corn
oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl
myristate, and benzyl benzoate.
[00495] The present disclosure further encompasses anhydrous
pharmaceutical compositions and
dosage forms comprising an active ingredient, since water can facilitate the
degradation of some compounds. For
example, water can be added (e.g., about 5%) in the pharmaceutical arts as a
means of simulating long-term storage
123

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
in order to determine characteristics such as shelf-life or the stability of
formulations over time. Anhydrous
pharmaceutical compositions and dosage forms can be prepared using anhydrous
or low moisture containing
ingredients and low moisture or low humidity conditions. For example,
pharmaceutical compositions and dosage
forms which contain lactose can be made anhydrous if substantial contact with
moisture and/or humidity during
manufacturing, packaging, and/or storage is expected. An anhydrous
pharmaceutical composition can be prepared
and stored such that its anhydrous nature is maintained. Accordingly,
anhydrous pharmaceutical compositions can
be packaged using materials known to prevent exposure to water such that they
can be included in suitable
formulary kits. Examples of suitable packaging include, but are not limited
to, hermetically sealed foils, plastic or
the like, unit dose containers, blister packs, and strip packs.
Therapeutic Methods
[00496] Phosphoinositide 3-kinases (PI3Ks) are members of a conserved
family of lipid kinases that
regulate numerous cell functions, including proliferation, differentiation,
cell survival and metabolism. Several
classes of PI3Ks exist in mammalian cells, including Class IA subgroup (e.g.,
PI3K-a, 13, (5), which are generally
activated by receptor tyrosine kinases (RTKs); Class IB (e.g., PI3K-7), which
is activated by G-protein coupled
receptors (GPCRs), among others. PI3Ks exert their biological activities via a
"PI3K-mediated signaling pathway"
that includes several components that directly and/or indirectly transduce a
signal triggered by a PI3K, including the
generation of second messenger phophotidylinositol, 3, 4, 5-triphosphate
(PIP3) at the plasma membrane, activation
of heterotrimeric G protein signaling, and generation of further second
messengers such as cAMP, DAG, and IP3,
all of which leads to an extensive cascade of protein kinase activation
(reviewed in Vanhaesebroeck, B. et al. (2001)
Annu Rev Biochem. 70:535-602). For example, PI3K-6 is activated by cellular
receptors through interaction
between the PI3K regulatory subunit (p85) 5H2 domains, or through direct
interaction with RAS. PIP3 produced by
PI3K activates effector pathways downstream through interaction with plextrin
homology (PH) domain containing
enzymes (e.g., PDK-1 and AKT [PKB]). (Fung-Leung WP. (2011) Cell Signal.
23(4):603-8). Unlike PI3K-6, PI3K-
is not associated with a regulatory subunit of the p85 family, but rather with
a regulatory subunit in the p101
family. PI3K-7 is associated with GPCRs, and is responsible for the very rapid
induction of PIP3. PI3K-7 can be
also activated by RAS.
[00497] In some embodiments, provided herein are methods of modulating a
PI3K kinase activity (e.g.,
selectively modulating) by contacting the kinase with an effective amount of a
compound, or a pharmaceutically
acceptable form (e.g., pharmaceutically acceptable salts, hydrates, solvates,
isomers, prodrugs, and isotopically
labeled derivatives) thereof, or pharmaceutical compositions as provided
herein. Modulation can be inhibition (e.g.,
reduction) or activation (e.g., enhancement) of kinase activity. In some
embodiments, provided herein are methods
of inhibiting kinase activity by contacting the kinase with an effective
amount of a compound as provided herein in
solution. In some embodiments, provided herein are methods of inhibiting the
kinase activity by contacting a cell,
tissue, organ that express the kinase of interest with a compound provided
herein. In some embodiments, provided
herein are methods of inhibiting kinase activity in a subject by administering
into the subject an effective amount of
a compound as provided herein. In some embodiments, the kinase activity is
inhibited (e.g., reduced) by more than
124

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
about 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% when contacted with a compound
provided herein as
compared to the kinase activity without such contact. In some embodiments,
provided herein are methods of
inhibiting PI3 kinase activity in a subject (including mammals such as humans)
by contacting said subject with an
amount of a compound as provided herein sufficient to inhibit or reduce the
activity of the PI3 kinase in said subject.
[00498]
In some embodiments, the kinase is a lipid kinase or a protein kinase. In some
embodiments, the
kinase is selected from a PI3 kinase including different isoforms such as PI3
kinase a, PI3 kinase p, PI3 kinase 7,
PI3 kinase 6; DNA-PK; mTor; Abl, VEGFR, Ephrin receptor B4 (EphB4); TEK
receptor tyrosine kinase (TIE2);
FMS-related tyrosine kinase 3 (FLT-3); Platelet derived growth factor receptor
(PDGFR); RET; ATM; ATR; hSmg-
1; Hck; Src; Epidermal growth factor receptor (EGFR); KIT; Inulsin Receptor
(IR); and IGFR.
[00499]
As used herein, a "PI3K-mediated disorder" refers to a disease or condition
involving aberrant
PI3K-mediated signaling pathway. In one embodiment, provided herein is a
method of treating a PI3K mediated
disorder in a subject, the method comprising administering a therapeutically
effective amount of a compound or a
pharmaceutical composition as provided herein. In some embodiments, provided
herein is a method of treating a
PI3K-6 or PI3K-7 mediated disorder in a subject, the method comprising
administering a therapeutically effective
amount of a compound or a pharmaceutical composition as provided herein. In
some embodiments, provided herein
is a method for inhibiting at least one of PI3K-6 and PI3K-7, the method
comprising contacting a cell expressing
PI3K in vitro or in vivo with an effective amount of the compound or
composition provided herein. PI3Ks have
been associated with a wide range of conditions, including immunity, cancer
and thrombosis (reviewed in
Vanhaesebroeck, B. et al. (2010) Current Topics in Microbiology and
Immunology, DOI 10.1007/82_2010_65). For
example, Class I PI3Ks, particularly PI3K-7 and PI3K-6 isoforms, are highly
expressed in leukocytes and have been
associated with adaptive and innate immunity; thus, these PI3Ks are believed
to be important mediators in
inflammatory disorders and hematologic malignancies (reviewed in Harris, SJ et
al. (2009) Curr Opin Investig
Drugs 10(11):1151-62); Rommel C. et al. (2007) Nat Rev Immunol 7(3):191-201;
Durand CA et al. (2009) J
Immunol. 183(9):5673-84; Dil N, Marshall AJ. (2009) Mol Immunol. 46(10):1970-
8; Al-Alwan MM et al. (2007) J
Immunol. 178(4):2328-35; Zhang TT, et al. (2008) J Allergy Clin Immunol.
2008;122(4):811-819.e2; Srinivasan L,
et al. (2009) Cell 139(3):573-86).
[00500]
Numerous publications support roles of PI3K-6, PI3K-7, and PI3K-I3 in the
differentiation,
maintenance, and activation of immune and malignant cells, as described in
more detail below.
[00501]
The importance of PI3K-6 in the development and function of B-cells is
supported from inhibitor
studies and genetic models. PI3K-6 is an important mediator of B-cell receptor
(BCR) signaling, and is upstream of
AKT, calcium flux, PLC7, MAP kinase, P70S6k, and FOX03a activation. PI3K-6 is
also important in IL4R, S1P,
and CXCR5 signaling, and has been shown to modulate responses to toll-like
receptors 4 and 9. Inhibitors of P13 K-
(5 have shown the importance of PI3K-6 in B-cell development (Marginal zone
and B1 cells), B-cell activation,
chemotaxis, migration and homing to lymphoid tissue, and in the control of
immunoglobulin class switching
leading to the production of IgE. Clayton E et al. (2002) J Exp Med.
196(6):753-63; Bilancio A, et al. (2006) Blood
107(2):642-50; Okkenhaug K. et al. (2002) Science 297(5583):1031-4; Al-Alwan
MM et al. (2007) J Immunol.
125

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
178(4):2328-35; Zhang TT, et al. (2008) J Allergy Clin Immunol.
2008;122(4):811-819.e2; Srinivasan L, et al.
(2009) Cell 139(3):573-86)
[00502] In T-cells, PI3K-6 has been demonstrated to have a role in T-cell
receptor and cytokine signaling,
and is upstream of AKT, PLC7, and GSK3b. In PI3K-6 deletion or kinase-dead
knock-in mice, or in inhibitor
studies, T-cell defects including proliferation, activation, and
differentiation have been observed, leading to reduced
T helper cell 2 (TH2) response, memory T-cell specific defects (DTH
reduction), defects in antigen dependent
cellular trafficking, and defects in chemotaxis/migration to chemokines (e.g.,
SIP, CCR7, CD62L). (Garcon F. et al.
(2008) Blood 111(3):1464-71; Okkenhaug K et al. (2006). J Immunol. 177(8):5122-
8; Soond DR, et al. (2010)
Blood 115(11):2203-13; Reif K, (2004). J Immunol. 2004;173(4):2236-40; Ji H.
et al. (2007) Blood 110(8):2940-7;
Webb LM, et al. (2005) J Immunol. 175(5):2783-7; Liu D, et al. (2010) J
Immunol. 184(6):3098-105; Haylock-
Jacobs S, et al. (2011)J Autoimmun. 2011;36(3-4):278-87; Jarmin SJ, et al.
(2008)J Clin Invest. 118(3):1154-64).
[00503] In neutrophils, PI3K-6 along with PI3K-7, and PI3K-I3, contribute
to the responses to immune
complexes, FC7RII signaling, including migration and neutrophil respiratory
burst. Human neutrophils undergo
rapid induction of PIP3 in response to formyl peptide receptor (FMLP) or
complement component C5a (C5a) in a
PI3K-7 dependent manner, followed by a longer PIP3 production period that is
PI3K-6 dependent, and is essential
for respiratory burst. The response to immune complexes is contributed by PI3K-
6, PI3K-7, and PI3K-I3, and is an
important mediator of tissue damage in models of autoimmune disease (Randis TM
et al. (2008) Eur J Immunol.
38(5):1215-24; Pinho V, (2007) J Immunol. 179(11):7891-8; Sadhu C. et al.
(2003) J Immunol. 170(5):2647-54;
Condliffe AM et al. (2005) Blood 106(4):1432-40). It has been reported that in
certain autoimmune diseases,
perfrential activation of PI3KI3 may be involved. (Kulkarni et al., Immunology
(2011) 4(168) ra23: 1-11). It was
also reported that PI3KI3-deficient mice were highly protected in an Fc7R-
dependent model of autoantibody-induced
skin blistering and partially protected in an Fc7R-dependent model of
inflammatory arthritis, whereas combined
deficiency of PI3KI3 and PI3K6 resulted in near complete protection in
inflammatory arthritis. (Id.).
[00504] In macrophages collected from patients with chronic obstructive
pulmonary disease (COPD),
glucocorticoid responsiveness can be restored by treatment of the cells with
inhibitors of PI3K-43. Macrophages also
rely on PI3K-6 and PI3K-7 for responses to immune complexes through the arthus
reaction (FCgR and C5a
signaling) (Randis TM, et al. (2008) Eur J Immunol. 38(5):1215-24 ; Marwick JA
et al. (2009) Am J Respir Crit
Care Med. 179(7):542-8; Konrad S, et al. (2008)J Biol Chem. 283(48):33296-
303).
[00505] In mast cells, stem cell factor- (SCF) and 1L3-dependent
proliferation, differentiation and function
are PI3K-6 dependent, as is chemotaxis. The allergen/IgE crosslinking of FCgR1
resulting in cytokine release and
degranulation of the mast cells is severely inhibited by treatment with PI3K-6
inhibitors, suggesting a role for PI3K-
in allergic disease (Ali K et al. (2004) Nature 431(7011):1007-11; Lee KS, et
al. (2006) FASEB J. 20(3):455-65;
Kim MS, et al. (2008) Trends Immunol. 29(10):493 -501).
[00506] Natural killer (NK) cells are dependent on both PI3K-6 and PI3K-7
for efficient migration towards
chemokines including CXCL10, CCL3, SIP and CXCL12, or in response to LPS in
the peritoneum (Guo H, et al.
(2008) J Exp Med. 205(10):2419-35; Tassi I, et al. (2007) Immunity 27(2):214-
27; Saudemont A, (2009) Proc Natl
Acad Sci USA. 106(14):5795-800; Kim N, et al. (2007) Blood 110(9):3202-8).
126

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00507] The roles of PI3K-6, PI3K-7, and PI3K-I3 in the differentiation,
maintenance, and activation of
immune cells support a role for these enzymes in inflammatory disorders
ranging from autoimmune diseases (e.g.,
rheumatoid arthritis, multiple sclerosis) to allergic inflammatory disorders,
such as asthma, and inflammatory
respiratory disease such as COPD. Extensive evidence is available in
experimental animal models, or can be
evaluated using art-recognized animal models. In an embodiment, described
herein is a method of treating
inflammatory disorders ranging from autoimmune diseases (e.g., rheumatoid
arthritis, multiple sclerosis) to allergic
inflammatory disorders, such as asthma and COPD using a compound described
herein.
[00508] For example, inhibitors of PI3K-6 and/or -7 have been shown to
have anti-inflammatory activity in
several autoimmune animal models for rheumatoid arthritis (Williams, 0. et al.
(2010) Chem Biol, 17(2):123-34;
WO 2009/088986; W02009/088880; WO 2011/008302). PI3K-6 is expressed in the RA
synovial tissue (especially
in the synovial lining which contains fibroblast-like synoviocytes (FLS), and
selective PI3K-6 inhibitors have been
shown to be effective in inhibiting synoviocyte growth and survival (Bartok et
al. (2010) Arthritis Rheum 62 Suppl
10:362). Several PI3K-6 and -7 inhibitors have been shown to ameliorate
arthritic symptoms (e.g., swelling of
joints, reduction of serum-induced collagen levels, reduction of joint
pathology and/or inflammation), in art-
recognized models for RA, such as collagen-induced arthritis and adjuvant
induced arthritis (WO 2009/088986;
W02009/088880; WO 2011/008302).
[00509] The role of PI3K-6 has also been shown in models of T-cell
dependent response, including the
DTH model. In the murine experimental autoimmune encephalomyelitis (EAE) model
of multiple sclerosis, the
PI3K-7/6- double mutant mice are resistant. PI3K-6 inhibitors have also been
shown to block EAE disease
induction and development of TH-17 cells both in vitro and in vivo (Haylock-
Jacobs, S. et al. (2011) J.
Autoimmunity 36(3-4):278-87).
[00510] Systemic lupus erythematosus (SLE) is a complex disease that at
different stages requires memory
T-cells, B-cell polyclonal expansion and differentiation into plasma cells,
and the innate immune reasponse to
endogenous damage associated molecular pattern molecules (DAMPS), and the
inflammatory responses to immune
complexes through the complement system as well as the Fc receptors. The role
of PI3K-6 and PI3K-7 together in
these pathways and cell types suggest that blockade with an inhibitor would be
effective in these diseases. A role
for PI3K in lupus is also predicted by two genetic models of lupus. The
deletion of phosphatase and tensin homolog
(PTEN) leads to a lupus-like phenotype, as does a transgenic activation of
ClasslA PI3Ks, which includes PI3K-43.
The deletion of PI3K-7 in the transgenically activated class lA lupus model is
protective, and treatment with a
PI3K-7 selective inhibitor in the murine MLR//pr model of lupus improves
symptoms (Barber, DF et al. (2006) J.
Immunol. 176(1): 589-93).
[00511] In allergic disease, PI3K-6 has been shown by genetic models and
by inhibitor treatment to be
essential for mast-cell activation in a passive cutaneous anaphalaxis assay
(Ali K et al. (2008) J Immunol.
180(4):2538-44; Ali K, (2004) Nature 431(7011):1007-11). In a pulmonary
measure of response to immune
complexes (Arthus reaction) a PI3K-6 knockout is resistant, showing a defect
in macrophage activation and C5a
production. Knockout studies and studies with inhibitors for both PI3K-6 and
PI3K-7 support a role for both of
these enzymes in the ovalbumin induced allergic airway inflammation and hyper-
responsiveness model (Lee KS et
127

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
al. (2006) FASEB J. 20(3):455-65). Reductions of infiltration of eosinophils,
neutrophils, and lymphocytes as well
as TH2 cytokines (IL4, IL5, and IL13) were seen with both PI3K-6 specific and
dual PI3K-6 and PI3K-7 inhibitors
in the Ova induced asthma model (Lee KS et al. (2006)J Allergy Clin Immunol
118(2):403-9).
[00512] PI3K-6 and PI3K-7 inhibition can be used in treating COPD. In the
smoked mouse model of
COPD, the PI3K-6 knockout does not develop smoke induced glucocorticoid
resistance, while wild-type and PI3K-7
knockout mice do. An inhaled formulation of dual PI3K-6 and PI3K-7 inhibitor
blocked inflammation in a LPS or
smoke COPD models as measured by neutrophilia and glucocorticoid resistance
(Doukas J, et al. (2009) J
Pharmacol Exp Ther. 328(3):758-65).
[00513] Class I PI3Ks, particularly PI3K-6 and PI3K-7 isoforms, are also
associated with cancers
(reviewed, e.g., in Vogt, PK et al. (2010) Curr Top Microbiol Immunol. 347:79-
104; Fresno Vara, JA et al. (2004)
Cancer Treat Rev. 30(2):193-204; Zhao, L and Vogt, PK. (2008) Oncogene
27(41):5486-96). Inhibitors of PI3K,
e.g., PI3K-6 and/or -7, have been shown to have anti-cancer activity (e.g.,
Courtney, KD et al. (2010) J Clin Oncol.
28(6):1075-1083); Markman, B et al. (2010) Ann Oncol. 21(4):683-91; Kong, D
and Yamori, T (2009) Curr Med
Chem. 16(22):2839-54; Jimeno, Act al. (2009) J Clin Oncol. 27:156s (suppl;
abstr 3542); Flinn, IW et al. (2009) J
Clin Oncol. 27:156s (suppl; abstr 3543); Shapiro, Get al. (2009) J Clin Oncol.
27:146s (suppl; abstr 3500); Wagner,
AJ et al. (2009)J Clin Oncol. 27:146s (suppl; abstr 3501); Vogt, PK et al.
(2006) Virology 344(1):131-8; Ward, Set
al. (2003) Chem Biol. 10(3):207-13; WO 2011/041399; US 2010/0029693; US
2010/0305096; US 2010/0305084).
In an embodiment, described herein is a method of treating cancer.
[00514] Types of cancer that can be treated with an inhibitor of PI3K
(particularly, PI3K-6 and/or -7)
include, e.g., leukemia, chronic lymphocytic leukemia, acute myeloid leukemia,
chronic myeloid leukemia (e.g.,
Salmena, L et al. (2008) Cell 133:403-414; Chapuis, N et al. (2010) Clin
Cancer Res. 16(22):5424-35; Khwaja, A
(2010) Curr Top Microbiol Immunol. 347:169-88); lymphoma, e.g., non-Hodgkin's
lymphoma (e.g., Salmena, L et
al. (2008) Cell 133:403-414); lung cancer, e.g., non-small cell lung cancer,
small cell lung cancer (e.g., Herrera, VA
et al. (2011) Anticancer Res. 31(3):849-54); melanoma (e.g., Haluska, F et al.
(2007) Semin Oncol. 34(6):546-54);
prostate cancer (e.g., Sarker, D et al. (2009) Clin Cancer Res. 15(15):4799-
805); glioblastoma (e.g., Chen, JS et al.
(2008) Mol Cancer Ther. 7:841-850); endometrial cancer (e.g., Bansal, N et al.
(2009) Cancer Control. 16(1):8-13);
pancreatic cancer (e.g., Furukawa, T (2008) J Gastroenterol. 43(12):905-11);
renal cell carcinoma (e.g., Porta, C
and Figlin, RA (2009) J Urol. 182(6):2569-77); colorectal cancer (e.g., Saif,
MW and Chu, E (2010) Cancer J.
16(3):196-201); breast cancer (e.g., Torben, NE et al. (2008) Biochem J.
415:97-100); thyroid cancer (e.g.,
Brzezianska, E and Pastuszak-Lewandoska, D (2011) Front Biosci. 16:422-39);
and ovarian cancer (e.g., Mazzoletti,
M and Broggini, M (2010) Curr Med Chem. 17(36):4433-47).
[00515] Numerous publications support a role of PI3K-6 and PI3K-7 in
treating hematological cancers.
PI3K-6 and PI3K-7 are highly expressed in the heme compartment, and some solid
tumors, including prostate, breast
and glioblastomas (Chen J.S. et al. (2008) Mol Cancer Ther. 7(4):841-50; Ikeda
H. et al. (2010) Blood 116(9):1460-
8).
[00516] In hematological cancers including acute myeloid leukemia (AML),
multiple myeloma (MM), and
chronic lymphocytic leukemia (CLL), overexpression and constitutive activation
of PI3K-6 supports the model that
128

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
PI3K-6 inhibition would be therapeutic Billottet C, et al. (2006) Oncogene
25(50):6648-59; Billottet C, et al. (2009)
Cancer Res. 69(3):1027-36; Meadows, SA, 52nd Annual ASH Meeting and
Exposition; 2010 Dec 4-7; Orlando, FL;
Ikeda H, et al. (2010) Blood 116(9):1460-8; Herman SE et al. (2010) Blood
116(12):2078-88; Herman SE et al.
(2011). Blood 117(16):4323-7. In an embodiment, described herein is a method
of treating hematological cancers
including, but not limited to acute myeloid leukemia (AML), multiple myeloma
(MM), and chronic lymphocytic
leukemia (CLL).
[00517]
A PI3K-6 inhibitor (CAL-101) has been evaluated in a phase 1 trial in patients
with
haematological malignancies, and showed activity in CLL in patients with poor
prognostic characteristics. In CLL,
inhibition of PI3K-6 not only affects tumor cells directly, but it also
affects the ability of the tumor cells to interact
with their microenvironment. This microenvironment includes contact with and
factors from stromal cells, T-cells,
nurse like cells, as well as other tumor cells. CAL-101 suppresses the
expression of stromal and T-cell derived
factors including CCL3, CCL4, and CXCL13, as well as the CLL tumor cells'
ability to respond to these factors.
CAL-101 treatment in CLL patients induces rapid lymph node reduction and
redistribution of lymphocytes into the
circulation, and affects tonic survival signals through the BCR, leading to
reduced cell viability, and an increase in
apoptosis. Single agent CAL-101 treatment was also active in mantle cell
lymphoma and refractory non Hodgkin's
lymphoma (Furman, RR, et al. 52nd Annual ASH Meeting and Exposition; 2010 Dec
4-7; Orlando, FL;
Hoellenriegel, J, et al. 52nd Annual ASH Meeting and Exposition; 2010 Dec 4-7;
Orlando, FL; Webb, HK, et al. 52nd
Annual ASH Meeting and Exposition; 2010 Dec 4-7; Orlando, FL; Meadows, et al.
52nd Annual ASH Meeting and
Exposition; 2010 Dec 4-7; Orlando, FL; Kahl, B, et al. 52nd Annual ASH Meeting
and Exposition; 2010 Dec 4-7;
Orlando, FL; Lannutti BJ, et al. (2011) Blood 117(2):591-4).
[00518]
PI3K-6 inhibitors have shown activity against PI3K-6 positive gliomas in vitro
(Kashishian A, et
al. Poster presented at: The American Association of Cancer Research 102nd
Annual Meeting; 2011 Apr 2-6;
Orlando, FL). PI3K-6 is the PI3K isoform that is most commonly activated in
tumors where the PTEN tumor
suppressor is mutated (Ward S, et al. (2003) Chem Biol. 10(3):207-13). In this
subset of tumors, treatment with the
PI3K-6 inhibitor either alone or in combination with a cytotoxic agent can be
effective.
[00519]
Another mechanism for PI3K-6 inhibitors to have an affect in solid tumors
involves the tumor
cells' interaction with their micro-environment. PI3K-6, PI3K-7, and PI3K-I3
are expressed in the immune cells that
infiltrate tumors, including tumor infiltrating lymphocytes, macrophages, and
neutrophils. PI3K-6 inhibitors can
modify the function of these tumor-associated immune cells and how they
respond to signals from the stroma, the
tumor, and each other, and in this way affect tumor cells and metastasis
(Hoellenriegel, J, et al. 52nd Annual ASH
Meeting and Exposition; 2010 Dec 4-7; Orlando, FL).
[00520]
PI3K-6 is also expressed in endothelial cells. It has been shown that tumors
in mice treated
with PI3K-6 selective inhibitors are killed more readily by radiation therapy.
In this same study, capillary network
formation is impaired by the PI3K inhibitor, and it is postulated that this
defect contributes to the greater killing with
radiation. PI3K-6 inhibitors can affect the way in which tumors interact with
their microenviroment, including
stromal cells, immune cells, and endothelial cells and be therapeutic either
on its own or in conjunction with another
129

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
therapy (Meadows, SA, et al. Paper presented at: 52nd Annual ASH Meeting and
Exposition; 2010 Dec 4-7;
Orlando, FL; Geng L, et al. (2004) Cancer Res. 64(14):4893-9).
[00521] In other embodiments, inhibition of PI3K (such as PI3K-43 and/or
¨y) can be used to treat a
neuropsychiatric disorder, e.g., an autoimmune brain disorder. Infectious and
immune factors have been implicated
in the pathogenesis of several neuropsychiatric disorders, including, but not
limited to, Sydenham's chorea (SC)
(Garvey, M.A. et al. (2005)J. Child Neurol. 20:424-429), Tourette's syndrome
(TS), obsessive compulsive disorder
(OCD) (Asbahr, F.R. et al. (1998) Am. J. Psychiatry 155:1122-1124), attention
deficit/hyperactivity disorder
(AD/HD) (Hirschtritt, M.E. et al. (2008) Child Neuropsychol. 1:1-16; Peterson,
B.S. et al. (2000) Arch. Gen.
Psychiatry 57:364-372), anorexia nervosa (Sokol, M.S. (2000)J. Child Adolesc.
Psychopharmacol. 10:133-145;
Sokol, M.S. et al. (2002)Am. J. Psychiatry 159:1430-1432), depression (Leslie,
D.L. et al. (2008) J Am. Acad.
Child Adolesc. Psychiatry 47:1166-1172), and autism spectrum disorders (ASD)
(Hollander, E. et al. (1999) Am. J.
Psychiatry 156:317-320; Margutti, P. et al. (2006) Curr. Neurovasc. Res. 3:149-
157). A subset of childhood
obsessive compulsive disorders and tic disorders has been grouped as Pediatric
Autoimmune Neuropsychiatric
Disorders Associated with Streptococci (PANDAS). PANDAS disorders provide an
example of disorders where the
onset and exacerbation of neuropsychiatric symptoms is preceded by a
streptococcal infection (Kurlan, R., Kaplan,
E.L. (2004) Pediatrics 113:883-886; Garvey, M.A. et al. (1998) J Clin. Neurol.
13:413-423). Many of the
PANDAS disorders share a common mechanism of action resulting from antibody
responses against streptococcal
associated epitopes, such as GlcNAc, which produces neurological effects
(Kirvan. C.A. et al. (2006)J
Neuroimmunol. 179:173-179). Autoantibodies recognizing central nervous system
(CNS) epitopes are also found in
sera of most PANDAS subjects (Yaddanapudi, K. et al. (2010) Mol. Psychiatry
15:712-726). Thus, several
neuropsychiatric disorders have been associated with immune and autoimmune
components, making them suitable
for therapies that include PI3K-43 and/or --)1 inhibition.
[00522] In certain embodiments, a method of treating (e.g., reducing or
ameliorating one or more
symptoms of) a neuropsychiatric disorder, (e.g., an autoimmune brain
disorder), using a PI3K-43 and/or --)1 inhibitor
is described, alone or in combination therapy. For example, one or more PI3K-
43 and/or --)1 inhibitors described
herein can be used alone or in combination with any suitable therapeutic agent
and/or modalities, e.g., dietary
supplement, for treatment of neuropsychiatric disorders. Exemplary
neuropsychiatric disorders that can be treated
with the PI3K-43 and/or --)1 inhibitors described herein include, but are not
limited to, PANDAS disorders,
Sydenham's chorea, Tourette's syndrome, obsessive compulsive disorder,
attention deficit/hyperactivity disorder,
anorexia nervosa, depression, and autism spectrum disorders. Pervasive
Developmental Disorder (PDD) is an
exemplary class of autism spectrum disorders that includes Autistic Disorder,
Asperger's Disorder, Childhood
Disintegrative Disorder (CDD), Rett's Disorder and PDD-Not Otherwise Specified
(PDD-NOS). Animal models for
evaluating the activity of the PI3K-43 and/or --)1 inhibitor are known in the
art. For example, a mouse model of
PANDAS disorders is described in, e.g., Yaddanapudi, K. et al. (2010) supra;
and Hoffman, K.I. et al. (2004)J.
Neurosci. 24:1780-1791.
[00523] In some embodiments, provided herein are methods of using the
compounds, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
130

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein to treat disease
conditions, including, but not limited to, diseases associated with
malfunctioning of one or more types of PI3 kinase.
A detailed description of conditions and disorders mediated by p1106 kinase
activity is set forth in Sadu et al., WO
01/81346, which is incorporated herein by reference in its entirety for all
purposes.
[00524]
In some embodiments, the disclosure relates to a method of treating a
hyperproliferative
disorder in a subject that comprises administering to said subject a
therapeutically effective amount of a compound,
or a pharmaceutically acceptable form (e.g., pharmaceutically acceptable
salts, hydrates, solvates, isomers, prodrugs,
and isotopically labeled derivatives) thereof, or pharmaceutical compositions
as provided herein. In some
embodiments, said method relates to the treatment of cancer such as acute
myeloid leukemia, thymus, brain, lung,
squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity
and oropharyngeal, bladder, gastric,
stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney,
liver, ovarian, prostate, colorectal,
esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS-related (e.g.,
Lymphoma and Kaposi's Sarcoma) or
viral-induced cancer. In some embodiments, said method relates to the
treatment of a non-cancerous
hyperproliferative disorder such as benign hyperplasia of the skin (e. g.,
psoriasis), restenosis, or prostate (e. g.,
benign prostatic hypertrophy (BPH)).
[00525]
Patients that can be treated with compounds, or a pharmaceutically acceptable
form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, according to the
methods as provided herein include,
for example, but not limited to, patients that have been diagnosed as having
psoriasis; restenosis; atherosclerosis;
BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary
gland, medullary carcinomas, colloid
carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian
cancer, including epithelial ovarian tumors
such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated
from the ovary into the abdominal
cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix
epithelial including squamous cell
carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer
selected from the following: an
adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic
cancer such as epitheliod carcinoma
in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct;
bladder cancer such as a transitional cell
carcinoma in urinary bladder, urothelial carcinomas (transitional cell
carcinomas), tumors in the urothelial cells that
line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell
cancers; leukemia such as acute
myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic
leukemia, chronic myeloid leukemia,
hairy cell leukemia, myelodysplasia, myeloproliferative disorders, NK cell
leukemia (e.g., blastic plasmacytoid
dendritic cell neoplasm), acute myelogenous leukemia (AML), chronic
myelogenous leukemia (CML),
mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and
myelodysplastic syndrome
(MDS); bone cancer; lung cancer such as non-small cell lung cancer (NSCLC),
which is divided into squamous cell
carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and
small cell lung cancer; skin cancer
such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic
keratosis, which is a skin condition
that sometimes develops into squamous cell carcinoma; eye retinoblastoma;
cutaneous or intraocular (eye)
melanoma; primary liver cancer (cancer that begins in the liver); kidney
cancer; thyroid cancer such as papillary,
131

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
follicular, medullary and anaplastic; lymphoma such as diffuse large B-cell
lymphoma, B-cell immunoblastic
lymphoma, NK cell lymphoma (e.g., blastic plasmacytoid dendritic cell
neoplasm), and small non-cleaved cell
lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus
(HBV), hepatitis C virus (HCV),
and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-1) and
adult T-cell leukemia/lymphoma;
and human papilloma virus (HPV) and cervical cancer; central nervous system
cancers (CNS) such as primary brain
tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or
glioblastoma multiforme),
Oligodendroglioma, Ependymoma, Meningioma, Lymphoma, Schwannoma, and
Medulloblastoma; peripheral
nervous system (PNS) cancers such as acoustic neuromas and malignant
peripheral nerve sheath tumor (MPNST)
including neurofibromas and schwannomas, malignant fibrous cytoma, malignant
fibrous histiocytoma, malignant
meningioma, malignant mesothelioma, and malignant mixed MilHenan tumor; oral
cavity and oropharyngeal cancer
such as, hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and
oropharyngeal cancer; stomach
cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors;
testicular cancer such as germ cell tumors
(GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors,
which include Leydig cell
tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic
carcinomas, Hodgkin disease, non-
Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon
cancer.
[00526]
In one embodiment, provided herein is a method of treating an inflammation
disorder,
including autoimmune diseases in a subject. The method comprises administering
to said subject a therapeutically
effective amount of a compound, or a pharmaceutically acceptable form (e.g.,
pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and isotopically labeled derivatives)
thereof, or pharmaceutical compositions
as provided herein. Examples of autoimmune diseases includes but is not
limited to acute disseminated
encephalomyelitis (ADEM), Addison's disease, antiphospholipid antibody
syndrome (APS), aplastic anemia,
autoimmune hepatitis, autoimmune skin disease, coeliac disease, Crohn's
disease, Diabetes mellitus (type 1),
Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS),
Hashimoto's disease, lupus
eiythematosus, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus
syndrome (OMS), optic neuritis, Ord's
thyroiditis, oemphigus, polyarthritis, primary biliary cirrhosis, psoriasis,
rheumatoid arthritis, Reiter's syndrome,
Takayasu's arteritis, temporal arteritis (also known as "giant cell
arteritis"), warm autoimmune hemolytic anemia,
Wegener's granulomatosis, alopecia universalis (e.g., inflammatory alopecia),
Chagas disease, chronic fatigue
syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial
cystitis, neuromyotonia, sarcoidosis,
scleroderma, ulcerative colitis, vitiligo, and vulvodynia. Other disorders
include bone-resorption disorders and
thrombosis.
[00527]
Inflammation takes on many forms and includes, but is not limited to, acute,
adhesive,
atrophic, catarrhal, chronic, cirrhotic, diffuse, disseminated, exudative,
fibrinous, fibrosing, focal, granulomatous,
hyperplastic, hypertrophic, interstitial, metastatic, necrotic, obliterative,
parenchymatous, plastic, productive,
proliferous, pseudomembranous, purulent, sclerosing, seroplastic, serous,
simple, specific, subacute, suppurative,
toxic, traumatic, and/or ulcerative inflammation.
[00528]
Exemplary inflammatory conditions include, but are not limited to,
inflammation associated
with acne, anemia (e.g., aplastic anemia, haemolytic autoimmune anaemia),
asthma, arteritis (e.g., polyarteritis,
132

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
temporal arteritis, periarteritis nodosa, Takayasu's arteritis), arthritis
(e.g., crystalline arthritis, osteoarthritis,
psoriatic arthritis, gout flare, gouty arthritis, reactive arthritis,
rheumatoid arthritis and Reiter's arthritis), ankylosing
spondylitis, amylosis, amyotrophic lateral sclerosis, autoimmune diseases,
allergies or allergic reactions,
atherosclerosis, bronchitis, bursitis, chronic prostatitis, conjunctivitis,
Chagas disease, chronic obstructive
pulmonary disease, cermatomyositis, diverticulitis, diabetes (e.g., type I
diabetes mellitus, type 2 diabetes mellitus),
a skin condition (e.g., psoriasis, eczema, burns, dermatitis, pruritus
(itch)), endometriosis, Guillain-Barre syndrome,
infection, ischaemic heart disease, Kawasaki disease, glomerulonephritis,
gingivitis, hypersensitivity, headaches
(e.g., migraine headaches, tension headaches), ileus (e.g., postoperative
ileus and ileus during sepsis), idiopathic
thrombocytopenic purpura, interstitial cystitis (painful bladder syndrome),
gastrointestinal disorder (e.g., selected
from peptic ulcers, regional enteritis, diverticulitis, gastrointestinal
bleeding, eosinophilic gastrointestinal disorders
(e.g., eosinophilic esophagitis, eosinophilic gastritis, eosinophilic
gastroenteritis, eosinophilic colitis), gastritis,
diarrhea, gastroesophageal reflux disease (GORD, or its synonym GERD),
inflammatory bowel disease (IBD) (e.g.,
Crohn's disease, ulcerative colitis, collagenous colitis, lymphocytic colitis,
ischaemic colitis, diversion colitis,
Behcet's syndrome, indeterminate colitis) and inflammatory bowel syndrome
(IBS)), lupus, multiple sclerosis,
morphea, myeasthenia gravis, myocardial ischemia, nephrotic syndrome,
pemphigus vulgaris, pernicious aneaemia,
peptic ulcers, polymyositis, primary biliary cirrhosis, neuroinflammation
associated with brain disorders (e.g.,
Parkinson's disease, Huntington's disease, and Alzheimer's disease),
prostatitis, chronic inflammation associated
with cranial radiation injury, pelvic inflammatory disease, polymyalgia
rheumatic, reperfusion injury, regional
enteritis, rheumatic fever, systemic lupus erythematosus, scleroderma,
scierodoma, sarcoidosis,
spondyloarthopathies, Sjogren's syndrome, thyroiditis, transplantation
rejection, tendonitis, trauma or injury (e.g.,
frostbite, chemical irritants, toxins, scarring, burns, physical injury),
vasculitis, vitiligo and Wegener's
granulomatosis. In certain embodiments, the inflammatory disorder is selected
from arthritis (e.g., rheumatoid
arthritis), inflammatory bowel disease, inflammatory bowel syndrome, asthma,
psoriasis, endometriosis, interstitial
cystitis and prostatistis. In certain embodiments, the inflammatory condition
is an acute inflammatory condition
(e.g., for example, inflammation resulting from infection). In certain
embodiments, the inflammatory condition is a
chronic inflammatory condition (e.g., conditions resulting from asthma,
arthritis and inflammatory bowel disease).
The compounds can also be useful in treating inflammation associated with
trauma and non-inflammatory myalgia.
[00529] Immune disorders, such as auto-immune disorders, include, but are
not limited to, arthritis
(including rheumatoid arthritis, spondyloarthopathies, gouty arthritis,
degenerative joint diseases such as
osteoarthritis, systemic lupus erythematosus, Sjogren's syndrome, ankylosing
spondylitis, undifferentiated
spondylitis, Behcet's disease, haemolytic autoimmune anaemias, multiple
sclerosis, amyotrophic lateral sclerosis,
amylosis, acute painful shoulder, psoriatic, and juvenile arthritis), asthma,
atherosclerosis, osteoporosis, bronchitis,
tendonitis, bursitis, skin condition (e.g., psoriasis, eczema, burns,
dermatitis, pruritus (itch)), enuresis, eosinophilic
disease, gastrointestinal disorder (e.g., selected from peptic ulcers,
regional enteritis, diverticulitis, gastrointestinal
bleeding, eosinophilic gastrointestinal disorders (e.g., eosinophilic
esophagitis, eosinophilic gastritis, eosinophilic
gastroenteritis, eosinophilic colitis), gastritis, diarrhea, gastroesophageal
reflux disease (GORD, or its synonym
GERD), inflammatory bowel disease (IBD) (e.g., Crohn's disease, ulcerative
colitis, collagenous colitis,
133

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
lymphocytic colitis, ischaemic colitis, diversion colitis, Behcet's syndrome,
indeterminate colitis) and inflammatory
bowel syndrome (IBS)), relapsing polychondritis (e.g., atrophic polychondritis
and systemic polychondromalacia),
and disorders ameliorated by a gastroprokinetic agent (e.g., ileus,
postoperative ileus and ileus during sepsis;
gastroesophageal reflux disease (GORD, or its synonym GERD); eosinophilic
esophagitis, gastroparesis such as
diabetic gastroparesis; food intolerances and food allergies and other
functional bowel disorders, such as non-
ulcerative dyspepsia (NUD) and non-cardiac chest pain (NCCP, including costo-
chondritis)). In certain
embodiments, a method of treating inflammatory or autoimmune diseases is
provided comprising administering to a
subject (e.g., a mammal) a therapeutically effective amount of a compound, or
a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, that selectively
inhibit PI3K-6 and/or PI3K-7 as
compared to all other type I PI3 kinases.
[00530]
Such selective inhibition of PI3K-6 and/or PI3K-7 can be advantageous for
treating any of the
diseases or conditions described herein. For example, selective inhibition of
PI3K-6 can inhibit inflammatory
responses associated with inflammatory diseases, autoimmune disease, or
diseases related to an undesirable immune
response including, but not limited to asthma, emphysema, allergy, dermatitis,
rheumatoid arthritis, psoriasis, lupus
erythematosus, anaphylaxsis, or graft versus host disease. Selective
inhibition of PI3K-6 can further provide for a
reduction in the inflammatory or undesirable immune response without a
concomittant reduction in the ability to
reduce a bacterial, viral, and/or fungal infection. Selective inhibition of
both PI3K-6 and PI3K-7 can be
advantageous for inhibiting the inflammatory response in the subject to a
greater degree than that would be provided
for by inhibitors that selectively inhibit PI3K -6 or PI3K-7 alone. In one
aspect, one or more of the subject methods
are effective in reducing antigen specific antibody production in vivo by
about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold,
10-fold, 25-fold, 50-fold, 100-fold, 250-fold, 500-fold, 750-fold, or about
1000-fold or more. In another aspect, one
or more of the subject methods are effective in reducing antigen specific IgG3
and/or IgGM production in vivo by
about 2-fold, 3-fold, 4-fold, 5-fold, 7.5-fold, 10-fold, 25-fold, 50-fold, 100-
fold, 250-fold, 500-fold, 750-fold, or
about 1000-fold or more.
[00531]
In one aspect, one of more of the subject methods are effective in
ameliorating symptoms
associated with rheumatoid arthritis including, but not limited to a reduction
in the swelling of joints, a reduction in
serum anti-collagen levels, and/or a reduction in joint pathology such as bone
resorption, cartilage damage, pannus,
and/or inflammation. In another aspect, the subject methods are effective in
reducing ankle inflammation by at least
about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 50%, 60%, or about 75% to 90%. In
another aspect, the subject
methods are effective in reducing knee inflammation by at least about 2%, 5%,
10%, 15%, 20%, 25%, 30%, 50%,
60%, or about 75% to 90% or more. In still another aspect, the subject methods
are effective in reducing serum anti-
type II collagen levels by at least about 10%, 12%, 15%, 20%, 24%, 25%, 30%,
35%, 50%, 60%, 75%, 80%, 86%,
87%, or about 90% or more. In another aspect, the subject methods are
effective in reducing ankle histopathology
scores by about 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90% or
more. In still another aspect,
the subject methods are effective in reducing knee histopathology scores by
about 5%, 10%, 15%, 20%, 25%, 30%,
40%, 50%, 60%, 75%, 80%, 90% or more.
134

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00532]
In some embodiments, provided herein are methods for treating disorders or
conditions in
which the 6 isoform of PI3K is implicated to a greater extent than other PI3K
isoforms such as PI3K-a and/or -13.
Selective inhibition of PI3K-6 and/or PI3K-7 can provide advantages over using
less selective compounds which
inhibit PI3K-a and/or -13, such as an improved side effects profile or
lessened reduction in the ability to reduce a
bacterial, viral, and/or fungal infection.
[00533]
In other embodiments, provided herein are methods of using a compound, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein, to treat respiratory
diseases including, but not limited to diseases affecting the lobes of lung,
pleural cavity, bronchial tubes, trachea,
upper respiratory tract, or the nerves and muscle for breathing. For example,
methods are provided to treat
obstructive pulmonary disease. Chronic obstructive pulmonary disease (COPD) is
an umbrella term for a group of
respiratory tract diseases that are characterized by airflow obstruction or
limitation. Conditions included in this
umbrella term include, but are not limited to: chronic bronchitis, emphysema,
and bronchiectasis.
[00534]
In another embodiment, the compounds, or a pharmaceutically acceptable form
(e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein are used for the
treatment of asthma. Also, the
compounds or pharmaceutical compositions described herein can be used for the
treatment of endotoxemia and
sepsis. In one embodiment, the compounds or pharmaceutical compositions
described herein are used to for the
treatment of rheumatoid arthritis (RA). In yet another embodiment, the
compounds or pharmaceutical compositions
described herein is used for the treatment of contact or atopic dermatitis.
Contact dermatitis includes irritant
dermatitis, phototoxic dermatitis, allergic dermatitis, photoallergic
dermatitis, contact urticaria, systemic contact-
type dermatitis and the like. Irritant dermatitis can occur when too much of a
substance is used on the skin of when
the skin is sensitive to certain substance. Atopic dermatitis, sometimes
called eczema, is a kind of dermatitis, an
atopic skin disease.
[00535]
In some embodiments, the disclosure provides a method of treating diseases
related to
vasculogenesis or angiogenesis in a subject that comprises administering to
said subject a therapeutically effective
amount of a compound, or a pharmaceutically acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates,
solvates, isomers, prodrugs, and isotopically labeled derivatives) thereof, or
pharmaceutical compositions as
provided herein. In some embodiments, said method is for treating a disease
selected from tumor angiogenesis,
chronic inflammatory disease such as rheumatoid arthritis and chronic
inflammatory demyelinating polyneuropathy,
atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis,
eczema, and scleroderma, diabetes,
diabetic retinopathy, retinopathy of prematurity, age-related macular
degeneration, hemangioma, glioma, melanoma,
Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and
epidermoid cancer.
[00536]
In addition, the compounds described herein can be used for the treatment of
arteriosclerosis,
including atherosclerosis. Arteriosclerosis is a general term describing any
hardening of medium or large arteries.
Atherosclerosis is a hardening of an artery specifically due to an
atheromatous plaque.
135

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00537]
In some embodiments, provided herein is a method of treating a cardiovascular
disease in a
subject that comprises administering to said subject a therapeutically
effective amount of a compound as provided
herein, or a pharmaceutically acceptable form (e.g., pharmaceutically
acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives) thereof. Examples of
cardiovascular conditions include, but are not
limited to, atherosclerosis, restenosis, vascular occlusion and carotid
obstructive disease.
[00538]
In some embodiments, the disclosure relates to a method of treating diabetes
in a subject that
comprises administering to said subject a therapeutically effective amount of
a compound as provided herein, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein.
[00539]
In addition, the compounds, or a pharmaceutically acceptable form (e.g.,
pharmaceutically
acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically
labeled derivatives) thereof, or
pharmaceutical compositions as provided herein, can be used to treat acne. In
certain embodiments, the
inflammatory condition and/or immune disorder is a skin condition. In some
embodiments, the skin condition is
pruritus (itch), psoriasis, eczema, burns or dermatitis. In certain
embodiments, the skin condition is psoriasis. In
certain embodiments, the skin condition is pruritis.
[00540]
In certain embodiments, the inflammatory disorder and/or the immune disorder
is a
gastrointestinal disorder. In some embodiments, the gastrointestinal disorder
is selected from gastrointestinal
disorder (e.g., selected from peptic ulcers, regional enteritis,
diverticulitis, gastrointestinal bleeding, eosinophilic
gastrointestinal disorders (e.g., eosinophilic esophagitis, eosinophilic
gastritis, eosinophilic gastroenteritis,
eosinophilic colitis), gastritis, diarrhea, gastroesophageal reflux disease
(GORD, or its synonym GERD),
inflammatory bowel disease (IBD) (e.g., Crohn's disease, ulcerative colitis,
collagenous colitis, lymphocytic colitis,
ischaemic colitis, diversion colitis, Behcet's syndrome, indeterminate
colitis) and inflammatory bowel syndrome
(IBS)). In certain embodiments, the gastrointestinal disorder is inflammatory
bowel disease (IBD).
[00541]
Further, the compounds, or a pharmaceutically acceptable form (e.g.,
pharmaceutically
acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically
labeled derivatives) thereof, or
pharmaceutical compositions as provided herein, can be used for the treatment
of glomerulonephritis.
Glomerulonephritis is a primary or secondary autoimmune renal disease
characterized by inflammation of the
glomeruli. It can be asymptomatic, or present with hematuria and/or
proteinuria. There are many recognized types,
divided in acute, subacute or chronic glomerulonephritis. Causes are
infectious (bacterial, viral or parasitic
pathogens), autoimmune or paraneoplastic.
[00542]
In some embodiments, provided herein are compounds, or a pharmaceutically
acceptable form
(e.g., pharmaceutically acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, for the treatment
of multiorgan failure. Also provided
herein are compounds, or a pharmaceutically acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates,
solvates, isomers, prodrugs, and isotopically labeled derivatives) thereof, or
pharmaceutical compositions as
provided herein, for the treatment of liver diseases (including diabetes),
gall bladder disease (inluding gallstones),
136

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
pancreatitis or kidney disease (including proliferative glomerulonephritis and
diabetes- induced renal disease) or
pain in a subject.
[00543]
In some embodiments, provided herein are compounds, or a pharmaceutically
acceptable
form (e.g., pharmaceutically acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical compositions as provided herein, for
the prevention of blastocyte
implantation in a subject.
[00544]
In some embodiments, provided herein are compounds, or a pharmaceutically
acceptable form
(e.g., pharmaceutically acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, for the treatment
of disorders involving platelet
aggregation or platelet adhesion, including, but not limited to Idiopathic
thrombocytopenic purpura, Bernard-Soulier
syndrome, Glanzmann's thrombasthenia, Scott's syndrome, von Willebrand
disease, Hermansky-Pudlak Syndrome,
and Gray platelet syndrome.
[00545]
In some embodiments, compounds, or a pharmaceutically acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, are provided for
treating a disease which is skeletal
muscle atrophy, skeletal or muscle hypertrophy. In some embodiments, provided
herein are compounds, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein, for the treatment of
disorders that include, but are not limited to, cancers as discussed herein,
transplantation-related disorders (e.g.,
lowering rejection rates, graft-versus-host disease, etc.), muscular sclerosis
(MS), allergic disorders (e.g., arthritis,
allergic encephalomyelitis) and other immunosuppressive-related disorders,
metabolic disorders (e.g., diabetes),
reducing intimal thickening following vascular injury, and misfolded protein
disorders (e.g., Alzheimer's Disease,
Gaucher's Disease, Parkinson's Disease, Huntington's Disease, cystic fibrosis,
macular degeneration, retinitis
pigmentosa, and prion disorders) (as mTOR inhibition can alleviate the effects
of misfolded protein aggregates).
The disorders also include hamartoma syndromes, such as tuberous sclerosis and
Cowden Disease (also termed
Cowden syndrome and multiple hamartoma syndrome).
[00546]
Additionally, the compounds, or a pharmaceutically acceptable form (e.g.,
pharmaceutically
acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically
labeled derivatives) thereof, or
pharmaceutical compositions as provided herein, can be used for the treatment
of bursitis, lupus, acute disseminated
encephalomyelitis (ADEM), Addison's disease, antiphospholipid antibody
syndrome (APS), amyloidosis (including
systemic and localized amyloidosis; and primary and secondary amyloidosis),
aplastic anemia, autoimmune
hepatitis, coeliac disease, crohn's disease, diabetes mellitus (type 1),
eosinophilic gastroenterides, goodpasture's
syndrome, graves' disease, guillain-barre syndrome (GBS), hashimoto's disease,
inflammatory bowel disease, lupus
erythematosus (including cutaneous lupus erythematosus and systemic lupus
erythematosus), myasthenia gravis,
opsoclonus myoclonus syndrome (OMS), optic neuritis, ord's
thyroiditis,ostheoarthritis, uveoretinitis, pemphigus,
polyarthritis, primary biliary cirrhosis, reiter's syndrome, takayasu's
arteritis, temporal arteritis, warm autoimmune
hemolytic anemia, wegener's granulomatosis, alopecia universalis, chagas'
disease, chronic fatigue syndrome,
137

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis,
neuromyotonia, sarcoidosis, scleroderma,
ulcerative colitis, vitiligo, vulvodynia, appendicitis, arteritis, arthritis,
blepharitis, bronchiolitis, bronchitis, cervicitis,
cholangitis, cholecystitis, chorioamnionitis, colitis, conjunctivitis,
cystitis, dacryoadenitis, dermatomyositis,
endocarditis, endometritis, enteritis, enterocolitis, epicondylitis,
epididymitis, fasciitis, fibrositis, gastritis,
gastroenteritis, gingivitis, hepatitis, hidradenitis, ileitis, iritis,
laryngitis, mastitis, meningitis, myelitis, myocarditis,
myositis, nephritis, omphalitis, oophoritis, orchitis, osteitis, otitis,
pancreatitis, parotitis, pericarditis, peritonitis,
pharyngitis, pleuritis, phlebitis, pneumonitis, proctitis, prostatitis,
pyelonephritis, rhinitis, salpingitis, sinusitis,
stomatitis, synovitis, tendonitis, tonsillitis, uveitis (e.g., ocular
uveitis), vaginitis, vasculitis, or vulvitis.
[00547]
In another aspect, provided herein are methods of disrupting the function of a
leukocyte or
disrupting a function of an osteoclast. The method includes contacting the
leukocyte or the osteoclast with a
function disrupting amount of a compound as provided herein.
[00548]
In another aspect, methods are provided for treating ophthalmic disease by
administering one
or more of the subject compounds or pharmaceutical compositions to the eye of
a subject.
[00549]
In certain embodiments, provided herein are methods of treating, preventing
and/or managing a
disease or a disorder using a compound, or a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable
salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical
compositions as provided herein, wherein the disease or disorder is: Crohn's
disease; cutaneous lupus; multiple
sclerosis; rheumatoid arthritis; and systemic lupus erythematosus.
[00550]
In other embodiments, provided herein are methods of treating, preventing
and/or managing a
disease or a disorder using a compound, or a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable
salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical
compositions as provided herein, wherein the disease or disorder is:
ankylosing spondylitis; chronic obstructive
pulmonary disease; myasthenia gravis; ocular uveitis, psoriasis; and psoriatic
arthritis.
[00551]
In other embodiments, provided herein are methods of treating, preventing
and/or managing a
disease or a disorder using a compound, or a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable
salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical
compositions as provided herein, wherein the disease or disorder is: adult-
onset Still's disease; inflammatory
alopecia; amyloidosis; antiphospholipid syndrome; autoimmune hepatitis;
autoimmune skin disease, Behcet's
disease; chronic inflammatory demyelinating polyneuropathy; eosinophilic
gastroenteritis; inflammatory
myopathies, pemphigus, polymyalgia rheumatica; relapsing polychondritis;
Sjorgen's syndrome; temporal arthritis;
ulcerative colitis; vasculis; vitiligo, and Wegner's granulomatosis.
[00552]
In other embodiments, provided herein are methods of treating, preventing
and/or managing a
disease or a disorder using a compound, or a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable
salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical
compositions as provided herein, wherein the disease or disorder is: gout
flare; sacoidosis; and systemic sclerosis.
[00553]
In certain embodiments, provided herein are methods of treating, preventing
and/or managing a
disease or a disorder using a compound, or a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable
138

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled
derivatives) thereof, or pharmaceutical
compositions as provided herein, wherein the disease or disorder is: asthma;
arthritis (e.g., rheumatoid arthritis and
psoriatic arthritis); psoriasis; scleroderma; myositis (e.g.,
dermatomyositis); lupus (e.g., cutaneous lupus
eiythematosus ("CLE") or systemic lupus erythematosus ("SLE")); or Sjogren's
syndrome.
[00554] Efficacy of a compound provided herein in treating, preventing
and/or managing the disease or
disorder can be tested using various animal models known in the art. For
example: efficacy in treating, preventing
and/or managing asthma can be assessed using ova induced asthma model
described, for example, in Lee et al.
(2006) J Allergy Clin Immunol 118(2):403-9; efficacy in treating, preventing
and/or managing arthritis (e.g.,
rheumatoid or psoriatic arthritis) can be assessed using autoimmune animal
models described, for example, in
Williams et al. (2010) Chem Biol, 17(2):123-34, WO 2009/088986, W02009/088880,
and WO 2011/008302;
efficacy in treating, preventing and/or managing psoriasis can be assessed
using transgenic or knockout mouse
model with targeted mutations in epidermis, vasculature or immune cells, mouse
model resulting from spontaneous
mutations, and immunodeficient mouse model with xenotransplantation of human
skin or immune cells, all of which
are described, for example, in Boehncke et al. (2007) Clinics in Dermatology,
25: 596-605; efficacy in treating,
preventing and/or managing fibrosis or fibrotic condition can be assessed
using the unilateral ureteral obstruction
model of renal fibrosis (see Chevalier et al., Kidney International (2009)
75:1145-1152), the bleomycin induced
model of pulmonary fibrosis (see Moore and Hogaboam, Am. J. Physiol. Lung.
Cell. Mol. Physiol. (2008) 294:L152-
L160), a variety of liver/biliary fibrosis models (see Chuang et al., Clin
Liver Dis (2008) 12:333-347 and Omenetti,
A. et al. (2007) Laboratory Investigation 87:499-514 (biliary duct-ligated
model)), or a number of myelofibrosis
mouse models (see Varicchio, L. et al. (2009) Expert Rev. Hematol. 2(3):315-
334); efficacy in treating, preventing
and/or managing scleroderma can be assessed using mouse model induced by
repeated local injections of bleomycin
("BLM") described, for example, in Yamamoto et al. (1999) J Invest Dermatol
112: 456-462; efficacy in treating,
preventing and/or managing dermatomyositis can be assessed using myositis
mouse model induced by immunization
with rabbit myosin described, for example, in Phyanagi et al. (2009) Arthritis
& Rheumatism, 60(10): 3118-3127;
efficacy in treating, preventing and/or managing lupus (e.g., CLE or SLE) can
be assessed using various animal
models described, for example, in Ghoreishi et al. (2009) Lupus, 19: 1029-
1035, Ohl et al. (2011) Journal of
Biomedicine and Biotechnology, Article ID 432595 (14 pages), Xia et al. (2011)
Rheumatology, 50:2187-2196, Pau
et al. (2012) PLoS ONE, 7(5):e36761 (15 pages), Mustafa et al. (2011)
Toxicology, 290:156-168, Ichikawa et al.
(2012) Arthritis and Rheumatism, 62(2): 493-503, Ouyang et al. (2012)J Mol
Med, DOT 10.1007/s00109-012-0866-
3 (10 pages), Rankin et al. (2012) Journal of Immunology, 188:1656-1667; and
efficacy in treating, preventing
and/or managing Sjogren's syndrome can be assessed using various mouse models
described, for example, in
Chiorini et al. (2009) Journal of Autoimmunity, 33: 190-196.
[00555] In one embodiment, provided herein is a method of treating,
preventing and/or managing
asthma. As used herein, "asthma" encompasses airway constriction regardless of
the cause. Common triggers of
asthma include, but are not limited to, exposure to an environmental
stimulants (e.g., allergens), cold air, warm air,
perfume, moist air, exercise or exertion, and emotional stress. Also provided
herein is a method of treating,
139

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
preventing and/or managing one or more symptoms associated with asthma.
Examples of the symptoms include, but
are not limited to, severe coughing, airway constriction and mucus production.
[00556] In one embodiment, provided herein is a method of treating,
preventing and/or managing
arthritis. As used herein, "arthritis" encompasses all types and
manifestations of arthritis. Examples include, but are
not limited to, crystalline arthritis, osteoarthritis, psoriatic arthritis,
gouty arthritis, reactive arthritis, rheumatoid
arthritis and Reiter's arthritis. In one embodiment, the disease or disorder
is rheumatoid arthritis. In another
embodiment, the disease or disorder is psoriatic arthritis. Also provided
herein is a method of treating, preventing
and/or managing one or more symptoms associated with arthritis. Examples of
the symptoms include, but are not
limited to, joint pain, which progresses into joint deformation, or damages in
body organs such as in blood vessels,
heart, lungs, skin, and muscles.
[00557] In one embodiment, provided herein is a method of treating,
preventing and/or managing
psoriasis. As used herein, "psoriasis" encompasses all types and
manifestations of psoriasis. Examples include, but
are not limited to, plaque psoriasis (e.g., chronic plaque psoriasis, moderate
plaque psoriasis and severe plaque
psoriasis), guttate psoriasis, inverse psoriasis, pustular psoriasis,
pemphigus vulgaris, erythrodermic psoriasis,
psoriasis associated with inflammatory bowel disease (IBD), and psoriasis
associated with rheumatoid arthritis
(RA). Also provided herein is a method of treating, preventing and/or managing
one or more symptoms associated
with psoriasis. Examples of the symptoms include, but are not limited to: red
patches of skin covered with silvery
scales; small scaling spots; dry, cracked skin that may bleed; itching;
burning; soreness; thickened, pitted or ridged
nails; and swollen and stiff joints.
[00558] In one embodiment, provided herein is a method of treating,
preventing and/or managing
fibrosis and fibrotic condition. As used herein, "fibrosis" or "fibrotic
condition encompasses all types and
manifestations of fibrosis or fibrotic condition. Examples include, but are
not limited to, formation or deposition of
tissue fibrosis; reducing the size, cellularity (e.g., fibroblast or immune
cell numbers), composition; or cellular
content, of a fibrotic lesion; reducing the collagen or hydroxyproline
content, of a fibrotic lesion; reducing
expression or activity of a fibrogenic protein; reducing fibrosis associated
with an inflammatory response;
decreasing weight loss associated with fibrosis; or increasing survival.
[00559] In certain embodiments, the fibrotic condition is primary
fibrosis. In one embodiment, the
fibrotic condition is idiopathic. In other embodiments, the fibrotic condition
is associated with (e.g., is secondary to)
a disease (e.g., an infectious disease, an inflammatory disease, an autoimmune
disease, a malignant or cancerous
disease, and/or a connective disease); a toxin; an insult (e.g., an
environmental hazard (e.g., asbestos, coal dust,
polycyclic aromatic hydrocarbons), cigarette smoking, a wound); a medical
treatment (e.g., surgical incision,
chemotherapy or radiation), or a combination thereof.
[00560] In some embodiments, the fibrotic condition is associated with an
autoimmune disease selected
from scleroderma or lupus, e.g., systemic lupus erythematosus. In some
embodiments, the fibrotic condition is
systemic. In some embodiments, the fibrotic condition is systemic sclerosis
(e.g., limited systemic sclerosis, diffuse
systemic sclerosis, or systemic sclerosis sine scleroderma), nepluogenic
systemic fibrosis, cystic fibrosis, chronic
graft vs. host disease, or atherosclerosis.
140

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00561] In certain embodiments, the fibrotic condition is a fibrotic
condition of the lung, a fibrotic
condition of the liver, a fibrotic condition of the heart or vasculature, a
fibrotic condition of the kidney, a fibrotic
condition of the skin, a fibrotic condition of the gastrointestinal tract, a
fibrotic condition of the bone marrow or a
hematopoietic tissue, a fibrotic condition of the nervous system, a fibrotic
condition of the eye, or a combination
thereof.
[00562] In other embodiment, the fibrotic condition affects a tissue
chosen from one or more of muscle,
tendon, cartilage, skin (e.g., skin epidermis or endodermis), cardiac tissue,
vascular tissue (e.g., artery, vein),
pancreatic tissue, lung tissue, liver tissue, kidney tissue, uterine tissue,
ovarian tissue, neural tissue, testicular tissue,
peritoneal tissue, colon, small intestine, biliary tract, gut, bone marrow,
hematopoietic tissue, or eye (e.g., retinal)
tissue.
[00563] In some embodiments, the fibrotic condition is a fibrotic
condition of the eye. In some
embodiments, the fibrotic condition is glaucoma, macular degeneration (e.g.,
age-related macular degeneration),
macular edema (e.g., diabetic macular edema), retinopathy (e.g., diabetic
retinopathy), or dry eye disease.
[00564] In certain embodiments, the fibrotic condition is a fibrotic
condition of the lung. In certain
embodiments, the fibrotic condition of the lung is chosen from one or more of:
pulmonary fibrosis, idiopathic
pulmonary fibrosis (IPF), usual interstitial pneumonitis (UIP), interstitial
lung disease, cryptogenic fibrosing
alveolitis (CFA), bronchiectasis, and scleroderma lung disease. In one
embodiment, the fibrosis of the lung is
secondary to a disease, a toxin, an insult, a medical treatment, or a
combination thereof. For example, the fibrosis of
the lung can be associated with (e.g., secondary to) one or more of: a disease
process such as asbestosis and
silicosis; an occupational hazard; an environmental pollutant; cigarette
smoking; an autoimmune connective tissue
disorders (e.g., rheumatoid arthritis, scleroderma and systemic lupus
erythematosus (SLE)); a connective tissue
disorder such as sarcoidosis; an infectious disease, e.g., infection,
particularly chronic infection; a medical treatment,
including but not limited to, radiation therapy, and drug therapy, e.g.,
chemotherapy (e.g., treatment with as
bleomycin, methotrexate, amiodarone, busulfan, and/or nitrofurantoin). In one
embodiment, the fibrotic condition
of the lung treated with the methods of the invention is associated with
(e.g., secondary to) a cancer treatment, e.g.,
treatment of a cancer (e.g., squamous cell carcinoma, testicular cancer,
Hodgkin's disease with bleomycin). In one
embodiment, the fibrotic condition of the lung is associated with an
autoimmune connective tissue disorder (e.g.,
scleroderma or lupus, e.g., SLE).
[00565] In certain embodiments, the fibrotic condition is a fibrotic
condition of the liver. In certain
embodiments, the fibrotic condition of the liver is chosen from one or more
of: fatty liver disease, steatosis (e.g.,
nonalcoholic steatohepatitis (NASH), cholestatic liver disease (e.g., primary
biliary cirrhosis (PBC)), cirrhosis,
alcohol induced liver fibrosis, biliary duct injury, biliary fibrosis, or
cholangiopathies. In other embodiments,
hepatic or liver fibrosis includes, but is not limited to, hepatic fibrosis
associated with alcoholism, viral infection,
e.g., hepatitis (e.g., hepatitis C, B or D), autoimmune hepatitis, non-
alcoholic fatty liver disease (NAFLD),
progressive massive fibrosis, exposure to toxins or irritants (e.g., alcohol,
pharmaceutical drugs and environmental
toxins).
141

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00566] In certain embodiments, the fibrotic condition is a fibrotic
condition of the heart. In certain
embodiments, the fibrotic condition of the heart is myocardial fibrosis (e.g.,
myocardial fibrosis associated with
radiation myocarditis, a surgical procedure complication (e.g., myocardial
post-operative fibrosis), infectious
diseases (e.g., Chagas disease, bacterial, trichinosis or fungal
myocarditis)); granulomatous, metabolic storage
disorders (e.g., cardiomyopathy, hemochromatosis); developmental disorders
(e.g, endocardial fibroelastosis);
arteriosclerotic, or exposure to toxins or irritants (e.g., drug induced
cardiomyopathy, drug induced cardiotoxicity,
alcoholic cardiomyopathy, cobalt poisoning or exposure). In certain
embodiments, the myocardial fibrosis is
associated with an inflammatory disorder of cardiac tissue (e.g., myocardial
sarcoidosis). In some embodiments, the
fibrotic condition is a fibrotic condition associated with a myocardial
infarction. In some embodiments, the fibrotic
condition is a fibrotic condition associated with congestive heart failure.
[00567] In certain embodiments, the fibrotic condition is a fibrotic
condition of the kidney. In certain
embodiments, the fibrotic condition of the kidney is chosen from one or more
of: renal fibrosis (e.g., chronic kidney
fibrosis), nepluppathies associated with injury/fibrosis (e.g., chronic
nepluppathies associated with diabetes (e.g.,
diabetic nephropathy)), lupus, scleroderma of the kidney, glomerular
nephritis, focal segmental glomerular sclerosis,
IgA nephropathyrenal fibrosis associated with human chronic kidney disease
(CKD), chronic progressive
nephropathy (CPN), tubulointerstitial fibrosis, ureteral obstruction, chronic
uremia, chronic interstitial nephritis,
radiation nephropathy, glomerulosclerosis, progressive glomerulonephrosis
(PGN), endothelial/thrombotic
microangiopathy injury, HIV-associated nephropathy, or fibrosis associated
with exposure to a toxin, an irritant, or a
chemotherapeutic agent. In one embodiment, the fibrotic condition of the
kidney is scleroderma of the kidney. In
some embodiments, the fibrotic condition of the kidney is transplant
nephropathy, diabetic nephropathy, lupus
nephritis, or focal segmental glomerulosclerosis (FSGS).
[00568] In certain embodiments, the fibrotic condition is a fibrotic
condition of the skin. In certain
embodiments, the fibrotic condition of the skin is chosen from one or more of:
skin fibrosis (e.g., hypertrophic
scarring, keloid), scleroderma, nepluogenic systemic fibrosis (e.g., resulting
after exposure to gadolinium (which is
frequently used as a contrast substance for MRIs) in patients with severe
kidney failure), and keloid.
[00569] In certain embodiments, the fibrotic condition is a fibrotic
condition of the gastrointestinal
tract. In certain embodiments, the fibrotic condition is chosen from one or
more of: fibrosis associated with
scleroderma; radiation induced gut fibrosis; fibrosis associated with a
foregut inflammatory disorder such as
Barrett's esophagus and chronic gastritis, and/or fibrosis associated with a
hindgut inflammatory disorder, such as
inflammatory bowel disease (IBD), ulcerative colitis and Crohn's disease. In
some embodiments, the fibrotic
condition of the gastrointestinal tract is fibrosis associated with
scleroderma.
[00570] In certain embodiments, the fibrotic condition is a fibrotic
condition of the bone marrow or a
hematopoietic tissue. In certain embodiments, the fibrotic condition of the
bone marrow is an intrinsic feature of a
chronic myeloproliferative neoplasm of the bone marrow, such as primary
myelofibrosis (also referred to herein as
agnogenic myeloid metaplasia or chronic idiopathic myelofibrosis). In other
embodiments, the bone marrow
fibrosis is associated with (e.g., is secondary to) a malignant condition or a
condition caused by a clonal proliferative
disease. In other embodiments, the bone marrow fibrosis is associated with a
hematologic disorder (e.g., a
142

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
hematologic disorder chosen from one or more of polycythemia vera, essential
thrombocythemia, myelodysplasia,
hairy cell leukemia, lymphoma (e.g., Hodgkin or non-Hodgkin lymphoma),
multiple myeloma or chronic
myelogeneous leukemia (CML)). In yet other embodiments, the bone marrow
fibrosis is associated with (e.g.,
secondary to) a non-hematologic disorder (e.g., a non-hematologic disorder
chosen from solid tumor metastasis to
bone marrow, an autoimmune disorder (e.g., systemic lupus erythematosus,
scleroderma, mixed connective tissue
disorder, or polymyositis), an infection (e.g., tuberculosis), or secondary
hyperparathyroidism associated with
vitamin D deficiency. In some embodiments, the fibrotic condition is
idiopathic or drug-induced myelofibrosis. In
some embodiments, the fibrotic condition of the bone marrow or hematopoietic
tissue is associated with systemic
lupus erythematosus or scleroderma.
[00571] In one embodiment, provided herein is a method of treating,
preventing and/or managing
scleroderma. Scleroderma is a group of diseases that involve hardening and
tightening of the skin and/or other
connective tissues. Scleroderma may be localized (e.g., affecting only the
skin) or systemic (e.g., affecting other
systems such as, e.g., blood vessels and/or internal organs). Common symptoms
of scleroderma include Raynaud's
phenomenon, gastroesophageal reflux disease, and skin changes (e.g., swollen
fingers and hands, or thickened
patches of skin). In some embodiments, the scleroderma is localized, e.g.,
morphea or linear scleroderma. In some
embodiments, the condition is a systemic sclerosis, e.g., limited systemic
sclerosis, diffuse systemic sclerosis, or
systemic sclerosis sine scleroderma.
[00572] Localized scleroderma (localized cutaneous fibrosis) includes
morphea and linear scleroderma.
Morphea is typically characterized by oval-shaped thickened patches of skin
that are white in the middle, with a
purple border. Linear scleroderma is more common in children. Symptoms of
linear scleroderma may appear
mostly on one side of the body. In linear scleroderma, bands or streaks of
hardened skin may develop on one or
both arms or legs or on the forehead. En coup de sabre (frontal linear
scleroderma or morphea en coup de sabre) is a
type of localized scleroderma typically characterized by linear lesions of the
scalp or face.
[00573] Systemic scleroderma (systemic sclerosis) includes, e.g., limited
systemic sclerosis (also
known as limited cutaneous systemic sclerosis, or CREST syndrome), diffuse
systemic sclerosis (also known as
diffuse cutaneous systemic sclerosis), and systemic sclerosis sine
scleroderma. CREST stands for the following
complications that may accompany limited scleroderma: calcinosis (e.g., of the
digits), Raynaud's phenomenon,
esophageal dysfunction, sclerodactyly, and telangiectasias. Typically, limited
scleroderma involves cutaneous
manifestations that mainly affect the hands, arms, and face. Limited and
diffuse subtypes are distinguished based on
the extent of skin involvement, with sparing of the proximal limbs and trunk
in limited disease. See, e.g., Denton,
C.P. et al. (2006), Nature Clinical Practice Rheumatology, 2(3):134-143. The
limited subtype also typically
involves a long previous history of Raynaud's phenomenon, whereas in the
diffuse subtype, onset of Raynaud's
phenomenon can be simultaneous with other manifestations or might occur later.
Both limited and diffuse subtypes
may involve internal organs. Typical visceral manifestations of limited
systemic sclerosis include isolated
pulmonary hypertension, severe bowel involvement, and pulmonary fibrosis.
Typical visceral manifestations of
diffuse systemic sclerosis include renal crisis, lung fibrosis, and cardiac
disease. Diffuse systemic sclerosis typically
progresses rapidly and affects a large area of the skin and one or more
internal organs (e.g., kidneys, esophagus,
143

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
heart, or lungs). Systemic sclerosis sine scleroderma is a rare disorder in
which patients develop vascular and
fibrotic damage to internal organs in the absence of cutaneous sclerosis.
[00574] In one embodiment, provided herein is a method of treating,
preventing and/or managing
inflammatory myopathies. As used herein, "inflammatory myopathies" encompass
all types and manifestations of
inflammatory myopathies. Examples include, but are not limited to, muscle
weakness (e.g., proximal muscle
weakness), skin rash, fatigue after walking or standing, tripping or falling,
dysphagia, dysphonia, difficulty
breathing, muscle pain, tender muscles, weight loss, low-grade fever, inflamed
lungs, light sensitivity, calcium
deposits (calcinosis) under the skin or in the muscle, as well as biological
concomitants of inflammatory myopathies
as disclosed herein or as known in the art. Biological concomitants of
inflammatory myopathies (e.g.,
dermatomyositis) include, e.g., altered (e.g., increased) levels of cytokines
(e.g., Type I interferons (e.g., IFN-a
and/or IFN-I3), interleukins (e.g., IL-6, IL-10, IL-15, IL-17 and IL-18), and
TNF-a), TGF-I3, B-cell activating factor
(BAFF), overexpression of IFN inducible genes (e.g., Type I IFN inducible
genes). Other biological concomitants
of inflammatory myopathies can include, e.g., an increased erythrocyte
sedimentation rate (ESR) and/or elevated
level of creatine kinase. Further biological concomitants of inflammatory
myopathies can include autoantibodies,
e.g., anti-synthetase autoantibodies (e.g., anti-Job antibodies), anti-signal
recognition particle antibodies (anti-SRP),
anti-Mi-2 antibodies, anti-p155 antibodies, anti-PM/Sci antibodies, and anti-
RNP antibodies.
[00575] The inflammatory myopathy can be an acute inflammatory myopathy
or a chronic
inflammatory myopathy. In some embodiments, the inflammatory myopathy is a
chronic inflammatory myopathy
(e.g., dermatomyositis, polymyositis, or inclusion body myositis). In some
embodiments, the inflammatory
myopathy is caused by an allergic reaction, another disease (e.g., cancer or a
connective tissue disease), exposure to
a toxic substance, a medicine, or an infectious agent (e.g., a virus). In some
embodiments, the inflammatory
myopathy is associated with lupus, rheumatoid arthritis, or systemic
sclerosis. In some embodiments, the
inflammatory myopathy is idiopathic. In some embodiments, the inflammatory
myopathy is selected from
polymyositis, dermatomyositis, inclusion body myositis, and immune-mediated
necrotizing myopathy. In some
embodiments, the inflammatory myopathy is dermatomyositis.
[00576] In another embodiment, provided herein is a method of treating,
preventing and/or managing a
skin condition (e.g., a dermatitis). In some embodiments, the methods provided
herein can reduce symptoms
associated with a skin condition (e.g., itchiness and/or inflammation). In
some such embodiments, the compound
provided herein is administered topically (e.g., as a topical cream, eyedrop,
nose drop or nasal spray). In some such
embodiments, the compound is a PI3K delta inhibitor (e.g., a PI3K inhibitor
that demonstrates greater inhibition of
PI3K delta than of other PI3K isoforms). In some embodiments, the PI3K delta
inhibitor prevents mast cell
degranulation.
[00577] As used herein, "skin condition" includes any inflammatory
condition of the skin (e.g., eczema
or dermatitis, e.g., contact dermatitis, atopic dermatitis, dermatitis
herpetiformis, seborrheic dermatitis, nummular
dermatitis, stasis dermatitis, perioral dermatitis), as well as accompanying
symptoms (e.g., skin rash, itchiness
(pruritis), swelling (edema), hay fever, anaphalaxis). Frequently, such skin
conditions are caused by an allergen. As
used herein, a "skin condition" also includes, e.g., skin rashes (e.g.,
allergic rashes, e.g., rashes resulting from
144

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
exposure to allergens such as poison ivy, poison oak, or poison sumac, or
rashes caused by other diseases or
conditions), insect bites, minor burns, sunburn, minor cuts, and scrapes. In
some embodiments, the symptom
associated with inflammatory myopathy, or the skin condition or symptom
associated with the skin condition, is a
skin rash or itchiness (pruritis) caused by a skin rash.
[00578] The skin condition (e.g., the skin rash) may be spontaneous, or
it may be induced, e.g., by
exposure to an allergen (e.g., poison ivy, poison oak, or poison sumac),
drugs, food, insect bite, inhalants, emotional
stress, exposure to heat, exposure to cold, or exercise. In some embodiments,
the skin condition is a skin rash (e.g.,
a pruritic rash, e.g., utricaria). In some embodiments, the skin condition is
an insect bite. In some embodiments, the
skin condition is associated with another disease (e.g., an inflammatory
myopathy, e.g., dermatomyositis).
[00579] In some embodiments, the subject (e.g., the subject in need of
treatment for an inflammatory
myopathy and/or a skin condition) exhibits an elevated level or elevated
activity of IFN-a, TNF-a, IL-6, IL-8, IL-1,
or a combination thereof. In certain embodiments, the subject exhibits an
elevated level of IFN-a. In some
embodiments, treating (e.g., decreasing or inhibiting) the inflammatory
myopathy, or the skin condition, comprises
inhibiting (e.g., decreasing a level of, or decreasing a biological activity
of) one or more of IFN-a, TNF-a, IL-6, IL-
8, or IL-1 in the subject or in a sample derived from the subject. In some
embodiments, the method decreases a
level of IFN-a, TNF-a, IL-6, IL-8, or IL-1 in the subject or in a sample
derived from the subject. In some
embodiments, the method decreases a level of IFN-a in the subject or in a
sample derived from the subject. In some
embodiments, the level of IFN-a, TNF-a, IL-6, IL-8, or IL-1 is the level
assessed in a sample of whole blood or
PBMCs. In some embodiments, the level of IFN-a, TNF-a, IL-6, IL-8, or IL-1 is
the level assessed in a sample
obtained by a skin biopsy or a muscle biopsy. In some embodiments, the sample
is obtained by a skin biopsy.
[00580] In one embodiment, provided herein is a method of treating,
preventing and/or managing
myositis. As used herein, "myositis" encompasses all types and manifestations
of myositis. Examples include, but
are not limited to, myositis ossificans, fibromyositis, idiopathic
inflammatory myopathies, dermatomyositis, juvenile
dermatomyositis, polymyositis, inclusion body myositis and pyomyositis. In one
embodiment, the disease or
disorder is dermatomyositis. Also provided herein is a method of treating,
preventing and/or managing one or more
symptoms associated with myositis. Examples of the symptoms include, but are
not limited to: muscle weakness;
trouble lifting arms; trouble swallowing or breathing; muscle pain; muscle
tenderness; fatigue; fever; lung problems;
gastrointestinal ulcers; intestinal perforations; calcinosis under the skin;
soreness; arthritis; weight loss; and rashes.
[00581] In one embodiment, provided herein is a method of treating,
preventing and/or managing
lupus. As used herein, "lupus" refers to all types and manifestations of
lupus. Examples include, but are not limited
to, systemic lupus erythematosus; lupus nephritis; cutaneous manifestations
(e.g., manifestations seen in cutaneous
lupus erythematosus, e.g., a skin lesion or rash); CNS lupus; cardiovascular,
pulmonary, hepatic, hematological,
gastrointestinal and musculoskeletal manifestations; neonatal lupus
erythematosus; childhood systemic lupus
erythematosus; drug-induced lupus erythematosus; anti-phospholipid syndrome;
and complement deficiency
syndromes resulting in lupus manifestations. In one embodiment, the lupus is
systemic lupus erythematosus (SLE),
cutaneous lupus erythematosus (CLE), drug-induced lupus, or neonatal lupus. In
another embodiment, the lupus is a
CLE, e.g., acute cutaneous lupus erythematosus (ACLE), subacute cutaneous
lupus erythematosus (SCLE),
145

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
intermittent cutaneous lupus erythematosus (also known as lupus erythematosus
tumidus (LET)), or chronic
cutaneous lupus. In some embodiments, the intermittent CLE is chronic discloid
lupus erythematosus (CDLE) or
lupus erythematosus profundus (LEP) (also known as lupus erythematosus
panniculitis). Types, symptoms, and
pathogenesis of CLE are described, for example, in Wenzel et al. (2010),
Lupus, 19, 1020-1028.
[00582]
In one embodiment, provided herein is a method of treating, preventing and/or
managing
Sjogren's syndrome. As used herein, "Sjogren's syndrome" refers to all types
and manifestations of Sjogren's
syndrome. Examples include, but are not limited to, primary and secondary
Sjogren's syndrome. Also provided
herein is a method of treating, preventing and/or managing one or more
symptoms associated with Sjogren's
syndrome. Examples of the symptoms include, but are not limited to: dry eyes;
dry mouth; joint pain; swelling;
stiffness; swollen salivary glands; skin rashes; dry skin; vaginal dryness;
persistent dry cough; and prolonged
fatigue.
[00583]
In some embodiments, a symptom associated with the disease or disorder
provided herein is
reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least
50%, at least 60%, at least 70%, at least
80%, at least 90%, or at least 95% relative to a control level. The control
level includes any appropriate control as
known in the art. For example, the control level can be the pre-treatment
level in the sample or subject treated, or it
can be the level in a control population (e.g., the level in subjects who do
not have the disease or disorder or the
level in samples derived from subjects who do not have the disease or
disorder). In some embodiments, the decrease
is statistically significant, for example, as assessed using an appropriate
parametric or non-parametric statistical
comparison.
Combination Therapy
[00584]
In some embodiments, provided herein are methods for combination therapies in
which an
agent known to modulate other pathways, or other components of the same
pathway, or even overlapping sets of
target enzymes are used in combination with a compound as provided herein, or
a pharmaceutically acceptable form
(e.g., pharmaceutically acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives)
thereof. In one aspect, such therapy includes, but is not limited to, the
combination of the subject compound with
chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to
provide a synergistic or additive
therapeutic effect.
[00585]
In one aspect, a compound as provided herein, or a pharmaceutically acceptable
form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, orpharmaceutical compositions as provided herein, can present
synergistic or additive efficacy when
administered in combination with agents that inhibit IgE production or
activity. Such combination can reduce the
undesired effect of high level of IgE associated with the use of one or more
PI3K-6 inhibitors, if such effect occurs.
This can be particularly useful in treatment of autoimmune and inflammatory
disorders (AIID) such as rheumatoid
arthritis. Additionally, the administration of PI3K-6 or PI3K-6/7 inhibitors
as provided herein in combination with
inhibitors of mTOR can also exhibit synergy through enhanced inhibition of the
PI3K pathway.
146

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00586] In a separate but related aspect, provided herein is a
combination treatment of a disease
associated with PI3K-43 comprising administering to a PI3K-43 inhibitor and an
agent that inhibits IgE production or
activity. Other exemplary PI3K-43 inhibitors are applicable for this
combination and they are described, e.g., US
Patent No. 6,800,620. Such combination treatment is particularly useful for
treating autoimmune and inflammatory
diseases (AIID) including, but not limited to rheumatoid arthritis.
[00587] Agents that inhibit IgE production are known in the art and
they include, but are not limited
to, one or more of TEI-9874, 2-(4-(6-cyclohexyloxy-2-
naphtyloxy)phenylacetamide)benzoic acid, rapamycin,
rapamycin analogs (i.e., rapalogs), TORC1 inhibitors, TORC2 inhibitors, and
any other compounds that inhibit
mTORC1 and mTORC2. Agents that inhibit IgE activity include, for example, anti-
IgE antibodies such as for
example Omalizumab and TNX-901.
[00588] For treatment of autoimmune diseases, a compound as provided
herein, or a pharmaceutically
acceptable form (e.g., pharmaceutically acceptable salts, hydrates, solvates,
isomers, prodrugs, and isotopically
labeled derivatives) thereof, or pharmaceutical compositions as provided
herein, can be used in combination with
commonly prescribed drugs including, but not limited to Enbrel , Remicade ,
Humira , Avonex , and Rebif . For
treatment of respiratory diseaseses, the subject compounds or pharmaceutical
compositions can be administered in
combination with commonly prescribed drugs including, but not limited to
Xolair , Advair , Singulair , and
Spiriva .
[00589] The compounds as provided herein, or a pharmaceutically
acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, can be formulated
or administered in conjunction with
other agents that act to relieve the symptoms of inflammatory conditions such
as encephalomyelitis, asthma, and the
other diseases described herein. These agents include non-steroidal anti-
inflammatory drugs (NSAIDs), e.g.,
acetylsalicylic acid; ibuprofen; naproxen; indomethacin; nabumetone; tolmetin;
etc. Corticosteroids are used to
reduce inflammation and suppress activity of the immune system. An exemplary
drug of this type is Prednisone.
Chloroquine (Aralen) or hydroxychloroquine (Plaquenil) can also be used in
some individuals with lupus. They can
be prescribed for skin and joint symptoms of lupus. Azathioprine (Imuran) and
cyclophosphamide (Cytoxan)
suppress inflammation and tend to suppress the immune system. Other agents,
e.g., methotrexate and cyclosporin
are used to control the symptoms of lupus. Anticoagulants are employed to
prevent blood from clotting rapidly.
They range from aspirin at very low dose which prevents platelets from
sticking, to heparin/coumadin. Other
compounds used in the treatment of lupus include belimumab (Benlysta0).
[00590] In another aspect, provided herein is a pharmaceutical
composition for inhibiting abnormal
cell growth in a subject which comprises an amount of a compound as provided
herein, or a pharmaceutically
acceptable form (e.g., pharmaceutically acceptable salts, hydrates, solvates,
isomers, prodrugs, and isotopically
labeled derivatives) thereof, in combination with an amount of an anti-cancer
agent (e.g., a chemotherapeutic agent).
Many chemotherapeutics are presently known in the art and can be used in
combination with the compounds as
provided herein.
147

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00591]
In some embodiments, the chemotherapeutic is selected from mitotic inhibitors,
alkylating
agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors,
cell cycle inhibitors, enzymes,
topoisomerase inhibitors, biological response modifiers, anti-hormones,
angiogenesis inhibitors, and anti-androgens.
Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-
peptide small molecules such as
Gleevec0 (Imatinib Mesylate), Velcade0 (bortezomib), Casodex (bicalutamide),
Iressa0, and Adriamycin as well
as a host of chemotherapeutic agents. Non-limiting examples of
chemotherapeutic agents include alkylating agents
such as thiotepa and cyclosphosphamide (CYTOXANTm); alkyl sulfonates such as
busulfan, improsulfan and
piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
ethylenimines and
methylamelamines including altretamine,
triethylenemelamine, trietylenephosphoramide,
triethylenethiophosphaoramide and trimethylolomelamine; BTK inhibitors such as
ibrutinib (PCI-32765) and AVL-
292; HDAC inhibitors usch as vorinostat, romidepsin, panobinostat, valproic
acid, belinostat, mocetinostat,
abrexinostat, entinostat, SB939, resminostat, givinostat, CUDC-101, AR-42, CHR-
2845, CHR-3996, 4SC-202,
CG200745, ACY-1215 and kevetrin; JAK/STAT inhibitors such as lestaurtinib,
tofacitinib, ruxolitinib, pacritinib,
CYT387, baricitinib, fostamatinib, GLPG0636, TG101348, INCB16562 and AZD1480;
nitrogen mustards such as
bedamustine, chlorambucil, chlornaphazine, cholophosphamide, estramustine,
ifosfamide, mechlorethamine,
mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine,
prednimustine, trofosfamide, uracil
mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine,
lomustine, nimustine, ranimustine; antibiotics
such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins,
cactinomycin, calicheamicin, carabicin,
carminomycin, carzinophilin, CasodexTm , chromomycins, dactinomycin,
daunorubicin, detorubicin, 6-diazo-5-oxo-
L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin,
mitomycins, mycophenolic acid,
nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin,
rodorubicin, streptonigrin,
streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites
such as methotrexate and 5-fluorouracil
(5-FU); folic acid analogues such as denopterin, methotrexate, pralatrexate,
pteropterin, trimetrexate; purine analogs
such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine
analogs such as ancitabine, azacitidine,
6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine,
enocitabine, floxuridine, androgens such as
calusterone, dromostanolone propionate, epitiostanol, mepitiostane,
testolactone; anti-adrenals such as
aminoglutethimide, mitotane, trilostane; folic acid replenisher such as
frolinic acid; aceglatone; aldophosphamide
glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene;
edatraxate; defofamine; demecolcine;
diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate;
hydroxyurea; lentinan; lonidamine;
mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet;
pirarubicin; podophyllinic acid; 2-
ethylhydrazide; procarbazine; PSK.RTM; razoxane; sizofiran; spirogermanium;
tenuazonic acid; triaziquone;
2,2',2"-trichlorotriethyla- mine; urethan; vindesine; dacarbazine;
mannomustine; mitobronitol; mitolactol;
pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa;
taxanes, e.g., paclitaxel (TAXOLTm,
Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERETm,
Rhone-Poulenc Rorer, Antony,
France) and ABRAXANE (paclitaxel protein-bound particles); retinoic acid;
esperamicins; capecitabine; and
pharmaceutically acceptable forms (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs,
and isotopically labeled derivatives) of any of the above. Also included as
suitable chemotherapeutic cell
148

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
conditioners are anti-hormonal agents that act to regulate or inhibit hormone
action on tumors such as anti-estrogens
including for example tamoxifen (NolvadexTm), raloxifene, aromatase inhibiting
4(5)-imidazoles, 4-
hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and
toremifene (Fareston); and anti-androgens
such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin;
chlorambucil; gemcitabine; 6-thioguanine;
mercaptopurine; methotrexate; platinum analogs such as cisplatin and
carboplatin; vinblastine; platinum; etoposide
(VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine;
navelbine; novantrone; teniposide;
daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11);
topoisomerase inhibitor RFS 2000;
difluoromethylornithine (DMFO). Where desired, the compounds or pharmaceutical
composition as provided
herein can be used in combination with commonly prescribed anti-cancer drugs
such as Herceptin , Avastin ,
Erbitux , Rituxan , Taxol , Arimidex , Taxotere , ABVD, AVICINE, Abagovomab,
Acridine carboxamide,
Ade catumumab, 17-N-Allylamino-17-demethoxygeldanamycin, Alpharadin,
Alvocidib, 3 -Aminopyridine-2 -
carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22
immunotoxins, Antineoplastic,
Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan,
Bendamustine, BIBW 2992, Biricodar,
Brostallicin, Bryostatin, Buthionine sulfoximine, CBV (chemotherapy),
Calyculin, Crizotinib, cell-cycle nonspecific
antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin,
Enocitabine, Epothilone, Eribulin,
Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE
chemotherapy regimen, IT-101, Imexon,
Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide,
Lucanthone, Lurtotecan,
Mafosfamide, Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1,
Pawpaw, Pixantrone, Proteasome
inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A,
Sapacitabine, Stanford V,
Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel,
Triplatin tetranitrate, Tris(2-
chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126,
and Zosuquidar.
[00592] In some embodiments, the chemotherapeutic is selected from
hedgehog inhibitors including, but
not limited to IPI-926 (See U.S. Patent 7,812,164). Other suitable hedgehog
inhibitors include, for example, those
described and provided in U.S. Patent 7,230,004, U.S. Patent Application
Publication No. 2008/0293754, U.S.
Patent Application Publication No. 2008/0287420, and U.S. Patent Application
Publication No. 2008/0293755, the
entire disclosures of which are incorporated by reference herein. Examples of
other suitable hedgehog inhibitors
include those described in U.S. Patent Application Publication Nos. US
2002/0006931, US 2007/0021493 and US
2007/0060546, and International Application Publication Nos. WO 2001/19800, WO
2001/26644, WO 2001/27135,
WO 2001/49279, WO 2001/74344, WO 2003/011219, WO 2003/088970, WO 2004/020599,
WO 2005/013800,
WO 2005/033288, WO 2005/032343, WO 2005/042700, WO 2006/028958, WO
2006/050351, WO 2006/078283,
WO 2007/054623, WO 2007/059157, WO 2007/120827, WO 2007/131201, WO
2008/070357, WO 2008/110611,
WO 2008/112913, and WO 2008/131354. Additional examples of hedgehog inhibitors
include, but are not limited
to, GDC-0449 (also known as RG3616 or vismodegib) described in, e.g., Von Hoff
D. et al., N Engl. J Med. 2009;
361(12):1164-72; Robarge K.D. et al., Bioorg Med Chem Lett. 2009; 19(19):5576-
81; Yauch, R. L. et al. (2009)
Science 326: 572-574; Sciencexpress: 1-3 (10.1126/science.1179386); Rudin, C.
et al. (2009) New England J of
Medicine 361-366 (10.1056/nejma0902903); BMS-833923 (also known as XL139)
described in, e.g., in Siu L. et
al., J Clin. Oncol. 2010; 28:15s (suppl; abstr 2501); and National Institute
of Health Clinical Trial Identifier No.
149

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
NCT006701891; LDE-225 described, e.g., in Pan S. et al., ACS Med. Chem. Lett.,
2010; 1(3): 130-134; LEQ-506
described, e.g., in National Institute of Health Clinical Trial Identifier No.
NCT01106508; PF-04449913 described,
e.g., in National Institute of Health Clinical Trial Identifier No.
NCT00953758; Hedgehog pathway antagonists
provided in U.S. Patent Application Publication No. 2010/0286114; SM0i2-17
described, e.g., U.S. Patent
Application Publication No. 2010/0093625; SANT-1 and SANT-2 described, e.g.,
in Rominger C.M. et al., J
Pharmacol. Exp. Ther. 2009; 329(3):995-1005; 1-piperaziny1-4-arylphthalazines
or analogues thereof, described in
Lucas B.S. et al., Bioorg. Med. Chem. Lett. 2010; 20(12):3618-22.
[00593] Other chemotherapeutic agents include, but are not limited to, anti-
estrogens (e.g. tamoxifen, raloxifene,
and megestrol), LHRH agonists (e.g. goscrclin and leuprolide), anti-androgens
(e.g. flutamide and bicalutamide),
photodynamic therapies (e.g. vertoporfin (BPD-MA), phthalocyanine,
photosensitizer Pc4, and demethoxy-
hypocrellin A (2BA-2-DMHA)), nitrogen mustards (e.g. cyclophosphamide,
ifosfamide, trofosfamide, chlorambucil,
estramustine, and melphalan), nitrosoureas (e.g. carmustine (BCNU) and
lomustine (CCNU)), alkylsulphonates (e.g.
busulfan and treosulfan), triazenes (e.g. dacarbazine, temozolomide), platinum
containing compounds (e.g. cisplatin,
carboplatin, oxaliplatin), vinca alkaloids (e.g. vincristine, vinblastine,
vindesine, and vinorelbine), taxoids (e.g.
paclitaxel or a paclitaxel equivalent such as nanoparticle albumin-bound
paclitaxel (Abraxane), docosahexaenoic
acid bound-paclitaxel (DHA-paclitaxel, Taxoprexin), polyglutamate bound-
paclitaxel (PG-paclitaxel, paclitaxel
poliglumex, CT-2103, XYOTAX), the tumor-activated prodrug (TAP) ANG1005
(Angiopep-2 bound to three
molecules of paclitaxel), paclitaxel-EC-1 (paclitaxel bound to the erbB2-
recognizing peptide EC-1), and glucose-
conjugated paclitaxel, e.g., 2'-paclitaxel methyl 2-glucopyranosyl succinate;
docetaxel, taxol), epipodophyllins (e.g.
etoposide, etoposide phosphate, teniposide, topotecan, 9-aminocamptothecin,
camptoirinotecan, irinotecan,
crisnatol, mytomycin C), anti-metabolites, DHFR inhibitors (e.g. methotrexate,
dichloromethotrexate, trimetrexate,
edatrexate), IMP dehydrogenase inhibitors (e.g. mycophenolic acid, tiazofurin,
ribavirin, and EICAR), ribonuclotide
reductase inhibitors (e.g. hydroxyurea and deferoxamine), uracil analogs (e.g.
5-fluorouracil (5-FU), floxuridine,
doxifluridine, ratitrexed, tegafur-uracil, capecitabine), cytosine analogs
(e.g. cytarabine (ara C), cytosine
arabinoside, and fludarabine), purine analogs (e.g. mercaptopurine and
Thioguanine), Vitamin D3 analogs (e.g. EB
1089, CB 1093, and KH 1060), isoprenylation inhibitors (e.g. lovastatin),
dopaminergic neurotoxins (e.g. 1-methyl-
4-phenylpyridinium ion), cell cycle inhibitors (e.g. staurosporine),
actinomycin (e.g. actinomycin D, dactinomycin),
bleomycin (e.g. bleomycin A2, bleomycin B2, peplomycin), anthracycline (e.g.
daunorubicin, doxorubicin,
pegylated liposomal doxorubicin, idarubicin, epirubicin, pirarubicin,
zorubicin, mitoxantrone), MDR inhibitors (e.g.
verapamil), Ca2+ ATPase inhibitors (e.g. thapsigargin), imatinib, thalidomide,
lenalidomide, tyrosine kinase
inhibitors (e.g., axitinib (AG013736), bosutinib (SKI-606), cediranib
(RECENTINTM, AZD2171), dasatinib
(SPRYCELO, BMS-354825), erlotinib (TARCEVAO), gefitinib (IRESSAO), imatinib
(GleevecO, CGP57148B,
STI-571), lapatinib (TYKERBO, TYVERBO), lestaurtinib (CEP-701), neratinib (HKI-
272), nilotinib
(TASIGNAO), semaxanib (semaxinib, SU5416), sunitinib (SUTENTO, SU11248),
toceranib (PALLADIA ),
vandetanib (ZACTIMAO, ZD6474), vatalanib (PTK787, PTK/ZK), trastuzumab
(HERCEPTINO), bevacizumab
(AVASTINO), rituximab (RITUXANO), cetuximab (ERBITUXO), panitumumab
(VECTIBIXO), ranibizumab
(Lucentis0), nilotinib (TASIGNAO), sorafenib (NEXAVARO), everolimus
(AFINITORO), alemtuzumab
150

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
(CAMPATHO), gemtuzumab ozogamicin (MYLOTARGO), temsirolimus (TORISELO), ENMD-
2076, PCI-32765,
AC220, dovitinib lactate (TKI258, CHIR-258), BIBW 2992 (TOVOKTM), SGX523, PF-
04217903, PF-02341066,
PF-299804, BMS-777607, ABT-869, MP470, BIBF 1120 (VARGATEFO), AP24534, JNJ-
26483327, MGCD265,
DCC-2036, BMS-690154, CEP-11981, tivozanib (AV-951), OSI-930, MM-121, XL-184,
XL-647, and/or XL228),
proteasome inhibitors (e.g., bortezomib (Velcade)), mTOR inhibitors (e.g.,
rapamycin, temsirolimus (CCI-779),
everolimus (RAD-001), ridaforolimus, AP23573 (Ariad), AZD8055 (AstraZeneca),
BEZ235 (Novartis), BGT226
(Norvartis), XL765 (Sanofi Aventis), PF-4691502 (Pfizer), GDC0980 (Genetech),
SF1126 (Semafoe) and OSI-027
(OSI)), oblimersen, gemcitabine, carminomycin, leucovorin, pemetrexed,
cyclophosphamide, dacarbazine,
procarbizine, prednisolone, dexamethasone, campathecin, plicamycin,
asparaginase, aminopterin, methopterin,
porfiromycin, melphalan, leurosidine, leurosine, chlorambucil, trabectedin,
procarbazine, discodermolide,
carminomycinõ aminopterin, and hexamethyl melamine.
[00594]
Exemplary biotherapeutic agents include, but are not limited to, interferons,
cytokines (e.g., tumor
necrosis factor, interferon a, interferon 7), vaccines, hematopoietic growth
factors, monoclonal serotherapy,
immunostimulants and/or immunodulatory agents (e.g., IL-1, 2, 4, 6, or 12),
immune cell growth factors (e.g., GM-
CSF) and antibodies (e.g. Herceptin (trastuzumab), T-DM1, AVASTIN
(bevacizumab), ERBITUX (cetuximab),
Vectibix (panitumumab), Rituxan (rituximab), Bexxar (tositumomab)).
[00595]
In some embodiments, the chemotherapeutic is selected from HSP90 inhibitors.
The HSP90
inhibitor can be a geldanamycin derivative, e.g., a benzoquinone or
hygroquinone ansamycin HSP90 inhbitor (e.g.,
IPI-493 and/or IPI-504). Non-limiting examples of HSP90 inhibitors include IPI-
493, IPI-504, 17-AAG (also
known as tanespimycin or CNF-1010), BIIB-021 (CNF-2024), BIIB-028, AUY-922
(also known as VER-49009),
SNX-5422, STA-9090, AT-13387, XL-888, MPC-3100, CU-0305, 17-DMAG, CNF-1010,
Macbecin (e.g.,
Macbecin I, Macbecin II), CCT-018159, CCT-129397, PU-H71, or PF-04928473 (SNX-
2112).
[00596]
In some embodiments, the chemotherapeutic is selected from PI3K inhibitors
(e.g., including
those PI3K inhibitors provided herein and those PI3K inhibitors not provided
herein). In some embodiment, the
PI3K inhibitor is an inhibitor of delta and gamma isoforms of P13 K. In some
embodiments, the PI3K inhibitor is an
inhibitor of alpha isoforms of P13 K. In other embodiments, the PI3K inhibitor
is an inhibitor of one or more alpha,
beta, delta and gamma isoforms of PI3K. Exemplary PI3K inhibitors that can be
used in combination are described
in, e.g., WO 09/088990, WO 09/088086, WO 2011/008302, WO 2010/036380, WO
2010/006086, WO 09/114870,
WO 05/113556; US 2009/0312310, and US 2011/0046165. Additional PI3K inhibitors
that can be used in
combination with the pharmaceutical compositions, include but are not limited
to, AMG-319, GSK 2126458, GDC-
0980, GDC-0941, Sanofi XL147, XL499, XL756, XL147, PF-46915032, BKM 120, CAL-
101 (GS-1101), CAL
263, SF1126, PX-886, and a dual PI3K inhibitor (e.g., Novartis BEZ235). In one
embodiment, the PI3K inhibitor is
an isoquinolinone.
[00597]
In some embodiments, provided herein is a method for using the a compound as
provided
herein, or a pharmaceutically acceptable form (e.g., pharmaceutically
acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives) thereof, or pharmaceutical
compositions as provided herein, in
combination with radiation therapy in inhibiting abnormal cell growth or
treating the hyperproliferative disorder in
151

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
the subject. Techniques for administering radiation therapy are known in the
art, and these techniques can be used in
the combination therapy described herein. The administration of the compound
as provided herein in this
combination therapy can be determined as described herein.
[00598] Radiation therapy can be administered through one of several
methods, or a combination of
methods, including without limitation external-beam therapy, internal
radiation therapy, implant radiation,
stereotactic radiosurgery, systemic radiation therapy, radiotherapy and
permanent or temporary interstitial
brachytherapy. The term "brachytherapy," as used herein, refers to radiation
therapy delivered by a spatially
confined radioactive material inserted into the body at or near a tumor or
other proliferative tissue disease site. The
term is intended without limitation to include exposure to radioactive
isotopes (e.g., At-211, 1-131, 1-125, Y-90, Re-
186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable
radiation sources for use as a cell
conditioner as provided herein include both solids and liquids. By way of non-
limiting example, the radiation
source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid
source, 1-125 as a solid source, or other
radionuclides that emit photons, beta particles, gamma radiation, or other
therapeutic rays. The radioactive material
can also be a fluid made from any solution of radionuclide(s), e.g., a
solution of 1-125 or 1-131, or a radioactive fluid
can be produced using a slurry of a suitable fluid containing small particles
of solid radionuclides, such as Au-198,
Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive
micro spheres.
[00599] Without being limited by any theory, the compounds as provided
herein, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein, can render abnormal
cells more sensitive to treatment with radiation for purposes of killing
and/or inhibiting the growth of such cells.
Accordingly, provided herein is a method for sensitizing abnormal cells in a
subject to treatment with radiation
which comprises administering to the subject an amount of a compound as
provided herein or pharmaceutically
acceptable forms (e.g., pharmaceutically acceptable salts, hydrates, solvates,
isomers, prodrugs, and isotopically
labeled derivatives) thereof, which amount is effective is sensitizing
abnormal cells to treatment with radiation. The
amount of the compound used in this method can be determined according to the
means for ascertaining effective
amounts of such compounds described herein.
[00600] The compounds as provided herein, or a pharmaceutically
acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, can be used in
combination with an amount of one or
more substances selected from anti-angiogenesis agents, signal transduction
inhibitors, and antiproliferative agents,
glycolysis inhibitors, or autophagy inhibitors.
[00601] Anti-angiogenesis agents, such as MMP-2 (matrix-
metalloproteinase 2) inhibitors, MMP-9
(matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11)
inhibitors, can be used in conjunction
with a compound as provided herein and pharmaceutical compositions described
herein. Anti-angiogenesis agents
include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001),
sorafenib, sunitinib, and
bevacizumab. Examples of useful COX-II inhibitors include CELEBREXTM
(alecoxib), valdecoxib, and rofecoxib.
Examples of useful matrix metalloproteinase inhibitors are described in WO
96/33172 (published October 24,
152

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
1996), WO 96/27583 (published March 7, 1996), European Patent Application No.
97304971.1 (filed July 8, 1997),
European Patent Application No. 99308617.2 (filed October 29, 1999), WO
98/07697 (published February 26,
1998), WO 98/03516 (published January 29, 1998), WO 98/34918 (published August
13, 1998), WO 98/34915
(published August 13, 1998), WO 98/33768 (published August 6, 1998), WO
98/30566 (published July 16, 1998),
European Patent Publication 606,046 (published July 13, 1994), European Patent
Publication 931, 788 (published
July 28, 1999), WO 90/05719 (published May 31, 1990), WO 99/52910 (published
October 21, 1999), WO
99/52889 (published October 21, 1999), WO 99/29667 (published June 17, 1999),
PCT International Application
No. PCT/IB98/01113 (filed July 21, 1998), European Patent Application No.
99302232.1 (filed March 25, 1999),
Great Britain Patent Application No. 9912961.1 (filed June 3, 1999), United
States Provisional Application No.
60/148,464 (filed August 12, 1999), United States Patent 5,863,949 (issued
January 26, 1999), United States Patent
5,861,510 (issued January 19, 1999), and European Patent Publication 780,386
(published June 25, 1997), all of
which are incorporated herein in their entireties by reference. In some
embodiments, MMP-2 and MMP-9 inhibitors
are those that have little or no activity inhibiting MMP-1. Other embodiments
include those that selectively inhibit
MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i.e., MAP-
1, MMP-3, MMP-4, MMP-5,
MMP-6, MMP- 7, MMP-8, MMP-10, MMP-11, MMP-12, andMMP-13). Some non-limiting
examples of MMP
inhibitors are AG-3340, RO 32-3555, and RS 13-0830.
[00602] Autophagy inhibitors include, but are not limited to,
chloroquine, 3-methyladenine,
hydroxychloroquine (PlaquenilTm), bafilomycin Al, 5-amino-4-imidazole
carboxamide riboside (AICAR), okadaic
acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of
type 2A or type 1, analogues of
cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-
mercaptopurine riboside, and
vinblastine. In addition, antisense or siRNA that inhibits expression of
proteins including, but not limited to ATG5
(which are implicated in autophagy), can also be used.
[00603] In some embodiments, provided herein is a method of and/or a
pharmaceutical composition
for treating a cardiovascular disease in a subject which comprises an amount
of a compound as provided herein, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, and an amount of one or more
therapeutic agents use for the treatment of
cardiovascular diseases.
[00604] Exemplary agents for use in cardiovascular disease applications
are anti-thrombotic agents,
e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase,
urokinase, tissue plasminogen activator
(TPA) and anisoylated plasminogen-streptokinase activator complex (APSAC),
anti-platelets agents, e.g., acetyl-
salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates,
calcium channel blocking drugs, anti-
proliferative agents, e.g., colchicine and alkylating agents, intercalating
agents, growth modulating factors such as
interleukins, transformation growth factor-beta and congeners of platelet
derived growth factor, monoclonal
antibodies directed against growth factors, anti-inflammatory agents, both
steroidal and non-steroidal, and other
agents that can modulate vessel tone, function, arteriosclerosis, and the
healing response to vessel or organ injury
post intervention. Antibiotics can also be included in combinations or
coatings. Moreover, a coating can be used to
153

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
effect therapeutic delivery focally within the vessel wall. By incorporation
of the active agent in a swellable
polymer, the active agent will be released upon swelling of the polymer.
[00605] The compounds as provided herein, or a pharmaceutically
acceptable form (e.g.,
pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives)
thereof, or pharmaceutical compositions as provided herein, can be formulated
or administered in conjunction with
liquid or solid tissue barriers also known as lubricants. Examples of tissue
barriers include, but are not limited to,
polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid.
[00606] Medicaments which can be administered in conjunction with the
compounds as provided
herein, or a pharmaceutically acceptable form (e.g., pharmaceutically
acceptable salts, hydrates, solvates, isomers,
prodrugs, and isotopically labeled derivatives) thereof, include any suitable
drugs usefully delivered by inhalation
for example, analgesics, e.g., codeine, dihydromorphine, ergotamine, fentanyl
or morphine; anginal preparations,
e.g., diltiazem; antiallergics, e.g. cromoglycate, ketotifen or nedocromil;
anti-infectives, e.g., cephalosporins,
penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine;
antihistamines, e.g., methapyrilene; anti-
inflammatories, e.g., beclomethasone, flunisolide, budesonide, tipredane,
triamcinolone acetonide or fluticasone;
antitussives, e.g., noscapine; bronchodilators, e.g., ephedrine, adrenaline,
fenoterol, formoterol, isoprenaline,
metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol,
rimiterol, salbutamol, salmeterol,
terbutalin, isoetharine, tulobuterol, orciprenaline or (-)-4-amino-3,5-
dichloro-a-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-
amino]methyl]benzenemethanol; diuretics, e.g., amiloride; anticholinergics
e.g., ipratropium, atropine or
oxitropium; hormones, e.g., cortisone, hydrocortisone or prednisolone;
xanthines e.g., aminophylline, choline
theophyllinate, lysine theophyllinate or theophylline; and therapeutic
proteins and peptides, e.g., insulin or glucagon.
It will be clear to a person skilled in the art that, where appropriate, the
medicaments can be used in the form of salts
(e.g., as alkali metal or amine salts or as acid addition salts) or as esters
(e.g., lower alkyl esters) to optimize the
activity and/or stability of the medicament.
[00607] Other exemplary therapeutic agents useful for a combination
therapy include, but are not
limited to, agents as described above, radiation therapy, hormone antagonists,
hormones and their releasing factors,
thyroid and antithyroid drugs, estrogens and progestins, androgens,
adrenocorticotropic hormone; adrenocortical
steroids and their synthetic analogs; inhibitors of the synthesis and actions
of adrenocortical hormones, insulin, oral
hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents
affecting calcification and bone
turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin,
vitamins such as water-soluble vitamins,
vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E,
growth factors, cytokines,
chemokines, muscarinic receptor agonists and antagonists; anticholinesterase
agents; agents acting at the
neuromuscular junction and/or autonomic ganglia; catecholamines,
sympathomimetic drugs, and adrenergic receptor
agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor
agonists and antagonists.
[00608] Therapeutic agents can also include agents for pain and
inflammation such as histamine and
histamine antagonists, bradykinin and bradykinin antagonists, 5-
hydroxytryptamine (serotonin), lipid substances that
are generated by biotransformation of the products of the selective hydrolysis
of membrane phospholipids,
eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal
anti-inflammatory agents, analgesic-
154

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
antipyretic agents, agents that inhibit the synthesis of prostaglandins and
thromboxanes, selective inhibitors of the
inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-
2, autacoids, paracrine hormones,
somatostatin, gastrin, cytokines that mediate interactions involved in humoral
and cellular immune responses, lipid-
derived autacoids, eicosanoids, 13-adrenergic agonists, ipratropium,
glucocorticoids, methylxanthines, sodium
channel blockers, opioid receptor agonists, calcium channel blockers, membrane
stabilizers and leukotriene
inhibitors.
[00609] Additional therapeutic agents contemplated herein include
diuretics, vasopressin, agents
affecting the renal conservation of water, rennin, angiotensin, agents useful
in the treatment of myocardial ischemia,
anti-hypertensive agents, angiotensin converting enzyme inhibitors, 13-
adrenergic receptor antagonists, agents for the
treatment of hypercholesterolemia, and agents for the treatment of
dyslipidemia.
[00610] Other therapeutic agents contemplated herein include drugs used
for control of gastric acidity,
agents for the treatment of peptic ulcers, agents for the treatment of
gastroesophageal reflux disease, prokinetic
agents, antiemetics, agents used in irritable bowel syndrome, agents used for
diarrhea, agents used for constipation,
agents used for inflammatory bowel disease, agents used for biliary disease,
agents used for pancreatic disease.
Therapeutic agents include, but are not limited to, those used to treat
protozoan infections, drugs used to treat
Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or
Leishmaniasis, and/or drugs used in the
chemotherapy of helminthiasis. Other therapeutic agents include, but are not
limited to, antimicrobial agents,
sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary
tract infections, penicillins,
cephalosporins, and other, P-Lactam antibiotics, an agent containing an
aminoglycoside, protein synthesis inhibitors,
drugs used in the chemotherapy of tuberculosis, mycobacterium avium complex
disease, and leprosy, antifungal
agents, antiviral agents including norn-etroviral agents and antiretroviral
agents.
[00611] Examples of therapeutic antibodies that can be combined with a
subject compound include
but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab,
panitumumab, trastuzumab), anti CD20
antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab,
bevacizumab, and gemtuzumab.
[00612] Moreover, therapeutic agents used for immunomodulation, such as
immunomodulators,
immunosuppressive agents, tolerogens, and immunostimulants are contemplated by
the methods herein. In addition,
therapeutic agents acting on the blood and the blood-forming organs,
hematopoietic agents, growth factors,
minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
[00613] For treating renal carcinoma, one can combine a a compound as
provided herein, or a
pharmaceutically acceptable form (e.g., pharmaceutically acceptable salts,
hydrates, solvates, isomers, prodrugs, and
isotopically labeled derivatives) thereof, or pharmaceutical compositions as
provided herein, with sorafenib and/or
avastin. For treating an endometrial disorder, one can combine a compound as
provided herein with doxorubincin,
taxotere (taxol), and/or cisplatin (carboplatin). For treating ovarian cancer,
one can combine a compound as
provided herein with cisplatin (carboplatin), taxotere, doxorubincin,
topotecan, and/or tamoxifen. For treating breast
cancer, one can combine a compound as provided herein with taxotere (taxol),
gemcitabine (capecitabine),
tamoxifen, letrozole, tarceva, lapatinib, PD0325901, avastin, herceptin, OSI-
906, and/or OSI-930. For treating lung
155

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
cancer, one can combine a compound as provided herein with taxotere (taxol),
gemcitabine, cisplatin, pemetrexed,
Tarceva, PD0325901, and/or avastin.
[00614] In some embodiments, the disorder to be treated, prevented and/or
managed is hematological
cancer, e.g., lymphoma (e.g., T-cell lymphoma; NHL), myeloma (e.g., multiple
myeloma), and leukemia (e.g.,
CLL), and a compound provided herein is used in combination with: HDAC
inhibitors such as vorinostat and
romidepsin; mTOR inhibitors such as everolmus; anti-folates such as
pralatrexate; nitrogen mustard such as
bendamustine; gemcitabine, optionally in further combination with oxaliplatin;
rituximab.cyclophosphamide
combination; PI3K inhibitors such as GS-1101, XL 499, GDC-0941, and AMG-319;
or BTK inhibitors such as
ibrutinib and AVL-292.
[00615] In certain embodiments, wherein inflammation (e.g., arthritis,
asthma) is treated, prevented
and/or managed, a compound provided herein can be combined with, for example:
PI3K inhibitors such as GS-1101,
XL 499, GDC-0941, and AMG-319; BTK inhibitors such as ibrutinib and AVL-292;
JAK inhibitors such as
tofacitinib, fostamatinib, and GLPG0636.
[00616] In certain embodiments wherein asthma is treated, prevented
and/or managed, a compound
provided herein can be combined with, for example: beta 2-agonists such as,
but not limited to, albuterol
(Proventi10, or Ventolin0), salmeterol (Serevent0), formoterol (Foradi10),
metaproterenol (Alupent0), pirbuterol
(MaxAir0), and terbutaline sulfate; corticosteroids such as, but not limited
to, budesonide (e.g., Pulmicort0),
flunisolide (e.g., AeroBid Oral Aerosol Inhaler or Nasalide Nasal Aerosol ),
fluticasone (e.g., Flonase0 or
Flovent0) and triamcinolone (e.g., Azmacort0); mast cell stabilizers such as
cromolyn sodium (e.g., Intal0 or
Nasalcrom0) and nedocromil (e.g., Tilade0); xanthine derivatives such as, but
not limited to, theophylline (e.g.,
AminophyllinO, Theo-24 or Theolair0); leukotriene receptor antagonists such
as, but are not limited to,
zafirlukast (Accolate0), montelukast (Singulair0), and zileuton (Zyflo0); and
adrenergic agonists such as, but are
not limited to, epinephrine (AdrenalinO, BronitinO, EpiPen0 or Primatene Mist
).
[00617] In certain embodiments wherein arthritis is treated, prevented
and/or managed, a compound
provided herein can be combined with, for example: TNF antagonist (e.g., a TNF
antibody or fragment, a soluble
TNF receptor or fragment, fusion proteins thereof, or a small molecule TNF
antagonist); an antirheumatic (e.g.,
methotrexate, auranofin, aurothioglucose, azathioprine, etanercept, gold
sodium thiomalate, hydroxychloroquine
sulfate, leflunomide, sulfasalzine); a muscle relaxant; a narcotic; a non-
steroid anti-inflammatory drug (NSAID); an
analgesic; an anesthetic; a sedative; a local anesthetic; a neuromuscular
blocker; an antimicrobial (e.g., an
aminoglycoside, an antifungal, an antiparasitic, an antiviral, a carbapenem,
cephalosporin, a fluoroquinolone, a
macrolide, a penicillin, a sulfonamide, a tetracycline, another
antimicrobial); an antipsoriatic; a corticosteroid; an
anabolic steroid; a cytokine or a cytokine antagonist.
[00618] In certain embodiments wherein psoriasis is treated, prevented
and/or managed, a compound
provided herein can be combined with, for example: budesonide, epidermal
growth factor, corticosteroids,
cyclosporine, sulfasalazine, aminosalicylates, 6-mercaptopurine, azathioprine,
metronidazole, lipoxygenase
inhibitors, mesalamine, olsalazine, balsalazide, antioxidants, thromboxane
inhibitors, IL-1 receptor antagonists, anti-
IL-1I3 monoclonal antibodies, anti-IL-6 monoclonal antibodies, growth factors,
elastase inhibitors, pyridinyl-
156

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
imidazole compounds, antibodies or agonists of TNF, LT, IL-1, IL-2, IL-6, IL-
7, IL-8, IL-15, IL-16, IL-18, EMAP-
II, GM-CSF, FGF, and PDGF, antibodies of CD2, CD3, CD4, CD8, CD25, CD28, CD30,
CD40, CD45, CD69,
CD90 or their ligands, methotrexate, cyclosporine, FK506, rapamycin,
mycophenolate mofetil, leflunomide,
NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase
inhibitors, adenosine agonists, antithrombotic
agents, complement inhibitors, adrenergic agents, IRAK, NIK, IKK, p38, MAP
kinase inhibitors, IL-113 converting
enzyme inhibitors, TNFa converting enzyme inhibitors, T-cell signaling
inhibitors, metalloproteinase inhibitors,
sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme
inhibitors, soluble cytokine
receptors, soluble p55 TNF receptor, soluble p75 TNF receptor, sIL-1RI, sIL-
1RII, sIL-6R, anti-inflammatory
cytokines, IL-4, IL-10, IL-11, IL-13 and TGFI3.
[00619] In certain embodiments wherein fibrosis or fibrotic condition of
the bone marrow is treated,
prevented and/or managed, a compound provided herein can be combined with, for
example, a Jak2 inhibitor
(including, but not limited to, INCB018424, XL019, TG101348, or TG101209), an
immunomodulator, e.g., an
IMIDO (including, but not limited to thalidomide, lenalidomide, or
panolinomide), hydroxyurea, an androgen,
erythropoietic stimulating agents, prednisone, danazol, HDAC inhibitors, or
other agents or therapeutic modalities
(e.g., stem cell transplants, or radiation).
[00620] In certain embodiments wherein fibrosis or fibrotic condition of
the heart is treated, prevented
and/or managed, a compound provided herein can be combined with, for example,
eplerenone, furosemide,
pycnogenol, spironolactone, TcNC100692, torasemide (e.g., prolonged release
form of torasemide), or combinations
thereof.
[00621] In certain embodiments wherein fibrosis or fibrotic condition of
the kidney is treated,
prevented and/or managed, a compound provided herein can be combined with, for
example, cyclosporine,
cyclosporine A, daclizumab, everolimus, gadofoveset trisodium (ABLAVARO),
imatinib mesylate (GLEEVECO),
matinib mesylate, methotrexate, mycophenolate mofetil, prednisone, sirolimus,
spironolactone, STX-100,
tamoxifen, TheraCLECTm, or combinations thereof.
[00622] In certain embodiments wherein fibrosis or fibrotic condition of
the skin is treated, prevented
and/or managed, a compound provided herein can be combined with, for example,
Bosentan (Tracleer), p144,
pentoxifylline; pirfenidone; pravastatin, STI571, Vitamin E, or combinations
thereof.
[00623] In certain embodiments wherein fibrosis or fibrotic condition of
the gastrointestinal system is
treated, prevented and/or managed, a compound provided herein can be combined
with, for example, ALTU-135,
bucelipase alfa (INN), DCI1020, EUR-1008 (ZENPEPTm), ibuprofen, Lym-X-Sorb
powder, pancrease MT,
pancrelipase (e.g., pancrelipase delayed release), pentade canoic acid (PA),
repaglinide, TheraCLECTm,
triheptadecanoin (THA), ULTRASE MT20, ursodiol, or combinations thereof.
[00624] In certain embodiments wherein fibrosis or fibrotic condition of
the lung is treated, prevented
and/or managed, a compound provided herein can be combined with, for example,
18-FDG, AB0024, ACT-064992
(macitentan), aerosol interferon-gamma, aerosolized human plasma-derived alpha-
1 antitrypsin, alphal-proteinase
inhibitor, ambrisentan, amikacin, amiloride, amitriptyline, anti-pseudomonas
IgY gargle, ARIKACETM,
AUREXISO (tefibazumab), AZAPRED, azathioprine, azithromycin, azithromycin,
AZLI, aztreonam lysine,
157

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
BIBF1120, Bio-25 probiotic, bosentan, Bramitob0 , calfactant aerosol,
captopril, CC-930, ceftazidime, ceftazidime,
cholecalciferol (Vitamin D3), ciprofloxacin (CIPROO, BAYQ3939), CNTO 888,
colistin CF, combined Plasma
Exchange (PEX), rituximab, and corticosteroids, cyclophosphamide, dapsone,
dasatinib, denufosol tetrasodium
(INS37217), dornase alfa (PULMOZYMEO), EPI-hNE4, erythromycin, etanercept, FG-
3019, fluticasone, FTI,
GC1008, GS-9411, hypertonic saline, ibuprofen, iloprost inhalation, imatinib
mesylate (GLEEVECO), inhaled
sodium bicarbonate, inhaled sodium pyruvate, interferon gamma-lb, interferon-
alpha lozenges, isotonic saline,
IWO01, KB001, losartan, lucinactant, mannitol, meropenem, meropenem infusion,
miglustat, minocycline,
Moli1901, MP-376 (levofloxacin solution for inhalation), mucoid
exopolysaccharide P. aeruginosa immune globulin
IV, mycophenolate mofetil, n-acetylcysteine, N-acetylcysteine (NAC), NaC1 6%,
nitric oxide for inhalation,
obramycin, octreotide, oligoG CF-5/20, Omalizumab, pioglitazone, piperacillin-
tazobactam, pirfenidone,
pomalidomide (CC-4047), prednisone, prevastatin, PRM-151, QAX576, rhDNAse, SB
656933, SB -656933 -AAA,
sildenafil, tamoxifen, technetium [Tc-99m] sulfur colloid and Indium [In-111]
DTPA, tetrathiomolybdate,
thalidomide, ticarcillin-clavulanate, tiotropium bromide, tiotropium RESPIMATO
inhaler, tobramycin
(GERNEBCINO), treprostinil, uridine, valganciclovir (VALCYTEO), vardenafil,
vitamin D3, xylitol, zileuton, or
combinations thereof.
[00625] In certain embodiments wherein fibrosis or fibrotic condition of
the liver is treated, prevented
and/or managed, a compound provided herein can be combined with, for example,
adefovir dipivoxil, candesartan,
colchicine, combined ATG, mycophenolate mofetil, and tacrolimus, combined
cyclosporine microemulsion and
tacrolimus, elastometry, everolimus, FG-3019, Fuzheng Huayu, GI262570,
glycyrrhizin (monoammonium
glycyrrhizinate, glycine, L-cysteine monohydrochloride), interferon gamma-lb,
irbesartan, losartan, oltipraz, ORAL
IMPACT , peginterferon alfa-2a, combined peginterferon alfa-2a and ribavirin,
peginterferon alfa-2b (SCH 54031),
combined peginterferon alpha-2b and ribavirin, praziquantel, prazosin,
raltegravir, ribavirin (REBETOLO, SCH
18908), ritonavir-boosted protease inhibitor, pentoxyphilline, tacrolimus,
tauroursodeoxycholic acid, tocopherol,
ursodiol, warfarin, or combinations thereof.
[00626] In certain embodiments wherein cystic fibrosis is treated,
prevented and/or managed, a
compound provided herein can be combined with, for example, 552-02, 5-
methyltetrahydrofolate and vitamin B12,
Ad5-CB-CFTR, Adeno-associated virus-CFTR vector, albuterol, alendronate, alpha
tocopherol plus ascorbic acid,
amiloride HC1, aquADEKTM, ataluren (PTC124), AZD1236, AZD9668, azithromycin,
bevacizumab, biaxin
(clarithromycin), BIIL 283 BS (amelubent), buprofen, calcium carbonate,
ceftazidime, cholecalciferol, choline
supplementation, CPX, cystic fibrosis transmembrane conductance regulator, DHA-
rich supplement, digitoxin,
cocosahexaenoic acid (DHA), doxycycline, ECGC, ecombinant human IGF-1, educed
glutathione sodium salt,
ergocalciferol (vitamin D2), fluorometholone, gadobutrol (GADOVISTO, BAY86-
4875), gentamicin, ghrelin,
glargine, glutamine, growth hormone, GS-9411, H5.001CBCFTR, human recombinant
growth hormone,
hydroxychloroquine, hyperbaric oxygen, hypertonic saline, IH636 grape seed
proanthocyanidin extract, insulin,
interferon gamma-lb, IoGen (molecular iodine), iosartan potassium, isotonic
saline, itraconazole, IV gallium nitrate
(GANITEO) infusion, ketorolac acetate, lansoprazole, L-arginine, linezolid,
lubiprostone, meropenem, miglustat,
MP-376 (levofloxacin solution for inhalation), normal saline IV, Nutropin AQ,
omega-3 triglycerides,
158

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
pGM169/GL67A, pGT-1 gene lipid complex, pioglitazone, PTC124, QAU145,
salmeterol, SB656933, SB656933,
simvastatin, sitagliptin, sodium 4-phenylbutyrate, standardized turmeric root
extract, tgAAVCF, TNF blocker,
TOBI, tobramycin, tocotrienol, unconjugated Isoflavones 100, vitamin: choline
bitartrate (2-hydroxyethyl)
trimethylammonium salt 1:1, VX-770, VX-809, Zinc acetate, or combinations
thereof.
[00627] In some embodiments, a compound provided herein is administered
in combination with an
agent that inhibits IgE production or activity. In some embodiments, the PI3K
inhibitor (e.g., PI3K6 inhibitor) is
administered in combination with an inhibitor of mTOR. Agents that inhibit IgE
production are known in the art
and they include but are not limited to one or more of TEI-9874, 2-(4-(6-
cyclohexyloxy-2-
naphtyloxy)phenylacetamide)benzoic acid, rapamycin, rapamycin analogs (i.e.
rapalogs), TORC1 inhibitors,
TORC2 inhibitors, and any other compounds that inhibit mTORC1 and mTORC2.
Agents that inhibit IgE activity
include, for example, anti-IgE antibodies such as for example Omalizumab and
TNX-901.
[00628] In certain embodiments wherein scleroderma is treated, prevented
and/or managed, a
compound provided herein can be combined with, for example: an
immunosuppressant (e.g., methotrexate,
azathioprine (Imuran0), cyclosporine, mycophenolate mofetil (Cellcept0), and
cyclophosphamide (Cytoxan0)); T-
cell-directed therapy (e.g., halofuginone, basiliximab, alemtuzumab,
abatacept, rapamycin); B-cell directed therapy
(e.g., rituximab); autologous hematopoietic stem cell transplantation; a
chemokine ligand receptor antagonist (e.g.,
an agent that targets the CXCL12/CSCR4 axis (e.g., AMD3100)); a DNA
methylation inhibitor (e.g., 5-azacytidine);
a histone dactylase inhibitor (e.g., trichostatin A); a statin (e.g.,
atorvastatin, simvastatin, pravastatin); an endothelin
receptor antagonist (e.g., Bosentan0); a phosphodiesterase type V inhibitor
(e.g., Sildenafil0); a prostacyclin analog
(e.g., trepostinil); an inhibitor of cytokine synthesis and/or signaling
(e.g., Imatinib mesylate, Rosiglitazone,
rapamycin, antitransforming growth factor 131 (anti-TGFI31) antibody,
mycophenolate mofetil, an anti-IL-6 antibody
(e.g., tocilizumab)); corticosteroids; nonsteroidal anti-inflammatory drugs;
light therapy; and blood pressure
medications (e.g., ACE inhibitors).
[00629] In certain embodiments wherein inflammatory myopathies are
treated, prevented and/or
managed, a compound provided herein can be combined with, for example: topical
creams or ointments (e.g., topical
corticosteroids, tacrolimus, pimecrolimus); cyclosporine (e.g., topical
cyclosporine); an anti-interferon therapy, e.g.,
AGS-009, Rontalizumab (rhuMAb IFNalpha), Vitamin D3, Sifalimumab (MEDI-545),
AMG 811, IFNa Kinoid, or
CEP33457. In some embodiments, the other therapy is an IFN-a therapy, e.g.,
AGS-009, Rontalizumab, Vitamin
D3, Sifalimumab (MEDI-545) or IFNa Kinoid; corticosteroids such as prednisone
(e.g., oral prednisone);
immunosuppressive therapies such as methotrexate (Trexa110, Methotrexate0,
Rheumatrex0), azathioprine
(AzasanO, Imuran0), intravenous immunoglobulin, tacrolimus (Prograf0),
pimecrolimus, cyclophosphamide
(Cytoxan0), and cyclosporine (Gengraf0, Neora10, Sandimmune0); anti-malarial
agents such as
hydroxychloroquine (Plaqueni10) and chloroquine (Aralen0); total body
irradiation; rituximab (Rituxan0); TNF
inhibitors (e.g., etanercept (Enbre10), infliximab (Remicade0)); AGS-009;
Rontalizumab (rhuMAb IFNalpha);
Vitamin D3; Sifalimumab (MEDI-545); AMG 811; IFNa Kinoid,; CEP33457; agents
that inhibit IgE production
such as TEI-9874, 2-(4-(6-cyclohexyloxy-2-naphtyloxy)phenylacetamide)benzoic
acid, rapamycin, rapamycin
analogs (i.e. rapalogs), TORC1 inhibitors, TORC2 inhibitors, and any other
compounds that inhibit mTORC1 and
159

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
mTORC2; agents that inhibit IgE activity such as anti-IgE antibodies (e.g.,
Omalizumab and TNX-90); and
additional therapies such as physical therapy, exercise, rest, speech therapy,
sun avoidance, heat therapy, and
surgery.
[00630]
In certain embodiments wherein myositis (e.g., dermatomysitis) is treated,
prevented and/or
managed, a compound provided herein can be combined with, for example:
corticosteroids; corticosteroid sparing
agents such as, but not limited to, azathioprine and methotrexate; intravenous
immunoglobulin; immunosuppressive
agents such as, but not limited to, tacrolimus, cyclophosphamide and
cyclosporine; rituximab; TNFa inhibitors such
as, but not limited to, etanercept and infliximab; growth hormone; growth
hormone secretagogues such as, but not
limited to, MK-0677, L-162752, L-163022, NN703 ipamorelin, hexarelin, GPA-748
(KP102, GHRP-2), and
LY444711 (Eli Lilly); other growth hormone release stimulators such as, but
not limited to, Geref, GHRH (1-44),
Somatorelin (GRF 1-44), ThGRF genotropin, L-DOPA, glucagon, and vasopressin;
and insulin-like growth factor.
[00631]
In certain embodiments wherein Sjogren's syndrome is treated, prevented and/or
managed, a
compound provided herein can be combined with, for example: pilocarpine;
cevimeline; nonsteroidal anti-
inflammatory drugs; arthritis medications; antifungal agents; cyclosporine;
hydroxychloroquine; prednisone;
azathioprine; and cyclophamide.
[00632]
Further therapeutic agents that can be combined with a subject compound can be
found in
Goodman and Gilman's "The Pharmacological Basis of Therapeutics" Tenth Edition
edited by Hardman, Limbird
and Gilman or the Physician's Desk Reference, both of which are incorporated
herein by reference in their entirety.
[00633]
The compounds described herein can be used in combination with the agents
provided herein
or other suitable agents, depending on the condition being treated. Hence, in
some embodiments, the compounds as
provided herein will be co-administered with other agents as described above.
When used in combination therapy,
the compounds described herein can be administered with the second agent
simultaneously or separately. This
administration in combination can include simultaneous administration of the
two agents in the same dosage form,
simultaneous administration in separate dosage forms, and separate
administration. That is, a compound described
herein and any of the agents described above can be formulated together in the
same dosage form and administered
simultaneously. Alternatively, a compound as provided herein and any of the
agents described above can be
simultaneously administered, wherein both the agents are present in separate
formulations. In another alternative, a
compound as provided herein can be administered just followed by and any of
the agents described above, or vice
versa. In the separate administration protocol, a compound as provided herein
and any of the agents described
above can be administered a few minutes apart, or a few hours apart, or a few
days apart.
[00634]
Administration of the compounds as provided herein can be effected by any
method that
enables delivery of the compounds to the site of action. An effective amount
of a compound as provided herein can
be administered in either single or multiple doses by any of the accepted
modes of administration of agents having
similar utilities, including rectal, buccal, intranasal and transdermal
routes, by intra-arterial injection, intravenously,
intraperitoneally, parenterally, intramuscularly, subcutaneously, orally,
topically, as an inhalant, or via an
impregnated or coated device such as a stent, for example, or an artery-
inserted cylindrical polymer.
160

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00635]
When a compound as provided herein is administered in a pharmaceutical
composition that
comprises one or more agents, and the agent has a shorter half-life than the
compound as provided herein, unit dose
forms of the agent and the compound as provided herein can be adjusted
accordingly.
[00636]
The examples and preparations provided below further illustrate and exemplify
the compounds
as disclosed herein and methods of preparing such compounds. It is to be
understood that the scope of the present
disclosure is not limited in any way by the scope of the following examples
and preparations. In the following
examples molecules with a single chiral center, unless otherwise noted, exist
as a racemic mixture. Those molecules
with two or more chiral centers, unless otherwise noted, exist as a racemic
mixture of diastereomers. Single
enantiomers/diastereomers can be obtained by methods known to those skilled in
the art.
EXAMPLES
Chemical Examples
[00637]
The chemical entities described herein can be synthesized according to one or
more illustrative
schemes herein and/or techniques well known in the art.
[00638]
Unless specified to the contrary, the reactions described herein take place at
atmospheric
pressure, generally within a temperature range from -10 C to 200 C. Further,
except as otherwise specified,
reaction times and conditions are intended to be approximate, e.g., taking
place at about atmospheric pressure within
a temperature range of about -10 C to about 110 C over a period that is, for
example, about 1 to about 24 hours;
reactions left to run overnight in some embodiments can average a period of
about 16 hours.
[00639]
The terms "solvent," "organic solvent," or "inert solvent" each mean a solvent
inert under the
conditions of the reaction being described in conjunction therewith including,
for example, benzene, toluene,
acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF"), chloroform,
methylene chloride (or
dichloromethane), diethyl ether, methanol, N-methylpyrrolidone ("NMP"),
pyridine and the like. Unless specified
to the contrary, the solvents used in the reactions described herein are inert
organic solvents. Unless specified to the
contrary, for each gram of the limiting reagent, one cc (or mL) of solvent
constitutes a volume equivalent.
[00640]
Isolation and purification of the chemical entities and intermediates
described herein can be
effected, if desired, by any suitable separation or purification procedure
such as, for example, filtration, extraction,
crystallization, column chromatography, thin-layer chromatography or thick-
layer chromatography, or a
combination of these procedures. Specific illustrations of suitable separation
and isolation procedures are given by
reference to the examples hereinbelow. However, other equivalent separation or
isolation procedures can also be
used.
[00641]
When desired, the (R)- and (S)-isomers of the non-limiting exemplary
compounds, if present,
can be resolved by methods known to those skilled in the art, for example by
formation of diastereoisomeric salts or
complexes which can be separated, for example, by crystallization; via
formation of diastereoisomeric derivatives
which can be separated, for example, by crystallization, gas-liquid or liquid
chromatography; selective reaction of
161

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
one enantiomer with an enantiomer-specific reagent, for example enzymatic
oxidation or reduction, followed by
separation of the modified and unmodified enantiomers; or gas-liquid or liquid
chromatography in a chiral
environment, for example on a chiral support, such as silica with a bound
chiral ligand or in the presence of a chiral
solvent. Alternatively, a specific enantiomer can be synthesized by asymmetric
synthesis using optically active
reagents, substrates, catalysts or solvents, or by converting one enantiomer
to the other by asymmetric
transformation.
[00642] The compounds described herein can be optionally contacted with
a pharmaceutically
acceptable acid to form the corresponding acid addition salts. Also, the
compounds described herein can be
optionally contacted with a pharmaceutically acceptable base to form the
corresponding basic addition salts.
[00643] In some embodiments, disclosed compounds can generally be
synthesized by an appropriate
combination of generally well known synthetic methods. Techniques useful in
synthesizing these chemical entities
are both readily apparent and accessible to those of skill in the relevant
art, based on the instant disclosure. Many of
the optionally substituted starting compounds and other reactants are
commercially available, e.g., from Aldrich
Chemical Company (Milwaukee, WI) or can be readily prepared by those skilled
in the art using commonly
employed synthetic methodology.
[00644] The discussion below is offered to illustrate certain of the
diverse methods available for use in
making the disclosed compounds and is not intended to limit the scope of
reactions or reaction sequences that can be
used in preparing the compounds provided herein.
General Synthetic Methods
[00645] The compounds herein being generally described, it will be more
readily understood by
reference to the following examples, which are included merely for purposes of
illustration of certain aspects and
embodiments, and are not intended.
(i) General methods
for the synthesis of Cl-Wd and Ts0-Wd heterocycles:
0 OH CI
HCONH2 Oxalyl chloride
I
NH2 130-180 C
A-1 A-2 A-3
Method A:
[00646] General
conditions for the preparation of 4-chloropyrido[3,2-d]pyrimidine:
[00647] 3-Aminopicolinic acid (A-1) (30 g, 217 mmol, 1.0 eq) is suspended
in formamide (75 mL, 1.88
mol, 8.66 eq). The resulting mixture is stirred at 140 C for 1 hand at 170 C
for 1 h and then at 180 C for an
additional 1 h. The mixture is cooled to RT and filtered. The filter cake is
washed with water, and dried in vacuo to
afford the product, pyrido[3,2-d]pyrimidin-4-ol (A-2).
[00648] To a stirred solution of pyrido[3,2-d]pyrimidin-4-ol (A-2) (16 g,
109 mmol, 1.0 eq) and DMF (0.4
mL) in DCM (200 mL) at RT, oxalyl chloride (23.2 mL, 272 mmol, 2.5 eq) is
added dropwise (over 5 min) and the
162

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
resulting mixture is stirred at reflux overnight. The mixture is allowed to
cool to RT and concentrated in vacuo. The
residue is diluted with water (200 mL), neutralized with saturated aqueous
NaHCO3 solution below 10 C to adjust
the pH to 8-9 and then extracted with ethyl acetate (4 x 100 mL). The combined
organic layers are washed with
brine, dried over Na2SO4 and filtered. The filtrate is concentrated in vacuo.
The residue is purified by flash column
chromatography on silica gel (16-50 % ethyl acetate-petro ether) to afford the
product, 4-chloropyrido[3,2-
d]pyrimidine (A-3).
0 OH CI
!N)LOH
NH2
NOH NCI
A-1 B-1 B-2
Method B:
[00649] General method for synthesis of 2,4-dichloropyrido[3,2-
d]pyrimidines:
[00650] 3-Aminopicolinic acid (A-1) (1.0 g, 7.24 mmol, 1.0 eq), potassium
cyanate (2.94 g, 36.2 mmol,
5.0 eq) and ammonium chloride (3.87 g, 72.4 mmol, 10.0 eq) are suspended in
water (40 mL). The slurry is heated
to 160 C and mixed manually for 2 h as water vapour is expelled from the
reaction vessel. The reaction temperature
is then increased to 200 C and stirred for 40 min. After cooling to 90 C,
hot water is added and then the mixture is
allowed to cool to RT. The mixture is acidified with concentrated HC1 to
adjust pH to 3-4. The precipitate is
collected by filtration, rinsed with water (2 x 10 mL) and dried in vacuo to
afford the product, pyrido[3,2-
d]pyrimidine-2,4-diol (B-1).
[00651] A mixture of pyrido[3,2-d]pyrimidine-2,4-diol (B-1) (360 mg, 2.21
mmol, 1.0 eq) and PC15 (1.84
g, 8.83 mmol, 8.0 eq) in POC13 (20 mL) is stirred at reflux for 8 h. An
additional amount of PC15 (1.84 g, 8.83 mmol,
8.0 eq) and POC13 (20 mL) are added, and the resulting mixture is stirred at
reflux for 16 h. After cooling to RT, the
mixture is concentrated in vacuo and the residue is poured into ice-water. The
mixture is neutralized with saturated
Na2CO3 to adjust the pH to 8-9, and then extracted with DCM (3 x 20 mL). The
combined organic layers are washed
with brine, dried over Na2SO4 and filtered. The filtrate is concentrated in
vacuo and the residue is purified by
column chromatography on silica gel (10-12% ethyl acetate/petro ether) to
afford the product, 2,4-
dichloropyrido[3,2-d]pyrimidine (B-2).
/=(CO2Et
Et0 CN Et0 C OH CI
2
C-2 \CN NCN NCN
I I -
NH2
C-1 C-3 C-4 C-5
Method C:
[00652] General method for synthesis of 4-chloro-1,5-naphthyridine-3-
carbonitriles:
[00653] A mixture of pyridin-3-amine (C-1) (10.0 g, 106.3 mmol, 1.0 eq)
and (E)-ethyl 2-cyano-3-
ethoxyacrylate (C-2) (21.5 g, 127 mmol, 1.2 eq) in toluene (40 mL) is stirred
at reflux under argon for 5 h and then
163

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
allowed to cool to RT. The precipitate is collected by filtration, rinsed with
a mixture of petroether and ethyl acetate
(v/v = 2:1, 3 x 10 mL), and then dried in vacuo to afford the product, ethyl 2-
cyano-3-(pyridin-3-ylamino)acrylate
(C-3).
[00654] The mixture of ethyl 2-cyano-3-(pyridin-3-ylamino)acrylate (C-3)
(2.0 g, 9.26 mmol, 1.0 eq) in
Dowtherm A (48 mL) is stirred at 260 C for 5 h. The mixture is cooled to RT
and poured into a mixture of Me0H
and DCM (v/v = 1:2, 20 mL). The precipitate is filtered, rinsed with DCM, and
dried in vacuo to afford the product,
4-hydroxy-1,5-naphthyridine-3-carbonitrile (C-4).
[00655] A mixture of 4-hydroxy-1,5-naphthyridine-3-carbonitrile (C-4) (2.0
g, 11.7 mmol, 1.0 eq) in
phosphoroxychloride (40 mL, 424 mol, 36.2 eq) is stirred at reflux for 3 h.
The mixture is cooled to RT and
concentrated in vacuo. The residue is poured into ice-water (50 mL) and
neutralized with saturated aqueous
NaHCO3 solution to adjust the pH to 8 while keeping the temperature below 5
C. The mixture is extracted with
DCM (3 x 50 mL). The combined organic layer is washed with brine, dried over
Na2SO4 and filtered. The filtrate is
concentrated in vacuo and the residue is purified by flash column
chromatography on silica gel (1.25% Me0H-
DCM) to afford the product, 4-chloro-1,5-naphthyridine-3-carbonitrile (C-5).
14F12 MCPBA conc. HCI
_______________________________________ H2N
DCM 1, 4-dioxane Hc
C-1 D-1
0
0
CH(OMe)3
D-1 0
0)L0 ____________________________________________
0 0 1 0
N,
DIEA
Reflux 0 0
isopropanol 0
0-2 0-3 0-4
HOAc
CI
Dowtherm A
Fe powder POCI 1
3
260 C AcOH / Pyridine
0 OH HOAc 011
0-5 0-6 0-7
Method D
[00656] General method for synthesis of 4-chloro-1,5-naphthyridines:
[00657] To a mixture of pyridin-3-amine (C-1) (10.0 g, 106.3 mmol, 1.0 eq)
in DCM (100 mL) at 0 C, m-
chloroperoxybenzoic acid (22.0 g, 128 mmol, 1.2 eq) is added in portions (over
15 min). The resulting mixture is
allowed to warm to RT and then stirred at RT for 2 h. The reaction is quenched
with water (100 mL). The organic
phase is separated and the aqueous layer is extracted with DCM (2 x 100 mL).
The resulting aqueous phase is
neutralized with concentrated HC1 to adjust the pH to 2-3. To this mixture,
1,4-dioxane (100 mL) is added and the
resulting mixture is concentrated in vacuo to afford the product, 3-
aminopyridine -1-oxide hydrochloride (D-1).
164

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00658] A mixture of 2,2-dimethy1-1,3-dioxane-4,6-dione (D-2) (10.0 g, 70
mmol, 1.0 eq) in trimethyl
orthoformate (100 mL, 365 mmol, 5.0 eq) is stirred at reflux for 3 h. The
resulting mixture is cooled to RT and
concentrated in vacuo. The residue is triturated with diethyl ether and then
filtered. The filter cake is washed with
cooled diethyl ether and dried in vacuo to afford the product, 5-
(methoxymethylene)-2,2-dimethy1-1,3-dioxane-4,6-
dione (D-3).
[00659] To a mixture of 5-(methoxymethylene)-2,2-dimethy1-1,3-dioxane-4,6-
dione (D-3) (7.6 g, 41.0
mmol, 1.0 eq) and 3-aminopyridine -1-oxide hydrochloride (D-1) (6.0 g, 41.0
mmol, 1.0 eq) in isopropanol (20
mL), N,N-diisopropylethylamine (7.2 mL, 53 mmol, 1.3 eq) is added and the
resulting mixture is stirred at 90 C for
20 min. The mixture is allowed to cool to RT and then poured into ice-water
(50 mL). The precipitate is collected by
filtration, rinsed with isopropanol (2 x 10 mL) and diethyl ether (2 x 10 mL),
and then dried in vacuo to afford the
product, 2,2-dimethy1-5-((pyridin-1-oxide 3-ylamino)methylene)-1,3-dioxane-4,6-
dione (D-4).
[00660] The mixture of 2,2-dimethy1-5-((pyridin-1-oxide 3-
ylamino)methylene)-1,3-dioxane-4,6-dione (D-
4) (5.1 g, 19.3 mmol, 1.0 eq) in Dowtherm A (48 mL) is stirred at 260 C for 5
- 10 min. The mixture is allowed to
cool to RT and then diluted with petroleum ether. The precipitate is filtered,
rinsed with petroleum ether, and dried
in vacuo to afford the product, 1,5-naphthyridin-5-oxide 4-ol (D-5).
[00661] To a solution of 1,5-naphthyridin-5-oxide 4-ol (D-5) (330 mg, 2.0
mmol, 1.0 eq) in acetic acid
(12 mL) and pyridine (2.5 mL) at RT, iron powder (456 mg, 8.15 mmol, 4.0 eq)
is added in portions (over 10 min)
and the resulting mixture is stirred for 2 h. The mixture is filtered and the
solid is rinsed with acetic acid. The
filtrate is concentrated in vacuo. The residue is triturated with acetone and
then filtered. The filter cake is rinsed
with acetone and dried in vacuo to afford the product, 1,5-naphthyridin-4-ol
acetic acid salt (D-6).
[00662] A mixture of 1,5-naphthyridin- 4-ol acetic acid salt (D-6) (800
mg, 3.0 mmol, 1.0 eq) in
phosphoroxychloride (5 mL, 53 mol, 17.6 eq) is stirred at 110 C for 30 min,
cooled to RT and then concentrated in
vacuo. The residue is poured into ice-water (50 mL) and neutralized with
saturated aqueous NaHCO3 solution to
adjust the pH to 8-9 while keeping the temperature below 5 C. The mixture is
stirred at RT for 30 min and then
extracted with DCM (3 x 40 mL). The combined organic layers are washed with
brine, dried over Na2SO4 and
filtered. The filtrate is concentrated in vacuo and the residue is purified by
flash column chromatography on silica
gel (1-2% Me0H-DCM) to afford the product, 4-chloro-1,5-naphthyridine (D-7).
-N
HCI
0
HCI(g )/ Et0H NH 0 NaHCO3( aq.) H2N
NCAO 0
CHCI3 ethanol
Z-1
0 Cl
N-N POCI3
I
H2N N H2N N
Z-2 Z-3
Method Z
165

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00663] General method for synthesis of 7-chloropyrazolo[1,5-a]pyrimidin-5-
amine:
[00664] A solution of ethyl 2-cyanoacetate (100 mL, 94 mmol) in chloroform
(100 mL) and ethanol (6.5
mL) is bubbled with HC1 at -10 for 4 h. The reaction mixture is allowed to
warm to RT slowly and then stirred at
RT overnight. The mixture is concentrated in vacuo to remove the solvent. The
residue is suspended in diethyl
ether (100 mL) and stirred for 30 min. The resulting solid is collected by
filtration and riseded with diethyl ether to
afford ethyl 3-ethoxy-3-iminopropanoate hydrochloride (Z-1) as a white solid.
[00665] Sodium bicarbonate (3.9 g, 46.2 mmol) is added in small portions
to a mixture of ethyl 3-ethoxy-
3-iminopropanoate hydrochloride (Z-1) (8.6 g, 44.0 mmol) in crushed ice (100
g) and ethyl acetate (50 mL) until the
pH value reaches 8-9. The organic layer is separated and the aqueous layer was
extracted with ethyl acetate (2 x 50
mL). The combined organic layers are dried over anhydrous Mg504 and filtered.
The filtrate is concentrated in
vacuo to afford a colourless oil which is added to a solution of 1H-pyrazol-5-
amine (3.65 g, 44.0 mmol) in ethanol
(100 mL). The resulting mixture is then degassed and backfilled with argon
(three cycles) and stirred at reflux
overnight. The mixture is then filtered while it was still warm. The solid is
rinsed with cold ethanol (25 mL) and
ether (2 x 25 mL), and then dried in vacuo to afford 5-aminopyrazolo[1,5-
a]pyrimidin-7(4H)-one (Z-2).
[00666] A solution of 5-aminopyrazolo[1,5-a]pyrimidin-7(4H)-one (Z-2) (1.5
g, 10 mmol) in phosphoryl
chloride (45 mL) is stirred at reflux overnight. The reaction mixture is
allowed to cool to RT and concentrated in
vacuo. The residue is then dissolved in ice-water (25 mL) and the resulting
mixture is neutralized with aqueous
NaOH solution (20%) to adjust the pH to 9 ¨ 10. The mixture is extracted with
ethyl acetate (3 x 50 mL) after
which the combined organic layers are washed with brine (500 mL), dried over
anhydrous Na2504 and filtered. The
filtrate is concentrated in vacuo to afford the crude product which is
slurried in a mixture of DCM/MTBE (3 mL/15
mL). The resulting solid is then collected by filtration to afford 7-
chloropyrazolo[1,5-a]pyrimidin-5-amine (Z-3).
0 CI 0 CI 0 NH2
EtO) NH4OH(N EtON EtON
N CI 1,4-dioxane NLNH2CI
Hinge-1
Method N
[00667] General method for synthesis of 2-amino-4-chloropyrimidine-5-
carboxylate:
[00668] To a stirred solution of ethyl 2,4-dichloropyrimidine-5-
carboxylate (1.6 g, 7.24 mmol) in 1,4-
dioxane (15 mL) at 0-5 C under argon, ammonium hydroxide (28-30%, 2.56 mL,
21.72 mmol) is added slowly and
the resulting mixture is stirred from 0 C to RT for 2 h. The reaction is
complete based on TLC (30% EA/Hex, Rf =
0.3 for ethyl 2-amino-4-chloropyrimidine-5-carboxylate (Hinge-1); Rf = 0.5 for
ethyl 4-amino-2-chloropyrimidine-
5-carboxylate) and LC-MS analysis. The mixture is partitioned between ethyl
acetate and water. The organic layer
is washed with brine, dried over Na2504 and filtered. The filtrate is mixed
with silica gel and then concentrated in
vacuo. The residue is loaded to the top of silica gel packed in a glass column
and eluted with 0-100% ethyl
acetate/hexanes to afford ethyl 2-amino-4-chloropyrimidine-5-carboxylate
(Hinge-1).
[00669] The other regio isomer, ethyl 4-amino-2-chloropyrimidine-5-
carboxylate, is also isolated.
166

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0"--\
ci CI OH CI
0¨\
N N H2SO4(con.) POCI3
N
Et3N, Et0H Nr 65-70 0C
CI
Reflux
A-1 A-2 A-3 A-4
Method 0
[00670] General method for synthesis of 5-chloroimidazo[1,2-c]pyrimidine:
[00671] The mixture of 2,4-dichloropyrimidine (A-1) (20 g, 134 mmol), 3,3-
diethoxypropan-1-amine (19.6
g, 147 mmol) and Et3N (20.5 mL, 147 mmol) in ethanol (250 mL) is stirred at RT
for 24 h. The mixture is
concentrated in vacuo and the residue is purified by flash column
chromatography to afford the 2-chloro-N-(2,2-
diethoxyethyl)pyrimidin-4-amine (A-2).
[00672] A mixture of 2-chloro-N-(2,2-diethoxyethyl)pyrimidin-4-amine (A-2)
(20 g, 81.3 mmol) and
H2SO4 (conc, 12 mL, 220 mmol) is stirred at 65-70 C for 2 h. The reaction
mixture is allowed to cool to RT,
poured into ice-water (50 mL) and then basified with cold aqueous NaOH
solution (10%) to adjust the pH to 1-3
while keeping inner temperature between 0-20 C. The precipitate is collected
by filtration and dried in vacuo to
afford imidazo[1,2-f]pyrimidin-5-ol (A-3).
[00673] A mixture of imidazo[1,2-f]pyrimidin-5-ol (A-3) (4 g , 29.6 mmol)
in phosphoroxychloride (50
mL, 530 mmol) is stirred at reflux overnight. The mixture is allowed to cool
to RT and concentrated in vacuo to
remove the phosphoroxychloride. The residue is poured into ice water (30 mL),
and then neutralized with saturated
aqueous NaHCO3 solution to adjust the pH to 6 while keeping the temperature
below 5 C. The mixture is stirred at
RT for 30 min and then extracted with ethyl acetate (20 mL x 3). The combined
organic layer is washed with brine,
dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo and the
residue was purified by flash column
chromatography on silica gel (1-5 % ethyl acetate-petroleum ether) to afford 5-
chloroimidazo[1,2-f]pyrimidine (A-
4).
0 0 0 CI
).LOH r)..LNH2 r).LI NH rLN
N NNH2 N N
NH2
B-1 B-2 B-3 B-4
Method P
[00674] General method for synthesis of 4-chloropyrido[3,4-d]pyrimidine:
[00675] The mixture of 3-aminoisonicotinic acid (B-1) (7 g, 50 mmol) and
DMF (0.2 mL) in thionyl
chloride (100 mL) is stirred at reflux for 2 h. The mixture is then
concentrated in vacuo. The residue is dissolved in
THF (30 mL) and the resulting solution (solution A) is used directly in the
next step.
167

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00676] To a stirred mixture of ammonium hydroxide (100 mL), the above
solution A (30 mL) is added
dropwise while the reaction temperature is maintained between 25 C to 30 C
using an ice-water bath. The resulting
mixture is stirred at RT for 2 h and then water (100 mL) is added. The organic
layer is separated, washed with water
(100 mL x 2), dried over Na2SO4 and filtered. The filtrate is concentrated in
vacuo to afford the amide (B-2).
[00677] The mixture of compound B-2 (2 g, 14 mmol) in triethoxy methane
(50 mL) is stirred at reflux
overnight. The mixture is allowed to cool to RT and filtered. The filter cake
is washed with water, and dried in
vacuo to afford compound (B-3).
[00678] To a stirred solution (B-3) (1 g, 6.7 mmol) and DMF (0.1 mL) in
CH3CN (20 mL) at RT, POC13
(10 mL) is added and the resulting mixture is stirred at reflux overnight. The
mixture is allowed to cool to RT and
concentrated in vacuo. The residue is diluted with water (30 mL), neutralized
with saturated aqueous NaHCO3
solution below 10 C to adjust the PH to 8-9 and then extracted with ethyl
acetate (100 mL x 4). The combined
organic layer is washed with brine, dried over Na2SO4 and filtered. The
filtrate is concentrated in vacuo. The
residue is purified by flash column chromatography on silica gel (16-50 %
ethyl acetate-petro ether) to afford
compound (B-4).
HON CI, õN
H2N_e_N
19
C-1 C-2 C-3
Method Q
[00679] General method for synthesis of 5-chloropyrazolo[1,5-a]pyrimidine:
[00680] To a stirred mixture of 1H-pyrazol-5-amine (4 g, 48 mmol, 1 eq) in
1,4-dioxane (20 mL), ethyl
propiolate (5.08 g. 52 mmol, 1.06 eq) is added dropwise and the resulting
mixture is stirred at reflux for 4 h. The
mixture is cooled to RT and filtered. The filter cake is washed with water,
and dried in vacuo to afford compound
(C-2).
[00681] The solution of (C-2) (1.35 g, 10 mmol) and DMF (0.1 mL) in POC13
(10 mL) is stirred at reflux
overnight. The mixture is allowed to cool to RT and concentrated in vacuo. The
residue is diluted with water (30
mL), neutralized with saturated aqueous NaHCO3 solution below 10 C to adjust
the PH to 8-9 and then extracted
with ethyl acetate (30 mL x 2). The combined organic layer is washed with
brine, dried over Na2SO4 and filtered.
The filtrate is concentrated in vacuo. The residue is purified by flash column
chromatography on silica gel (16-50 %
ethyl acetate-petro ether) to afford compound (C-3).
General method for the synthesis of amine cores:
168

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
Ri Ri
;\OH (C0C H2N¨R21)2, DMF (Cat.)
RT, DCM NEt3 , RT
I
E-1 E-2
Ri 0
BocHN N.0 conc. HCI
\N" R2
Me0H, reflux
"BuLi, iPrMgCI
-50 C - -20 C, THF
E-3 K1H2
Method E:
[00682] General conditions for the preparation of (S)-3-(1-aminoethyl)-
isoquinolin-1(2H)-ones:
[00683] To a stirred mixture of a given o-methylbenzoic acid (E-1) (1.5
mol, 1 eq) and DMF (2 mL) in
DCM (1275 mL) at RT, oxalyl chloride (1.65 mol, 1.1 eq) is added over 5 min
and the resulting mixture is stirred at
RT for 2 h. The mixture is then concentrated in vacuo. The residue is
dissolved in DCM (150 mL) and the resulting
solution (solution A) is used directly in the next step.
[00684] To a stirred mixture of aniline (1.58 mol, 1.05 eq) and
triethylamine (3.15 mol, 2.1 eq) in DCM
(1350 mL), the above solution A (150 mL) is added dropwise while the reaction
temperature is maintained between
25 C to 40 C by an ice-water bath. The resulting mixture is stirred at RT for
2 h and then water (1000 mL) is added.
The organic layer is separated, washed with water (2 x 1000 mL), dried over
Na2SO4 and filtered. The filtrate is
concentrated in vacuo. The product is suspended in n-heptane (1000 mL) and
stirred at RT for 30 min. The
precipitate is collected by filtration, rinsed with heptanes (500 mL) and
further dried in vacuo to afford the amide
(E-2).
[00685] To a stirred mixture of amide (E-2) (173 mmol, 1 eq) in anhydrous
THF (250 mL) at -30 C under
an argon atmosphere, a solution of n-butyllithium in hexanes (432 mol, 2.5 eq)
is added dropwise over 30 min while
keeping inner temperature between -30 C and -10 C. The resulting mixture is
then stirred at -30 C for 30 min.
[00686] To a stirred mixture of (S)-tert-butyl 1-(methoxy(methyl)amino)-1-
oxopropan-2- ylcarbamate (260
mmol, 1.5 eq) in anhydrous THF (250 mL) at -30 C under an argon atmosphere, a
solution of isopropylmagnesium
chloride in THF (286 mmol, 1.65 eq) is added dropwise over 30 min while
keeping inner temperature between -30
C and -10 C. The resulting mixture is stirred at -30 C for 30 min. This
solution is then slowly added to above
reaction mixture while keeping the inner temperature between -30 C and -10
C. The resulting mixture is stirred at -
15 C for 1 h. The reaction mixture is quenched with water (50 mL) and then
acidified with conc. HC1 at -10 C - 0
C to adjust the pH to 1-3. The mixture is allowed to warm to RT and
concentrated in vacuo. The residue is
dissolved in Me0H (480 mL), and then conc. HC1 (240 mL) is added quickly at
RT. The resulting mixture is stirred
at reflux for 1 h. The reaction mixture is concentrated in vacuo to reduce the
volume to about 450 mL. The residue is
extracted with a 2:1 mixture of heptane and ethyl acetate (2 x 500 mL). The
aqueous layer is basified with
concentrated ammonium hydroxide to adjust the pH to 9-10 while keeping the
inner temperature between -10 C and
0 C. The mixture is then extracted with DCM (3 x 300 mL), washed with brine,
dried over MgSO4 and filtered. The
169

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
filtrate is concentrated in vacuo and the residue is dissolved in Me0H (1200
mL) at RT. To this solution, D- (-) -
tartaric acid (21g, 140 mmol, 0.8 eq) is added in one portion at RT. After
stirring at RT for 30 min, white solid
precipitates out and the mixture is slurried at RT for 10 h. The solid is
collected by filtration and rinsed with Me0H
(3 x 50 mL). The collected solid is suspended in water (500 mL) and then
neutralized with concentrated ammonium
hydroxide solution at RT to adjust the pH to 9-10. The mixture is extracted
with DCM (3 x 200 mL). The combined
organic layers are washed with brine, dried over MgSO4 and filtered. The
filtrate is concentrated in vacuo to afford
(S)-3-(1-aminoethyl)-isoquinolin-1(2H)-ones (E-3).
0
0
R1 R1
0 CbzHN N.0 '\.)LN-R2 acid, IPAc f\N-R2
_____________________________________________________ )1.
'BuLi, iPrMgCI
F1H2
0
NHCbz E-3'
E-2'
Method E':
[00687] General conditions for the preparation of (S)-3-(1-aminoethyl)-
isoquinolin-1(2H)-ones:
[00688] In one embodiment, an amino compound (E-3') may be prepared
following Method E',
wherein the intermediate (E-1') may be prepared following procedures known in
the art or the procedure as
described in Method E. In one embodiment, intermediate (E-2') may be prepared
from intermediate (E-1') by
contacting intermediate (E-1') with a base (e.g., nBuLi) followed by (S)-
benzyl (1-(methoxy(methyl)amino)-1-
oxopropan-2-yl)carbamate. In one embodiment, the amino compound (E-3') may be
prepared from intermediate
(E-2') by cyclizing intermediate (E-2') in the presence of an acid. In one
embodiment, the acid is H2SO4. In
another embodiment, the acid is HC1. In one embodiment, the amount of the acid
is about 1 to 20 equivalents
relative to the amount of the intermediate (E-2'). In one embodiment, the acid
is about 5 equivalents of H2SO4. In
one embodiment, the cyclization occurs at about room temperature to 65 C.
[00689] In one embodiment, the cyclization provides the amino compound (E-
3') with a ratio of (S)-
enantiomer to (R)-enantiomer of about 1:1 to 20:1. In one embodiment, the
cyclization provides the amino
compound (E-3') with a ratio of (S)-enantiomer to (R)-enantiomer of about 1:1
to 10:1. In one embodiment, the
cyclization provides the amino compound (E-3') with a ratio of (S)-enantiomer
to (R)-enantiomer of about 1:1 to
4:1. It is to be understood that the methods provided herein are also suitable
for the preparation of (R)-enantiomer
of the amino compound (E-3') when (R)-benzyl (1-(methoxy(methyl)amino)-1-
oxopropan-2-yl)carbamate is used in
place of (S)-benzyl (1-(methoxy(methyl)amino)-1-oxopropan-2-yl)carbamate.
170

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 2 0 NH2
0 NH C0
CIA CI NaHS HO OC2H5
0 CI
______________________________________________________ . 1------
______________________________ ¨
pyridine / THF 0e0Et Et0H/H20 S
F-1 F-2 F-3
CO0C2H5 CO0C2H5
Tp HgO/AcOH
POCI3 CI
ozone
I \ I \ _____________ .
H HO
DMF S DMF S HCI / H20
0 0
F-4 F-5 R2
R2 0
CO0C2H5 COOH 0
CI NaOH/H20 C11.3_____
(C0C1)2 (I)_NH2
\c____:1H
\CS¨ _____________________ .- I \ ___________________ N __ CI
S S DCM pyridine I \
F-6 F-7 S
F-8
R2
CI 101
I. n-BuLi / THF
con. HCI N
2. i-Pr-MgCI
____________________ ¨ - S-
0 Me0H z
Y
NH
2 F-9
Boc'NH 1
Method F:
[00690] General conditions for the preparation of (S)-6-(1-aminoethyl)-3-
chloro-5-phenylthieno[3,2-
c]pyridin-4(5H)-ones:
[00691] To a solution of ethyl-3-aminocrotonate (F-1) (64.6 g, 0.5 mol,
1.0 eq) and pyridine (44.5 mL,
0.55 mol, 1.1 eq) in THF (600 mL) at -20 C, 2-chloroacetyl chloride (59.3 g,
0.53 mol, 1.05 eq) is added dropwise
within 1 h. The resulting mixture is stirred from -20 C to RT for an
additional 2 h and then it is poured into H20
(1200 mL). The mixture is extracted with ethyl acetate (3 x 1200 mL). The
combined organic layer is washed with
H20 (2 x 500 mL) and brine (500 mL), dried over anhydrous Na2SO4 and filtered.
The filtrate is concentrated in
vacuo to reduce the volume to about 200 mL. The residue is slurried in
petroether (400 mL) at RT for 30 min. The
solid is collected by filtration, rinsed with petroether (200 mL), and dried
in vacuo to afford the product, (E)-ethyl 3-
amino-2-(2-chloroacetyl) but-2-enoate (F-2).
[00692] To a suspension of (E)-ethyl 3-amino-2-(2-chloroacetyl) but-2-
enoate (F-2) (58.6 g, 0.29 mol, 1.0
eq) in Et0H (500 mL) at RT, 30% NaSH solution (190 g, 1.018 mol, 3.6 eq) is
added dropwise over 30 min and the
resulting mixture is stirred at RT for 3 h. The mixture is poured into H20
(1.5 L) and stirred for 30 min. The
precipitate is collected by filtration and rinsed with H20 (3 x 100 mL). The
collected solid is re-dissolved in ethyl
acetate (500 mL), dried over anhydrous Na2SO4 and filtered. The filtrate is
concentrated in vacuo to afford the
product, ethyl 4-hydroxy-2-methylthiophene-3-carboxylate (F-3).
[00693] To a stirred solution of ethyl 4-hydroxy-2-methylthiophene-3-
carboxylate (F-3) (20.0 g, 0.11 mol,
1.0 eq) in DMF (150 mL) at -20 C under argon, phosphoryl chloride (49.4 g,
0.32 mol, 3.0 eq) is added dropwise
171

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
while keeping the reaction temperature below 10 C. The resulting mixture is
stirred at RT for 30 min and then at 80
C for 1 h. The reaction mixture is cooled to RT and poured into ice-water (800
mL). The resulting mixture is
neutralized with Na0Ac to adjust the pH to 7 - 8. The precipitate is collected
by filtration, rinsed with H20 (150
mL), and dried in vacuo. The product is purified by flash column
chromatography on silica gel (5% ethyl acetate-
petroether) to afford the product, ethyl 4-chloro-5-formy1-2-methylthiophene-3-
carboxylate (F-4).
[00694] To a mixture of ethyl 4-chloro-5-formy1-2-methylthiophene-3-
carboxylate (F-4) (9.72 g, 41.77
mmol, 1.0 eq) in DMF (150 mL), Oxone (77.1 g, 125.3 mmol, 3.0 eq) is added in
portions and the resulting mixture
is stirred at RT overnight. The mixture is poured into H20 (800 mL) and
stirred for 30 min. The precipitate is
collected by filtration, rinsed with H20 (100 mL) and then slurried in H20 (80
mL) for 30 min. The solid is collected
by filtration, rinsed with H20 (3 x 60 mL) and petroether (3 x 60 mL), and
dried in vacuo to afford the product, 3-
chloro-4-(ethoxycarbony1)-5-methylthiophene-2-carboxylic acid (F-5).
[00695] To a stirred solution of 3-chloro-4-(ethoxycarbony1)-5-
methylthiophene-2-carboxylic acid (F-5)
(7.79 g, 31.3 mmol, 1.0 eq) in AcOH (150 mL) at RT, Hg0 (7.46 g, 34.4 mmol,
1.1 eq) is added and the resulting
mixture is stirred at reflux for 1 h. The reaction mixture is cooled to 50 C -
60 C and then aqueous HC1 (750 mL,
2.4 M, 1.8 mol) is added. The resulting mixture is stirred at reflux for an
additional 1 h. The mixture is allowed to
cool to RT and extracted with methyl tert-butyl ether (3 x 200 mL). The
combined organic layers are washed with
sat. NaHCO3 solution (3 x 200 mL), H20 (200 mL) and brine H20 (200 mL)
sequentially. The organic layer is dried
over anhydrous Na2SO4 and filtered. The filtrate is concentrated in vacuo and
the residue is purified by flash column
chromatography on silica gel (2% ethyl acetate-petroether) to afford the
product, ethyl 4-chloro-2-methylthiophene-
3-carboxylate (F-6).
[00696] To a mixture of ethyl 4-chloro-2-methylthiophene-3-carboxylate (F-
6) (4.98 g, 24.3 mmol, 1.0 eq)
in Et0H (50 mL) ar RT, aqueous NaOH solution (20 %, 14.6 g, 73.0 mmol, 3.0 eq)
is added and the resulting
mixture is stirred at reflux for 1 h. The mixture is allowed to cool to RT and
concentrated in vacuo. The residue is
poured into ice water (30 mL) and neutralized with concentrated HC1 to adjust
the pH to 1-2 while keeping the
temperature below 5 C. The precipitate is collected by filtration, rinsed
with H20 (80 mL) and dried in vacuo to
afford the product, 4-chloro-2-methylthiophene-3-carboxylic acid (F-7).
[00697] To a stirred mixture of 4-chloro-2-methylthiophene-3-carboxylic
acid (F-7) (6.8 mmol, 1.0 eq)
and DMF (three drops) in DCM (25 mL), oxalyl chloride (20.4 mmol, 3.0 eq) is
added at 0-5 C over 5 min. The
resulting mixture is stirred from 0 C to RT overnight. The mixture is
concentrated in vacuo. The residue is
dissolved in DCM (20 mL) and the resulting solution (solution C) is used
directly in the next step.
[00698] To a stirred mixture of a given (R3-substituted)phenylamine (7.47
mmol, 1.1 eq.) and pyridine
(20.4 mmol, 3.0 eq.) in DCM (10 mL) at 0-5 C, solution C is added and the
resulting mixture is stirred at RT for 2
h. The mixture is diluted with DCM (150 mL), washed with aqueous HC1 (1 M, 3 x
50 mL), H20 (2 x 50 mL) and
brine (2 x 50 mL) sequentially. The organic layer is dried over anhydrous
Na2SO4 and filtered. The filtrate is
concentrated in vacuo to afford the amide (F-8).
172

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00699] To a mixture of amide (F-8) (2.0 mmol, 1.0 eq.) in THF (3.5mL) at -
60 C under argon, n-
butyllithium solution (2.5 M in hexanes, 2.8 mL, 7.0 mmol, 3.5 eq.) is added
slowly while keeping the temperature
below -50 C. The resulting mixture is stirred below -50 C for 30 min to form
solution D.
[00700] To a stirred mixture of (S)-tert-butyl 1-(methoxy(methyl)amino)-1-
oxopropan-2-ylcarbamate (2.6
mmol, 1.3 eq) in anhydrous THF (3.0 mL) at -60 C under argon, a solution of
isopropylmagnesium chloride in THF
(2.0 M, 1.33 mL, 2.66 mmol, 1.33 eq.) is added dropwise over 30 min while
keeping inner temperature below -50
C. The resulting mixture is stirred below -50 C for an additional 30 min and
then it is added slowly to solution D
while keeping inner temperature at -50 C. The resulting mixture is stirred
between -50 C and -40 C for 1 h and
then at RT for an additional 1 h. The reaction mixture is quenched with water
(5.0 mL) and acidified with aqueous
HC1 (5 N) at 0 C to adjust the pH to 5-6. The mixture is concentrated in
vacuo. The residue is dissolved in Me0H
(3.0 mL) and concentrated HC1 (1.5 mL) is added. The resulting mixture is
stirred at reflux for 1 h and then
concentrated in vacuo to remove Me0H. The residue is suspended in H20 (5.0 mL)
and extracted with a 1:1 mixture
of heptane and ethyl acetate (3 x 20 mL). The aqueous layer is basified with
saturated aqueous NH4OH to adjust the
pH to 8 ¨ 9 and extracted with DCM (3 x 20 mL). The combined organic layers
are washed with brine, dried over
anhydrous Na2SO4 and filtered. The filtrate is concentrated in vacuo and the
residue is purified by flash column
chromatography on silica gel (10% Me0H-DCM) to afford the product (F-9).
0 / 1. POCI3 0 / \ siii
¨0 DMF, THF
____________________________ . _-0 mCPBA
__________________________________________________________________ INI ' 0
H2N 2. 2M NaHS H2N µS--
-
S
G-1 G-2 G-3
0 1. nBuLi (2.5 eq) 0 1. (C0C1)2
NaOH ____________________ Y -78 C / THF (OH N DCM
/ DMF
(1 OH _________________________________________________________________ II.
THF:MeOH:H20 'S ,S N)
OH

/ THF I 2. H2NR2 (1.5 eq)
(2:1:1) DIEA/THF
G-4 G-5
0 0
0 R2
=\
CI )LOEt N/ y
I N'
,.R SnBu3 / N1R2
N 1 H µs¨i 0Et
N 2 PP
_____________________________________________________ 1w
'S
S Pd(h4)3 NaH / DMF
/ 0
I /
G-6 G-7 G-8
173

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 \
0s04 / Na104 R2 CS2CO3N"R2 NaBH4
OEt Nss---r0Et
0 0 0
G-9 G-10
\ \ \
TPAP / NMO
4A MS powder N-R2 MeMgCI
0 OH
G-11
G-12 G-13
0
\
PPh3 / DPPA / DIAD R2 Pd/C / H2 INV R2
N
_________________ = Isiss
NH2
N3
G-14 G-15
Method G:
[00701] General conditions for the preparation of 6-(1-aminoethyl)-3-
methyl-isothiazolo[4,5-c]pyridin-
4(5H)-ones:
[00702] To a solution of methyl 3-aminocrotonate (G-1) (10.0 g. 86.9 mmol)
in anhydrous THF (200 mL)
at 0 C, a solution of phosphoryl chloride (12.0 mL. 95.6 mmol) in anhydrous
DMF (28 mL) is added dropwise
(over 10 min). The resulting mixture is stirred at 0 C for 1 h and then
stirred at 30 C for 4 h. The mixture is
allowed to stand overnight in a refrigerator. Chilled ether (800 mL) is then
added to the reaction mixture until a
semiclear/oil residue is formed. The yellow ether layer is decanted. The oil
residue is then dissolved in DCM (500
mL) and washed with NaHS aqueous solution (2.0 M). The organic layer is washed
with H20 (4 x 500 mL), dried
over Na2SO4 and filtered. The filtrate is concentrated in vacuo to afford the
product, methyl 3-amino-2-
thioformylbut-2-enoate (G-2).
[00703] To a solution of methyl 3-amino-2-thioformylbut-2-enoate (G-2)
(5.34 g, 33.5 mmol) in Et0H
(250 mL), a solution of meta-chloroperoxybenzoic acid (70-75%, 12.4 g, 50.3
mmol) in Et0H (150 mL) is added
and the resulting mixture is stirred at reflux for 3 h. The mixture is allowed
to cool to RT, quenched with saturated
aqueous NaOH solution and then extracted with ethyl acetate (2 x 200 mL). The
combined organic layers are
washed with brine, dried over Na2SO4 and filtered. The filtrate is
concentrated in vacuo to afford the product, 3-
methylisothiazole-4-carboxylate (G-3).
[00704] To a solution of 3-methylisothiazole-4-carboxylate (G-3) (4.73 g,
30.1 mmol) in THF-Me0H-H20
(2:1:1, 50 mL), NaOH (3.61 g, 90.3 mmol) is added and the resulting mixture is
stirred at 40 C for 16 h. The
mixture is allowed to cool to RT and then acidified with concentrated HC1 to
adjust the pH to 2-3. The precipitate is
174

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
collected by filtration, rinsed with water and dried in vacuo to afford the
product, 3-methylisothiazole-4-carboxylic
acid (G-4).
[00705] To a solution of 3-methylisothiazole-4-carboxylic acid (G-4) (3.9
g, 27.3 mmol) in anhydrous
THF (150 mL) at -78 C under argon, n-butyl lithium solution (27.3 mL, 68.3
mmol) is added dropwise and the
resulting mixture is stirred at -78 C for 1 h. To this mixture, a solution of
iodide (13.9 g, 54.6 mmol) in THF (50
mL) is added slowly and the resulting mixture is stirred at RT for 1 h. The
mixture is acidified with concentrated
HC1 to adjust the pH to 3-4, and then extracted with ethyl acetate. The
organic layer is washed with aqueous Na2S03
solution. The aqueous layer is extracted with ethyl acetate. The combined
organic layers are washed with brine,
dried over Na2SO4 and filtered. The filtrate is concentrated in vacuo to
afford the product, 5-iodo-3-
methylisothiazole-4-carboxylic acid (G-5).
[00706] To a solution of 5-iodo-3-methylisothiazole-4-carboxylic acid (G-
5) (4.45 g, 16.5 mmol) and
DMF (3 drops) in anhydrous DCM (60 mL), oxalyl chloride solution (2.0 M in
DCM, 16.5 mL, 33.1 mmol) is added
dropwise and the resulting mixture is stirred at RT for 2 h. The reaction
mixture is concentrated in vacuo to afford
the acyl chloride intermediate as an oil. The intermediate is dissolved in
anhydrous THF (100 mL). To this mixture,
a given amine R2NH2 (24.8 mmol) and N,N-diisopropylethylamine (4.09 mL, 24.8
mmol) are added dropwise. The
resulting mixture is stirred at RT for 1 h. The reaction mixture is quenched
with water and extracted with ethyl
acetate. The combined organic layers are washed with brine, dried over Na2SO4
and filtered. The filtrate is
concentrated in vacuo to afford the amide (G-6).
[00707] To a solution of amide (G-6) (18.6 mmol) and tributyl(vinyl)fin
(8,19 mL, 27.9 mmol) in DMF
(30 mL) under argon, Pd(PPh3)4 (1.07 g, 0.93 mmol) is added. The resulting
mixture is stirred at 90 C for 2 h. The
mixture is allowed to cool to RT, quenched with water and extracted with ethyl
acetate (2 x 300 mL). The combined
organic layers are washed with brine, dried over Na2SO4 and filtered. The
filtrate is concentrated in vacuo and the
residue is purified by flash column chromatography on silica gel (0-25% ethyl
acetate-hexanes) to afford the product
(G-7).
[00708] To a solution of (G-7)in anhydrous DMF (70 mL) at RT, sodium
hydride (60% in mineral oil,
1.52 g, 37.9 mmol) is added in portions and the resulting mixture is stirred
at RT for 45 min. To this mixture, ethyl
chloroacetate (4.73 mL, 44.2 mmol) is added dropwise and the resulting mixture
is stirred for 2 h. The reaction
mixture is quenched with water and extracted with ethyl acetate. The combined
organic layers are washed with
brine, dried over Na2SO4 and filtered. The filtrate is concentrated in vacuo
to afford the product (G-8). The product
obtained is used in the next step without purification.
[00709] To a solution of (G-8) (12.62 mmol) in 1,4-dioxane-H20 (3:1, 100
mL) at RT, osmium tetraoxide
(4% wt in H20, 1.0 mL, 0.13 mmol) is added and the resulting mixture is
stirred for 30 min. To this mixture, sodium
periodate (5.40 g, 25.24 mmol) is added and the resulting mixture is stirred
at RT for 3 h. The mixture is filtered
through celite and the filtrate is extracted with ethyl acetate (2 x 100 mL).
The combined organic layers are washed
with brine, dried over Na2SO4 and filtered. The filtrate is concentrated in
vacuo to afford the product aldehyde (G-
9). The product obtained is used in the next step without purification.
175

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00710] To a solution of (G-9) (12.68 mmol) in a mixture of Et0H and ethyl
acetate (3:1, 200 mL), cesium
carbonate (4.55 g, 13.95 mmol) is added and the resulting mixture is stirred
at 50 C for 2 h. The mixture is allowed
to cool to RT and concentrated in vacuo. The residue is partitioned between
water and ethyl acetate. The organic
layer is washed with brine, dried over Na2SO4 and filtered. The filtrate is
concentrated in vacuo and the residue is
purified by flash column chromatography on silica gel (0-40% ethyl acetate-
hexanes) to afford the product (G-10).
[00711] To a solution of (G-10) (2.48 mmol) in anhydrous Me0H (15 mL),
NaBH4 (936 mg, 24.75 mmol)
is added in portions and the resulting mixture is stirred at RT for 16 h. The
mixture is partitioned between water and
ethyl acetate. The organic layer is washed with brine, dried over Na2SO4 and
filtered. The filtrate is concentrated in
vacuo to afford product alcohol (G-11).
[00712] To a solution of (G-11) (5.21 mmol) in anhydrous DCM (100 mL), 4A
molecular sieves
(powder, 2.84 G), NMO (N-methylmorpholine-N-oxide) (1.22 g, 10.43 mmol) and
TPAP (tetrapropylammonium
perruthenate) (92 mg, 0.26 mmol) are added sequentially. The resulting mixture
is stirred at RT for 1 h and then
filtered through a celite/silica gel pad. The filtrate is concentrated in
vacuo to afford aldehyde (G-12).
[00713] To a solution of (G-12) (1.05 g, 3.90 mmol) in anhydrous THF (100
mL) at -78 C under argon,
methylmagnesium chloride solution (3.0M in THF, 3.25 mL, 9.75 mmol) is added
dropwise and the resulting
mixture is stirred from -78 C to RT for 2 h. The mixture is quenched with
water (50 mL) and extracted with ethyl
acetate (2 x 100 mL). The combined organic layers are washed with brine, dried
over Na2SO4 and filtered. The
filtrate is concentrated in vacuo to afford product (G-13).
[00714] To a solution of (G-13) (0.48 mmol) in anhydrous THF (8 mL) at 0
C under argon, triphenyl
phosphine (230 mg, 0.88 mmol) is added and the resulting mixture is stirred
for 5 min. To this mixture, diphenyl
phosphoryl azide (0.24 mL, 1.12 mmol) is added followed by slow addition of
diisopropyl azodicarboxylate (0.17
mL, 0.88 mmol) over a 20 min period of time. The resulting mixture is stirred
from 0 C to RT for 2 h. The mixture
is then partitioned between ethyl acetate and water. The organic layer is
washed with brine, dried over Na2SO4 and
filtered. The filtrate is concentrated in vacuo and the residue is purified by
ISCO (silica gel cartridge, 0-50% ethyl
acetate-hexanes) to afford the product azide (G-14).
[00715] A mixture of azide (G-14) (0.2489 mmol) and palladium (10% weight
on carbon, 23 mg, 20% of
starting material by weight) in anhydrous Me0H (5 mL) is degassed and flushed
with hydrogen (three cycles). The
reaction mixture is stirred under a hydrogen atmosphere at RT for 1 h. The
mixture is filtered through celite and
rinsed with ethyl acetate. The filtrate is then concentrated in vacuo to
afford the amine (G-15).
176

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0'
R1 CO2Et
0 ).Ri R1 CN Me2N )e ¨( HBr-AcOH
NC _________________ r )¨K
COOEt Et0H / refluxa- CN _______ _
=)-0Et Piperidine (cat.) /
AcOH / 55 C
AcOH/reflux Me2N
H-1 H-2 H-3
(Z/E isomer not determined)
R1 R1 R1
COOH COCI R2-NH2
>i COOEt NaOH aq Oxalyl chloride
1,4-dioxane &N Br DMF/DCM __ w I
NBr TEA/THF 1
NBr 110 C
H-4 H-5 H-6
CI
R1 0 R1 0 yo R1 0
Sn(Bu)3
_________________________ L' > ).1s1"
R2 OEt 0s04/Na104 >)LN,R2
H ' I H
&N Br Pd(PPh3)4 N NaH/DMF ID 1,4-dioxane-H20
N
DMF/90 C I I OEt
H-7 H-8 H-9
R1 0 R1 0 R1 0
R2 < CS2CO3 N NaBH4 - R2 TPAP/NMO /0*N-
R2
I1 1 _________________ 3.
,. yo tNr0Et
N Me0H NOH 4A MS/DCM
I
0 OEt 0
H-10 H-11 H-12
R1 0 R1 0
Ri 0
R2 MeMgCI ).14R2 " PPh3/DPPA/DIAD OLNIR2"
IN" I _____________________ . I
THF THF
N
OH N3
H-14 H-15
H-13
R1 0
H2 / Pd-C R2
0.N1"
Me0H - I
ley
NH2
H-16
Method H
[00716] General conditions for the preparation of 7-(1-aminoethy1-6-pheny1-
1,6-naphthyridin-5(6H)-ones:
[00717] To a mixture of ethyl 2-cyanoacetate (H-1) (45.2 g, 400 mmol) and a
given ketone It1C(0)Me
(800 mmol) in glacial acetic acid (50 mL), piperidine (2 mL, 20 mmol) is added
and the resulting mixture stirred at
reflux for 24 h. The reaction mixture is allowed to cool to RT, and then
concentrated in vacuo. The residue is diluted
with water (200 mL) and extracted with ethyl acetate (3 x 200 mL). The
combined organic layers are washed with
177

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
brine (50 mL), dried over Na2SO4 and filtered. The filtrate is concentrated in
vacuo and the residue is purified by
flash column chromatography on silica gel (0-2% ethyl acetate-petroether) to
afford the product (11-2).
[00718] To a solution of (11-2) (285 mol) in absolute Et0H (300 mL), N,N-
dimethylformamide dimethyl
acetal (37.3 g, 313 mmol) is added dropwise and the resulting mixture is
stirred at reflux for 6 h. The mixture is
allowed to cool to RT, and concentrated in vacuo to afford the product (II-3).
This material is used in the next step
without further purification.
[00719] Dienoate (II-3) (148 mmol) is dissolved in AcOH (120 mL) and the
mixture is stirred at 40 C. A
solution of 45% HBr-AcOH (120 mL) is added dropwise, and then the mixture is
stirred at 55 C for 2 h. The
mixture is allowed to cool to RT, poured into ice-water, neutralized with
solid Na2CO3, and then extracted with ethyl
acetate (3 x 150 mL). The combined organic layer is washed with brine (50 mL),
dried over Na2SO4 and filtered.
The filtrate is concentrated in vacuo and the residue is purified by flash
column chromatography on silica gel (5-
20% ethyl acetate-petroether) to afford the product (II-4).
[00720] To a solution of 4-substituted ethyl 2-bromolnicotinate (II-4) (52
mmol) in 1,4-dioxane (15 mL), a
solution of NaOH (8.0 g, 200 mmol) in H20 (15 mL) is added and the resulting
mixture is stirred at reflux for 12 h.
The mixture is allowed to cool to RT, diluted with H20, and washed with ethyl
acetate (3 x 30 mL). The aqueous
layer is acidified with concentrated hydrochloric acid to adjust the pH to 1,
and then extracted with ethyl acetate (3 x
50 mL). The combined organic layers are washed with brine (25 mL), dried over
Na2SO4 and filtered. The filtrate is
concentrated in vacuo to afford the product nicotinic acid (II-5).
[00721] Compound (II-5) is then converted to (II-16) in analogous fashion
to (G-15) in Method G.
General conditions for attachment of Wd substituents:
00
Wd-CI Ri
N R2
j\ N R2
or
H2
Wd-OTs
1-1HN.Wd
N
E-3
Method I
[00722] A mixture of compound (E-3) (1.0 mmol, 1.0 eq), Wd-Cl or Wd-OTs
(1.50 mmol, 1.5 eq) and
triethylamine (3.0 mmol, 3.0 eq) in n-BuOH (5 mL) is stirred at reflux for 1-5
h. The reaction mixture is allowed to
cool to RT and then concentrated in vacuo. The residue is purified by flash
column chromatography on silica gel
(eluting with a mixture solvent of Me0H and DCM) to afford the product (I-1).
The reaction can occur under other
conditions known in the art that are suitable for SNAr displacement reaction.
In one embodiment, the reaction
solvent is NMP.
(iv) General conditions for substitution of Wd:
178

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 0
R ii
iR 1kAN,R2
A
H IN N C N NH2
I I
N NN
J-1 J-2
Method J
[00723] Quinolinone (J-1) (0.11 mmol, 1.0 eq) is suspended in ammonium
hydroxide (10-35% solution,
20 mL) in a sealed tube and the resulting mixture is stirred at 140 C
overnight. The mixture is allowed to cool to
RT and then extracted with DCM (3 x 15 mL). The combined organic layers are
washed with brine, dried over
Na2SO4 and filtered. The filtrate is concentrated in vacuo and the residue is
purified by column chromatography on
silica gel (1-2% Me0H/DCM) to afford compound (J-2) as the product.
0
0 0 BocHN)-L
Ri Ri . OH
1\)"Lsoii SOCl2 / DMF(cat) R2-NH2 \N-R2 SOCl2 / DMF(cat)
I -1"" I
NO2 Toluene EtN3, RT NO2 Toluene DCM, DIPEA
K-1
Reflux K-2 Reflux
0 0
0
Zn,AcOH HCI / Me0H (2N)
,R2 R 2
I
NO2
NHBoc FIHBoc FIF12
K-3 K-4 K-5
Method K:
[00724] To a stirred mixture of nitrobenzoic acid (K-1) (1.0 mol, 1.0 eq)
and DMF (2.0 mL) in toluene
(800 mL), thionyl chloride (292 mL, 1.0 mol, 4.0 eq) is added dropwise (over
15 min) and the resulting mixture is
stirred at reflux for 1.5 h. The mixture is allowed to cool to RT and then
concentrated in vacuo. The residue is
dissolved in DCM (100 mL) to form solution A, which is used directly in the
next step.
[00725] To a stirred mixture of a given amine R2-NH2 (102.4 g, 1.1 mol,
1.1 eq) and triethylamine (280
mL, 2.0 mol, 2.0 eq) in DCM (700 mL), solution A is added dropwise while
keeping the reaction temperature below
C. The resulting mixture is allowed to warm to RT and then stirred at RT
overnight. The reaction mixture is
diluted with ice-water (1.0 L) and stirred for 15 min. The precipitate is
collected by filtration, rinsed with isopropyl
ether (3 x 100 mL) and petroleum ether (3 x 100 mL), and then dried in vacuo
to afford product amide (K-2).
[00726] A mixture of nitro- benzamide (K-2) (20.0 mmol, 1.0 eq) and DMF
(cat.) in toluene (60 mL) at
RT, thionyl chloride (12 mL, 164 mmol, 8.2 eq) is added dropwise (over 5 min)
and the resulting mixture is stirred
at reflux for 2 h. The mixture is allowed to cool to RT and then concentrated
in vacuo. The residue is dissolved in
DCM (10 mL) to form solution B, which is used directly in the next step.
179

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00727] To a stirred mixture of N-(tert-butoxycarbony1)-L-alanine (16.0
mmol, 0.8 eq) and N,N-
diisopropylethylamine (4.0 g, 31.0 mol, 1.5 eq) in DCM (20 mL), solution B is
added dropwise while keeping the
reaction temperature between 0-10 C. The resulting mixture is stirred at this
temperature for 1 h and then stirred at
RT overnight. The reaction mixture is quenched with ice-water (100 mL). The
organic layer is separated and the
aqueous layer is extracted with DCM (2 x 80 mL). The combined organic layers
are washed with brine, dried over
Na2SO4 and filtered. The filtrate is concentrated in vacuo and the residue is
slurried in isopropyl ether (100 mL) for
15 min. The solid is collected by filtration and dried in vacuo to afford
product (K-3).
[00728] To a suspension of zinc dust (7.2 g, 110 mmol, 10.0 eq) in glacial
acetic acid (40 mL) at 15 C, a
solution of (K-3) (11.0 mmmol, 1.0 eq) in glacial acetic acid (40 mL) is added
and the resulting mixture is stirred at
RT for 4 h. The mixture is poured into ice-water (200 mL) and neutralized with
saturated aqueous NaHCO3 solution
to adjust the pH to 8. The resulting mixture is extracted with DCM (3 x 150
mL). The combined organic layers are
washed with brine, dried over Na2SO4 and filtered. The filtrate is
concentrated in vacuo and the residue is purified
by flash chromatography on silica gel (7% ethyl acetate-petroleum ether) to
afford product (K-4).
[00729] Compound (K-4) (0.5 mmol, 1.0 eq) is dissolved in hydrochloric
methanol solution (2N, 20 mL)
and the resulting mixture is stirred at RT for 2 h. The mixture is
concentrated in vacuo. The residue is diluted with
water (30 mL) and then neutralized with saturated aqueous NaHCO3 to adjust the
pH to 8 while keeping the
temperature below 5 C. The resulting mixture is extracted with DCM (3 x 30
mL). The combined organic layers
are washed with brine, dried over Na2SO4 and filtered. The filtrate is
concentrated in vacuo and the residue is
slurried in petroleum ether (10 mL). The solid is collected by filtration and
dried in vacuo to afford product (K-5).
[00730] The quinazolinone (K-5) can be used to synthesize compounds
described herein using, for
example, Method I to couple the amine to Wd groups.
0
BocHNJ-N
L ,Me CI 0
CI 0
CI 0
- OMe OH
= 0
OH
L-2
Ac20
70 C
iPrMgCI o NHBoc
L-1 nHexLi NHBoc L-4
isobutyric acid L-3
THF H2N-R2
AlMe3
DCM
CI 0
CI 0
HCI
110 ,R2 i-PrOHR2
ref lux
Fi 2 0 .
NHBoc
L-6 L-5
Method L:
180

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00731] To 2-chloro-6-methylbenzoic acid (L-1) (8.00 g, 46.9 mmol) in a
dry round bottom flask under N2
is added 50 mL of dry THF. The resulting mixture is cooled to -25 C. Then, n-
hexyllithium (88 mL, 202 mmol)
(2.3 M in hexanes) is added, and the reaction is stirred at -20 C for about
20 min.
[00732] To compound (L-2) (14.16 g, 61.0 mmol) in a second dry round
bottom flask under N2 is added 70
mL of dry THF. The mixture is cooled to about -10 C. To the cold mixture is
slowly added isopropyl magnesium
chloride (63.3 ml, 2 M, 127 mmol). The resulting mixture is then stirred at -
10 C for about 20 min. Then, this
mixture is slowly cannulated drop wise into the flask containing the (L-1)
reaction while maintaining the
temperature at -20 C. After addition is complete, the reaction is slowly
warmed to RT and stirred at RT for about
1.6 hours. The reaction mixture is then cooled to -10 C and quickly
cannulated to another flask containing 15 mL
of ethyl acetate and 10 mL of isobutyric acid at -10 C under N2. After
stirring for about 5 minutes, 10 mL of water
is rapidly added. The cooling bath is removed, and the reaction mixture is
stirred for 10 minutes at RT. The mixture
is transferred to a separation funnel, and water (200 mL) is added. The water
layer is extracted with Et0Ac (3 X 400
mL). The aqueous layer is then acidified with HC1 (2M) to pH 3, and is
extracted with Et0Ac (3 X 500 mL), dried
over sodium sulfate and concentrated in vacuo. The resulting material is
purified by silica gel column
chromatography using 0 ¨ 10% Me0H in DCM to afford benzoic acid (L-3).
[00733] A mixture of benzoic acid (L-3) (5.00 g, 14.63 mmol) in acetic
anhydride (10 mL) is stirred in a
round bottom flask at 70 C for about 2.5 hours. Then, the remaining acetic
anhydride is removed in vacuo. The
residue is purified using silica gel column chromatography using Et0Ac/hexanes
to afford lactone (L-4).
[00734] To a mixture of R2-NH2 (197 mg, 1.54 mmol) in 2 mL of DCM is added
A1Me3 (0.772 ml, 1.54
mmol). The mixture is stirred for about 15 min. Then, a solution of lactone (L-
4) (100 mg, 0.309 mmol) in 2 mL of
DCM is added, and the reaction is stirred at RT for about 3 hours. The
reaction mixture is then quenched with
addition of 10 mL of Rochelle's salt and stirring for about 2 hours. The
mixture is diluted with DCM, washed with
brine, dried with Na2SO4 and concentrated in vacuo to afford the amide (L-5)
which is carried directly to the next
reaction.
[00735] To the amide (L-5) in 5 mL of isopropanol is added 3 mL of
concentrated HC1. The reaction is
heated at 65 C for about 3 hours. After cooling to RT, the mixture is
concentrated in vacuo. The solid is
suspended in 15 mL of DCM, followed by the addition of 10 mL of sat. NaHCO3.
This mixture is then stirred at RT
for about 30 min, then 50 mL of DCM is added. The layers are separated and the
organic layer is dried with Na2SO4
and concentrated in vacuo to afford isoquinolinone (L-6).
181

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
0 i) DCM, TFA 0
BocHNJ-LN 3 Me ii) CF3COOEt, TEA, Et0H F COCHN __ N Me
" s. "
z OMe z OMe
L-2 L-2'
Cl 0
Cl 0
Cl 0 g OH
2
L-'
OH Ac20
i) nHexLi 0 , NHCOCF3
L-1
ii) iPrMgCI NHCOCF3 L-4'
iii) citric acid
L-3'
iv) silica plug or crystallize H2N-R2
AlMe3
DCM
Cl 0
Cl 0
HCI
VI-R2
NJ' R2 i-PrOH
reflux
F11-12 0 .
NHCOCF3
L-6 L-5'
Method L':
[00736] General conditions for the preparation of (S)-3-(1-aminoethyl)-
isoquinolin-1(2H)-ones:
[00737] In some embodiments, isoquinolinone (L-6) may be prepared by
following the procedures
exemplified in Method L'. In one embodiment, intermediate (L-2') is prepared
by contacting intermediate (L-2)
with TFA followed by CF3COOEt. In one embodiment, benzoic acid (L-3') is
prepared by contacting o-methyl-
benzoic acid (L-1) with a base (e.g., nHexLi), followed by intermediate (L-
2'), and followed by purification by
silica plug or crystallization. In one embodiment, lactone (L-4') is prepared
by contacting benzoic acid (L-3') with
Ac20 under conditions suitable for cyclization. In one embodiment, amide (L-
5') is prepared by contacting lactone
(L-4') with corresponding amine or aniline in the presence of A1Me3. In one
embodiment, isoquinolinone (L-6) is
prepared by contacting amide (L-5') with an acid under conditions suitable for
cyclization and deprotection. In one
embodiment, the acid is HC1.
[00738] In one embodiment, the cyclization provides the amino compound (L-
6) with a ratio of (S)-
enantiomer to (R)-enantiomer of about 1:1 to 20:1. In one embodiment, the
cyclization provides the amino
compound (L-6) with a ratio of (S)-enantiomer to (R)-enantiomer of about 1:1
to 10:1. In one embodiment, the
cyclization provides the amino compound (L-6) with a ratio of (S)-enantiomer
to (R)-enantiomer of about 1:1 to 4:1.
It is to be understood that the methods provided herein are also suitable for
the preparation of (R)-enantiomer of the
amino compound (L-6) when (R)-enantiomer of intermediate (L-2) is used in
place of (S)-enantiomer of (L-2).
[00739] A compound provided herein may contain one or more chiral
center. Conventional techniques
for the preparation/isolation of individual enantiomers include synthesis from
a suitable optically pure precursor,
asymmetric synthesis from achiral starting materials, or resolution of an
enantiomeric mixture, for example, by
chiral chromatography, recrystallization, resolution, diastereomeric salt
formation, or derivatization into
182

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
diastereomeric adducts followed by separation. In one embodiment, the
enantiopurity of a compound provided
herein may be improved by contacting the compound provided herein with a
suitable solvent or mixture thereof,
followed by filtration. In one embodiment, the solvent is a mixture of water
and dichloromethane. In another
embodiment, the solvent is a mixture of dichloromethane and methanol.
OMe 0
OMe Me
pinacol 1 Mel

0 __________________________ / ____________________ ....
13,
HO' OH
M-1 M-2 M-3
Method M:
[00740] In one embodiment, the N-methyl pyridinone boronic ester (M-3)
may be prepared following
procedures exemplified in Method M. In one embodiment, pyridyl boronic ester
(M-2) may be prepared by
contacting pyridyl boronic acid (M-1) with pinacol under conditions suitable
for the formation of boronic esters. In
one embodiment, the N-methyl pyridinone boronic ester (M-3) may be prepared by
contacting pyridyl boronic ester
(M-2) with Mel under conditions suitable for 0-methyl-to-N-methyl shifting.
Example 1
oi
N N
CI 0 Si
CI o 0 N 0 ;
N A-3 -
3.-
101 / HN N
NH2
NN
1-1 1-2
[00741] Amine I-1 was prepared according to Method E and then coupled to
(A-3) using Method Ito
provide compound 1-2. ESI-MS m/z: 428.0 [M+1-1]+.
Example 2
oi
o 0
N N
0 0 10 ;
N A-3 :
0 - F HN N
F Fl- 112
NN
1-3 1-4
[00742] Amine 1-3 was prepared according to Method E and then coupled to
(A-3) using Method Ito
provide compound 1-4. ESI-MS m/z: 412.2 [M+1-1]+.
183

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 3
CI
N N
0 0
-'
0 40 k I4* 0 ;
F
N A-3 F
1 1 / HN N
NH2
N N
1-5 1-6
[00743] Amine 1-5 was prepared according to Method E and then coupled to
(A-3) using Method I to
provide compound 1-6. ESI-MS m/z: 412.0 [M+El]+.
Example 4
CI 0 CI 0 CI 0
HNO3,H2SO4 0 OH 02N 0 COCICOCI , DCM Et3N, DCM
1110 _1.,.. OH _________
OH and
Aniline
NO2
1-7 1-8 1-8'
CI 0 0
CI 0 Ii

CI 0 N
H2N 0 SnCl2 / DCM CI 0 41)
0 N 02N 0 _______ a 0 N *
H N H
H H
and and
N
NO2 H2
1-9 1-9 1-10 I-10'
CI 0 0 CI 0 0 CI 0
0 0
HBF / NaN0
4 2 0 120 C
____________________________________________________ a 0 11
11 _________________________ a- N
H
NH2 N2BF4 F
1-10 1-11 1-12
CI
N L-'11 CI o 1
CI 0 0
kN N
Method E
N A3 0 /
N
0 /
F HN
yy
_
F NH2 I
N N
,-...--
1-13 1-14
[00744] Intermediate amide 1-12 was prepared from 2-chloro-6-methylbenzoic
acid 1-7 according to the
following procedures:
[00745] To a solution of 2-chloro-6-methylbenzoic acid 1-7 (50 g, 294
mmol, 1.0 eq) in conc. H2SO4 (500
mL) at 0 C, fuming HNO3 (20.4 g, 324 mmol, 1.1 eq) was added slowly. The
resulting mixture was stirred at 0 C
for 5 min and then stirred at RT for 2 h. The reaction mixture was poured into
ice-water (800 mL) and extracted with
ethyl acetate (3 x 500 mL). The combined organic layers were washed with
brine, dried over Na2SO4 and filtered.
184

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
The filtrate was concentrated in vacuo to afford the product as a mixture of
two regio-isomers, 6-chloro-2-methy1-3-
nitrobenzoic acid 1-8 and 6-chloro-2-methyl-5-nitrobenzoic acid 1-8'. The
mixture was used directly in the next step.
[00746] To a stirred solution of above obtained regio-isomer mixture of 6-
chloro-2-methyl-3-nitrobenzoic
acid 8 and 6-chloro-2-methyl-5-nitrobenzoic acid 1-8' (61.8 g, 287 mmol, 1.0
eq) and DMF (catalytic amount) in
DCM (500 mL) at RT, oxalyl chloride (49 mL, 574 mmol, 2.0 eq) was added slowly
(over 5 min). The resulting
mixture was stirred at RT for 2 h and then concentrated in vacuo. The residue
was dissolved in DCM (150 mL) to
form solution A.
[00747] To a mixture of aniline (32 g, 344 mmol, 1.2 eq) and triethylamine
(87.5 g, 861 mmol, 3.0 eq) in
DCM (400 mL) at RT, solution A was added dropwise. The resulting mixture was
stirred for 15 min and then
quenched with water (500 mL). The organic layer was separated and the aqueous
layer was extracted with DCM (3
x 500 mL). The combined organic layers were washed with brine, dried over
Na2SO4 and filtered. The filtrate was
concentrated in vacuo to afford the product as a mixture of two regio-isomers,
6-chloro-2-methy1-3-nitro-N-
phenylbenzamide 1-9 and 2-chloro-6-methyl-3-nitro-N-phenylbenzamide 1-9'. The
mixture was used directly in the
next step.
[00748] To a stirred mixture of 6-chloro-2-methyl-3-nitro-N-
phenylbenzamide 9 and 2-chloro-6-methy1-3-
nitro-N-phenylbenzamide 1-9' (33.6.8 g, 116 mmol, 1.0 eq) in Et0H (400 mL),
SnC12.2H20 (105 g, 464 mmol, 4.0
eq) was added and the resulting mixture was stirred at reflux for 3 h. The
reaction mixture was allowed to cool and
concentrated in vacuo. The residue was poured into water (500 mL) and the
mixture was extracted with ethyl acetate
(5 x 500 mL). The combined organic layers were washed with brine, dried over
Na2SO4 and filtered. The filtrate was
concentrated in vacuo . The residue was purified by flash column
chromatography on silica gel (2-20% ethyl
acetate-petroleum ether) to afford the regio-isomer, 3-amino-6-chloro-2-methyl-
N-phenylbenzamide 1-10. The
remaining mixture of 3-amino-6-chloro-2-methyl-N-phenylbenzamide 1-10 and 5-
amino-6-chloro-2-methyl-N-
phenylbenzamide 1-10' was collected separately. Compound 1-10' was obtained by
further column chromatography
purification.
[00749] To a stirred mixture of 3-amino-6-chloro-2-methyl-N-
phenylbenzamide 1-10 (10.3 g, 39.6 mmol,
1.0 eq) in fluoroboric acid (42%, 72 mL) at 0 C, a solution of sodium nitrate
(5.5 g, 79.2 mmol, 2.0 eq) in H20 (10
mL) was added dropwise over 30 min while keeping the reaction temperature
between 0 C and 5 C. The resulting
mixture was stirred at RT for 16 h. The precipitate was collected by
filtration, rinsed with cold water (2 x 5 mL), and
dried in vacuo to afford the product I-11. The product was used directly in
the next step.
[00750] The intermediate I-11 (12.8 g, 35.7 mmol) was heated to 120 C and
the temperature was
maintained between 120 C and 130 C for 1 h. The mixture was cooled to 50 C
and poured into ice-water (50 mL).
The resulting mixture was extracted with ethyl acetate (3 x 50 mL). The
combined organic layers were washed with
brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo
and the residue was purified by flash
column chromatography on silica gel (2-20% ethyl acetate-hexanes) to afford
the product, 6-chloro-3-fluoro-2-
methyl-N-phenylbenzamide 1-12.
[00751] Amine 1-13 was prepared from amide 1-12 according to Method E and
was then coupled to A-3
using Method Ito provide compound 1-14. ESI-MS m/z: 446.0 [M+H]
185

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 5
ci ci 0 0
N N F
CI 0 00 F CI 0 0 kN 0 ;
H2N 0 N
N A-3 =
HN N
H __________________________________________________ _
_
RFI2 N N
1-10 1-15 1-16
[00752] Amine 1-15 was prepared from amide 1-10' in analogous fashion to
compound 1-13 in Example 4.
Amine 1-15 was then coupled to (A-3) using Method I to provide compound 1-16.
ESI-MS m/z: 446.0 [M+H] '.
Example 6
CI
N l'i o 0
0 0 k F is
N N
/ .
F
10I ; A-3
p.
HN N
- 'i
NH2
NN
1-17 1-18
[00753] Amine 1-17 was prepared according to Method E and was then coupled
to (A-3) using Method Ito
provide compound 1-18. ESI-MS m/z: 412.2 [M+H] '.
Example 7
Ci
N N
CI 0 el
CI 0 0 k ,,, ,
N -- N F
0 /
N F A-3 -
0 / FIR N
_
RH2
NN
1-19 1-20 Q.
[00754] Amine 1-19 was prepared according to Method E and was then coupled
to (A-3) using Method Ito
provide compound 1-20. ESI-MS m/z: 446.0 [M+H] '.
186

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 8
/
N¨N
yN¨N/ ,
,
B
0 /N
________________________________________ ..
FIN N FIN N
i i N
N N N
1-2 1-21
[00755] Compound 1-2 was converted to compound 1-21 according to the
following general procedure:
[00756] Compound 1-2 (0.2 mmol, 1.0 eq), boronic acid (0.41 mmol, 2.0 eq),
Pd(OAc)2 (0.04 mmol, 0.2
eq), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (0.12 mmol, 0.6
eq) and Na2CO3 (0.6 mmol, 3.0 eq)
were dissolved in 1-methy1-2-pyrrolidinone (10 mL). The resulting mixture was
degassed and back-filled with argon
three times, and then stirred at 160 C under an argon atmosphere for 1.5 h.
The mixture was concentrated in vacuo
and the residue was purified by flash column chromatography on silica gel
(1:30, Me0H-DCM) to afford the
product 1-21 as a solid; ESI-MS m/z: 474.2 [M+H]+.
Example 9
CI
N N CI 0 y\
0 0
CI 0 yx N
/ ; A-3 -
a.
FIFJ N
'I N
NH2
N
1-22 1-23
[00757] Amine 1-22 was prepared according to Method E and was then coupled
to (A-3) using Method Ito
provide compound 1-23. ESI-MS m/z: 392.2 [M+H] '.
Example 10
N¨N/
/
V 0
I\
0N /
_
FIR N
NN
1-24
[00758] Compound 1-24 was prepared from compound 1-23 using the analogous
coupling procedures for
the synthesis of compound 1-21 in Example 8. ESI-MS m/z: 438.2 [M+H] '.
187

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 11
/
N-N
/
, 0 0
110 /N F
1-111 N
:0
1-25 N
[00759] Compound 1-25 was prepared from compound 1-20 using the analogous
coupling procedures for
the synthesis of compound 1-21 in Example 8. ESI-MS m/z: 492.2 [M+H] .
Example 12
CI
is
0INI)N ........)CLN 0
\
ke Nsis I /
21---N
N 1 r A-3 i
sS---\\ NH
NH2 N N
L I
1-27
1-26 N
[00760] Amine 1-26 was prepared according to Method G and was then coupled
to (A-3) using Method I to
provide compound 1-27. ESI-MS m/z: 415.2 [M+H] '.
Example 13
CI
N )N
CI\ ii 0
k
CI N
\ ilri el
A-3
.. S
tr li'j
S- HN N
= '1
NH2 N N
1-28 1-29
[00761] Amine 1-28 was prepared according to Method F and was then coupled
to (A-3) using Method Ito
provide compound 1-29. ESI-MS m/z: 434.0 [M+H] '.
188

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 14
N N
S
1-1F1 N
NN
1-30 II
[00762] Compound 1-30 was prepared from compound 1-29 using the analogous
coupling procedures for
the synthesis of compound 1-21 in Example 8. ESI-MS m/z: 480.2 [M+H]
Example 15
CI,NH CI 0
N 2 1.n-BuLl/THF
COOH (C0C1)2N 2.HMPA
a. H 0
DCM DCM/pyridine
N
1-7 1-31 Boo'NH I
CI
N CI 0
CI 0 LNN
Con. HCI NN/
A-3
Me0H HN N
1;1-1-12 II
1-32 1-33 N
[00763] Compound 1-31 was prepared from 1-7 in analogous fashion to E-2 in
Method E and was then
converted to 1-32 according to the following procedure:
[00764] To a mixture of 2-chloro-6-methyl-N-(2-(pyrrolidin-1-
yl)ethyl)benzamide 1-31 (1.33 g, 5.0 mmol,
1.0 eq) and HMPA (0.87 mL, 5.0 mmol, 1.0 eq) in THF (12.5 mL) at -60 C under
argon, n-butyllithium solution
(2.5 M in hexanes, 5.0 mL, 12.5 mmol, 2.5 eq) was added dropwise while keeping
the temperature below -60 C.
The resulting mixture was stirred between -70 C and -60 C for 30 min. To
this mixture, (S)-tert-butyl 1-
(methoxy(methyl)amino)-1-oxopropan-2-ylcarbamate (1.39 g, 6.0 mmol, 1.2 eq)
was added quickly. The resulting
mixture was allowed to slowly warm to RT (over 1 h) and then stirred at RT for
an additional 2 h. The reaction
mixture was cooled to 0 C, quenched with water (5.0 mL), and then acidified
with aqueous HC1 (5 M) to adjust the
pH to 5-6. The mixture was concentrated in vacuo. The residue was suspended in
a mixture of Me0H (20 mL) and
H20 (5.0 mL), concentrated HC1 (10.0 mL) was added and the resulting mixture
was stirred at reflux for 2 h. The
mixture was cooled to -5 C and basified with saturated aqueous NH4OH to
adjust the pH to 8-9. The precipitate was
189

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
filtered off and the filtrate was extracted with DCM (3 x 20 mL). The combined
organic layers were dried over
anhydrous Na2SO4 and filtered. The filtrate was concentrated in vacuo and the
residue was purified by flash column
chromatography on silica gel (1-10% Me0H-DCM) to afford the product (S)-3-(1-
aminoethyl)-8-chloro-2-(2-
(pyrrolidin-1-y1)ethyl)isoquinolin-1(2H)-one 1-32.
[00765] Compound 1-32 was coupled to (A-3) using Method Ito provide
compound 1-33. ESI-MS m/z:
449.2 [M+H]
Example 16
N-N N-N
CI
N-N

N "1 0
ci o o'B'o z
o 401 N
B-2
7
________________________________ 01 ____________ 3
HIN N C
tz.1H2
NH2 NXN
1-1 1-34 1-35
[00766] Compound 1-35 was prepared in two steps from compound I-1.
Compound I-1 was coupled to 1-
methy1-1H-pyrazol-4-ylboronic acid pinacol ester using the procedure described
in Example 8 to provide compound
1-34. Compound 1-34 was then coupled to (B-2) using Method Ito provide
compound 1-35. ESI-MS m/z: 508.2
[M+H]
Example 17
N-N
0 N
1-111rNNH2
NN
1-36 II
[00767] Compound 1-36 was prepared from compound 1-35 using Method J. ESI-
MS m/z: 489.2 [M+H]
Example 18
N-N
0
NF
7
N CI
NN
1-37 II
190

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00768] Compound 1-37 was prepared in analogous fashion to compound 1-35 in
Example 16 except that
amine 1-19 was used in place of amine I-1. ESI-MS m/z: 526.0 [M+H] '.
Example 19
N-N/
0 ; F
HN N NH2
NN
1-38 II
[00769] Compound 1-38 was prepared from compound 1-37 using Method J. ESI-
MS m/z: 507.2 [M+H] '.
Example 20
N-N/
/ Z 0
NA
0 /
41 NY CI
NN
1-39 II
[00770] Compound 1-39 was prepared in analogous fashion to compound 1-35 in
Example 16 except that
amine 1-22 was used in place of amine I-1. ESI-MS m/z: 472.2 [M+H] '.
Example 21
N-N/
/ Z 0
NA
110
HI1 N NH2
1
NN
1-40 II
[00771] Compound 1-40 was prepared from compound 1-39 using Method J. ESI-
MS m/z: 453.2 [M+H] '.
191

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 22
1 1
CI 0 10,
HOOH N*I
/
HN N
N:0 õ HN N
'1
41
1-
1-2 N N
[00772] Compound 1-41 was prepared from compound 1-2 according to the
following coupling
procedures:
[00773] Compound 1-2 (230 umol), boronic acid (690 umol, 3 eq.), sodium
carbonate (1.15 mmol, 5 eq.),
RuPhos (701.tmol, 0.30 eq.), and palladium diacetate (35 umol, 0.15 eq.) were
combined in a 2 mL microwave-
reaction tube with a stir bar which was sealed with a septum. The atmosphere
was purged three times with vacuum,
backfilling with dry argon, then 1,4-dioxane (1.6 mL) and water (0.4 mL) were
added. The reaction was subjected
to microwave heating at 125 C for 3h. The reaction mixture was diluted with
DCM (ca. 30 mL), treated with silica
gel (ca. 1 g) and concentrated; flash chromatography of this residue (on 15 g
silica gel, eluting with a Me0H/DCM
or Et0Ac/hexanes gradient as required, approx. 750 mL total eluent) gave the
product 1-41 as a light-yellow to off-
white powder. ESI-MS m/z: 485.8 [M+H] '.
[00774] The following compounds were also prepared from compound 1-2 using
the analogous coupling
conditions of Example 22:
Example Compound Boronic Acid ESI-MS m/z
23 OMe OMe 502.4
.L
N ' N /I \
1 N N
o ei y
N
H
SI N N
1-42
HO OH
la/
24 mi2 NH 2 486.2
II
1 N N
o 0 10 y
N
H 1 N N B
T , ,
1 Cr.,,
1-43 NN ,
192

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
25 N 499.2
N
I j
7 o a
0N y
H
/ N N1, ,B, ,,
1-44 N)0
26 N OMe MeON 501.1
I
7 0 la
N y
H
0 / N NB
)0, HO' 1-45 N
27 / / 411.4
N-N
/ N-N
7
0
y
N
H
0 / N N
B,
:0 0
N
1-46
28 / / 487.2
N-N
/ N-N
y 0 N 0
H
101 N N,B,
'i vl() 0
NN
1-47
29 OMe OMe 501.24
1 14 )1 Isl
oNsi
0 y
H
/ N N ,B,
HO OH
NN
1-48
Example 30
CI
)NI Cl 0 0
Cl 0 0 I
N
0 ; D-7 /
... ,
. FIN1
F1H2 I
NN
1-1 1-49
193

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00775] Compound I-1 was coupled to (D-7) according to Method I to provide
compound 1-49. ESI-MS
m/z: 427.2 [M+H]
Example 31
CI
CI 0
CI 0 Thsr =N SF
D-7
________________________________________ 3.
HF,1
NN
1-19 1-50
[00776] Compound 1-19 was coupled to (D-7) using Method I to provide
compound 1-50. ESI-MS m/z:
445.2 [M+H]
Example 32
N-N/
0 N
141
1-51 NN
[00777] Compound 1-51 was prepared from compound 1-49 using the analogous
coupling procedures for
the synthesis of compound 1-21 in Example 8. ESI-MS m/z: 473.2 [M+H]
Example 33
CI
f N CI 0 el
CI 0
N*LCI N
B-2 =

FIF1 N CI
H2
NN
1-1 1-52
[00778] Compound 1 was coupled to (B-2) using Method Ito provide compound
52. ESI-MS m/z: 462.07
[M+H]
194

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 34
ci
SN
CI 0
C-5
CN
HNL
NH2 I N
1-1 1-53 Nal
[00779] Compound I-1 was coupled to (C-5) using Method I to provide
compound 1-53. ESI-MS m/z:
452.0 [M+H]
Example 35
CI
NCN CI 0 el
CI 0
C-5 CN
HN
NH2
1-19 1-54 NJN
[00780] Compound 1-19 was coupled to (C-5) using Method Ito provide
compound 1-54. ESI-MS m/z:
470.0 [M+H]
Example 36
N-N
0 N
CN
HL
I N
1-55 Na
[00781] Compound 1-55 was prepared from compound 1-53 using the analogous
coupling procedures for
the synthesis of compound 1-21 in Example 8. ESI-MS m/z: 498.2 [M+H]
Example 37
OMe OMe
CI 0 0, 0 N '1%1 N N
Method J
HO-B N
HN N,C1
HNNH2
N -*11
HN N H2
1-52 1-56
1-57 N
195

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00782] Compound 1-52 was converted to compound 1-56 using Method J.
Compound 1-57 was then
coupled to 2-methoxypyrimidin-5-ylboronic acid using the analogous procedure
in Example 22 to provide
compound 1-57. ESI-MS m/z: 517.25 [M+H] '.
Example 38
Me0
I r%1
/ 0 N ei
0 /
FIll N NH2
1-58 NN
[00783] Compound 1-58 was prepared in analogous fashion as compound 1-57 in
Example 37 except that
5-methoxypyridin-3-ylboronic acid was used in place of 2-methoxypyrimidin-5-
ylboronic acid. ESI-MS m/z: 516.25
[M+H] '.
Example 39
1
N N
I0 N 001
0 /
HNi N NH2
1-59 NN
[00784] Compound 1-59 was prepared in analogous fashion as compound 1-57 in
Example 37 except that
5-methylpyrimidin-5-ylboronic acid was used in place of 2-methoxypyrimidin-5-
ylboronic acid. ESI-MS m/z:
501.24 [M+H] '.
Example 40
HO 0
}21 0 0 0
, 0 0 0 ;
0 ;
0 ; _,..=
,
HN N
FIFI N
_ )0
FIFI N 1%1
NN
1-60 'i 1-61 1-62 N
NN
196

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00785] Compound 1-62 was prepared in 2 steps from compound 1-60. Styrene
1-60 was prepared using
the analogous coupling conditions in Example 22 except that styrylboronic acid
was used in place of 2-methy1-5-
(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)pyrimidine.
[00786] Compound 1-60 (0.98 mmol, 1 eq) was dissolved in THF (10 mL), tert-
butanol (5 mL), and water
(1.5 mL), and treated with N-methyl-morpholine N-oxide (700 mg, 6 eq) and a 4%
aqueous solution of osmium
tetroxide (1254, 0.02 eq). The reaction was stirred at ambient temperature
overnight. The solvent was removed in
vacuo and the residue was taken up in water (50 mL), excess oxidant quenched
with saturated sodium sulfite
solution (20 mL), and the product extracted three times with 40 mL of DCM. The
combined organic phases were
washed with brine and concentrated to a yellow solid.
[00787] This yellow solid was dissolved in THF (25 mL) and water (5 mL),
then treated with sodium
periodate (1 g, 4.7 eq) and allowed to stir at ambient temperature for 1 day.
The reaction mixture was diluted with
DCM, treated with silica gel (2 g) and concentrated in vacuo. Purification of
the product by flash chromatography
on silica gel, eluting with 40% to 80% Et0Ac in hexanes, afforded the aldehyde
1-61.
[00788] Aldehyde 1-61 (0.96 mmol, 1 eq) in THF (13 mL) and tert-butanol (3
mL) was treated with
isoprene (2 mL, 20 eq.), a 2.7M aqueous solution of monobasic sodium phosphate
(2 mL, 5 eq), and a 1M aqueous
solution of sodium chlorite (3 mL, 3 eq). The reaction was stirred at ambient
temperature during 2 days, then treated
with additional 1M chlorite solution (3 mL, 3 eq), and allowed to react
overnight. The reaction mixture was added
to water (90 mL) and DCM (60 mL) and acidified to pH 3 by slow addition of 6M
HC1 (ca. 30 drops). The layers
were separated and the aqueous phase extracted twice with 40 mL DCM. The
combined organic phases were
washed with brine, dried on sodium sulfate, and concentrated. Purification of
the product by flash chromatography
on silica gel, eluting with 1-5% Me0H in DCM containing 1% acetic acid,
afforded the acid 1-62 as an off white
solid. ESI-MS m/z: 436.17 [M+H] '.
Example 41
NHBoc NH2
N NN N
y
CI 0 0 1 ,
HO- 'OH 101 N0
FIF1 N -
N-,c HN N
, N
63 :- ,N
1-2 1 NC
[00789] Compound 1-63 was prepared from compound 1-2 using the analogous
coupling procedures in
Example 22 except that 2-(tert-butoxycarbonylamino)pyrimidin-5-ylboronic acid
was used in place of 2-methy1-5-
(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)pyrimidine. ESI-MS m/z: 487.23
[M+H] '.
Example 42
197

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
CI 0
N
0
ke
N =
A-3
NH
NH2 N-1
1-64 1-65 I
[00790] Amine 1-64 was prepared according to Method H and was then coupled
to (A-3) using Method I to
provide compound 1-65. ESI-MS m/z: 409.2 [M+H]
Example 43
OMe
OMe CI
0
kN 0
NH
0 =
A-3
N NH
N
NH
NC I
NH2
1-66 1-67 1-68
[00791] Amine 1-66 was prepared from (H-1) in analogous fashion to (II-16)
in Method H except that (4-
methoxyphenyl)methanamine was used in place of aniline in step 6 (amide
formation). Amine 1-66 was then
coupled to (A-3) according to Method Ito provide compound 1-67. Compound 1-67
was converted to 1-68
according to the following procedure:
[00792] 6-(4-Methoxybenzy1)-4-methyl-7-(1-(pyrido[3,2-d]pyrimidin-4-
ylamino)ethyl)-1,6-naphthyridin-
5(6H)-one 1-67 (60 mg, 0.13 mmol) was dissolved in trifluoroacetic acid (4 mL)
in a sealed tube and the resulting
mixture was stirred at 140 C for 48 h. The mixture was allowed to cool to RT,
quenched with water and then
neutralized with saturated NaHCO3 aqueous solution. The mixture was extracted
with ethyl acetate, washed with
brine, dried with Na2SO4 and filtered. The filtrate was concentrated in vacuo
to afford the product, 4-methy1-7-(1-
(pyrido[3,2-d]pyrimidin-4-ylamino)ethyl)-1,6-naphthyridin-5(6H)-one 1-68. ESI-
MS m/z: 333.2 [M+H]
Example 44
[00793] The following isoquinolinone compounds (L-6) were made in
analogous fashion to Method L
using the following R2-NH2 reagents.
Table 2. Compounds (L-6)
R2-NH2 Compound (L-6) ESI-MS m/z: [M+11]
CI 0
299.10
H2N
1101
112
198

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
0 CI CI 335
CI 0 .
H2N * ;
=
KIH2
s CN
H2N CN 324.12
0N
=
171 H2
0 Me Me 313.10
CI .H2N 0 N
0 /
=
171 H2
s OMe OMe 329.09
CI 0 .
H2N N
* /
=
171H2
0 OCF3 OCF3 383.13
CI .H2N 0 N
* /
=
FIH2
f1\1 N
CI 0 300.09
I)
H2N N
* /
=
171H2
Example 45
CI
N 0 0
I'l
0 0
k ,õ
N - 0 N F
0N F A-3
/ .
HN N
_
F1H2
NN
1-69
1-70
199

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00794] Amine 1-69 was prepared according to Method E. It was then coupled
to (A-3) using Method I to
provide compound 1-70. ESI-MS mh: 426.2 [M+H] '.
Example 46
CI 0 el CI 0 0
101 /
FIN Ny C 1 ____________ )1.- H Fl N 0
,..,i
N N
N N
1-52 1-71
[00795] To a stirred solution of compound 1-52 (0.1 mmol, 1.0 eq) in THF
(10 mL) at RT, sodium
methoxide (0.2 mmol, 2 eq) was added and the resulting mixture was stirred at
reflux for 1-2 h. The mixture was
allowed to cool to RT and then extracted with DCM (15 mL x 3). The combined
organic layers were washed with
brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo
and the residue was purified by
column chromatography on silica gel (1-2% Me0H/DCM) to afford compound 1-71.
ESI-MS m/z: 458.1 [M+H] '.
Example 47
o
N N
I / 0 N 00
0 /
NH
1-72 N ..= N.k..
I
0 N
[00796] Compound 1-72 was prepared from compound 1-71 in analogous fashion
to compound 1-21 in
Example 8. ESI-MS m/z: 532.2 [M+H] '.
Example 48
o
e
.I
NNV N N .1. c, ,
0
N N N N 0
I

B, I
CI , 0
N F
el 0
/
ei ; F 0 /)--(\
__________________________ 3...
S; F A-3
. .
HN N
AH2 -
NH 2 NN
1-19 I-73a 1-73 II
[00797] Compound 1-73 was prepared in two steps from compound 1-19.
Compound 1-19 was coupled to
2-methoxy-5-(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)pyrimidine using the
procedure described in Example 8
200

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
to provide compound I-73a. Compound I-73a was then coupled to (A-3) using
Method I to provide compound 1-73.
ESI-MS m/z: 520.1 [M+H] '.
Example 49
...--,..
N N
I /
0 N 0
H
0 / N N N H2
i
N N
1-74
[00798] Compound 1-74 was prepared from compound 1-56 using pyrimidin-5-
ylboronic acid in the
Suzuki reaction performed in analogous fashion to compound 1-21 in Example 8.
ESI-MS m/z: 487.2 [M+H] '.
Example 50
N
I
/ 0 N 0
0 /
HI1 N NH2
1
N N
1-75
[00799] Compound 1-75 was prepared from compound 1-56 using (5-
methylpyridin-3-yl)boronic acid in
the Suzuki reaction performed in analogous fashion to compound 1-21 in Example
8. ESI-MS m/z: 500.3 [M+H] '.
Example 51
N
,
1 / 0 N el
0 /
H Il N
i
N N
1-76
[00800] Compound 1-76 was prepared from compound 1-2 using 2-methylpyridin-
4-ylboronic acid in the
Suzuki reaction performed in analogous fashion to compound 1-21 in Example 8.
ESI-MS m/z: 485.1 [M+H] '.
201

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 52
io'
N ' N
CI 0 01 CI 0 I 0 / 0
lei
N [ I ________________ N N =
____________________________________________________ . ,
H IN N C FIR N OBn H BR N 0 n
u 'I 'r :Cr
N IsN N , N
1-52 I-77a I-77b
e
N ' N
I / 0 N so
.
HN N OH
6/;1N
1-77
[00801] To a stirred solution of compound 1-52 (0.1 mmol, 1.0 eq) in THF
(10 mL) at RT, sodium
benzyloxide (0.2 mmol, 2 eq) was added and the resulting mixture was stirred
at reflux for 1-2 h. The mixture was
allowed to cool to RT and then extracted with DCM (15 mL x 3). The combined
organic layers were washed with
brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo
and the residue was purified by
column chromatography on silica gel (1-2% Me0H/DCM) to afford compound I-77a.
ESI-MS m/z: 518.2 [M+H] '.
Example 53
0 0' o
0'.1. .1.
,N)....õN N ' N N ' N
I õI I
N ' N NCI ..-
0 I
-.-
0 ..
0
..... 0 (1
N 'F B-2 N F N F
_________________________________ A. 101
/
. . 0 ...-HIT1 N CI H
FIR N N 2
NH2
N
I
N N
N
I
..."
I-73a I-78a 1-78
[00802] Compound 1-78 was prepared in two steps from compound I-73a.
Compound I-73a was coupled
to (B-2) using Method Ito provide compound I-78a. Compound 1-78 was prepared
from compound I-78a using
Method J. ESI-MS m/z: 535.2 [M+H] '.
202

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 54
cl
N NN
NNq ,
IN I
y N
CI 0 0 I 0 0
,
Sn(Bu)3 0
0 ; 0 B-2 N
__________________________________________________ a- 0
_.... 0 N4 B-2

/ HN N CI
NH2 z
NH2 X:r
,N
1-1 I-79aN
I-79b
N
, N
I /
0 N 00
_____________ x 0 /
HN N NH2
:(7
,N
N
1-79 II
[00803] Compound 1-79 was prepared from compound I-1 in three steps.
Compound I-1 was coupled with
4-(tributylstannyl)pyridazine using the procedure described in Example 8 to
provide compound I-79a. Compound I-
79a was then coupled to (B-2) using Method I to provide compound I-79b.
Compound I-79b was converted to
compound 1-79 using Method J. ESI-MS m/z: 487.2 [M+H]+.
Example 55
CI
N,
N
N 1 N" N NI ,
I
N / NCI /
y
0
el
N F
Sn(Bu)3 0 0 B-2 N F
x 10
¨0" 0 N F ________________________________________________ /
0 /
/ . Hgl N CI
NH2 z
NH2:Cr
,N
N
1-19 I-80a I-80b
N,
1 'N
0 0
N F
_____________ x = /
H NH2
N N 2
uN
1-80
[00804] Compound 1-80 was prepared in three steps from compound 1-19 in
analogous fashion to 1-79 in
Example 54. ESI-MS m/z: 505.2 [M+H] '.
203

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 56
N
,
I /
0 N 0
0 /
FIF1 N NH2
NN
1-81
[00805] Compound 1-81 was prepared from compound 1-56 using 2-
methylpyridin-4-ylboronic acid in the
Suzuki reaction performed in analogous fashion to compound 1-21 in Example 8.
ESI-MS m/z: 500.3 [M+H]'.
Example 57
N
0 /
-
HN N NH2
NN
1-82 II
[00806] Compound 1-82 was prepared from compound 1-56 using quinolin-3-
ylboronic acid in the Suzuki
reaction performed in analogous fashion to compound 1-21 in Example 8. ESI-MS
m/z: 536.3 [M+H] '.
Example 58
HN
\
N
1 / 0 N 0
110 /
FIF1LN NH2
T,
1-83 N
[00807] Compound 1-83 was prepared from compound 1-56 using 5-(4,4,5,5-
tetramethy1-1,3,2-
dioxaborolan-2-y1)-1H-pyrrolo[2,3-b]pyridine in the Suzuki reaction performed
in analogous fashion to compound
1-21 in Example 8. ESI-MS m/z: 525.3 [M+H] '.
204

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 59
1 1
N N J y N N ci 1
N. N
N N 0
B, I
0 i
.--õ,---õ, ....,.,7- N i F
_____________________________ IP
N F N FCI-N
0 , .. _____ . ....- HF1 N CI
-
NH2 Y
NH2
N N
19 I-84a II
J. I-84b
N N
I / 0 el
N F
0 /
_
_______________ 1
FIF1 N NH2
i
NN
1-84
[00808] Compound 1-84 was prepared in two steps from compound 1-19 in
analogous fashion to
compound I-73a in Example 48. Compound I-84a was converted to compound I-84b
in analogous fashion to
compound 1-78 in Example 53. ESI-MS m/z: 519.2 [M+H]+.
Example 60
ci
1 _f_NI
0 0 0
0 0 B I.1
-2 N F N F
x
N F /
0 / FIR N CI
N H2 I Y HNNNH2
N N
NN
1-69
I-85a 1-85
[00809] Compound 1-85 was prepared in two steps from compound 1-69.
Compound 1-69 was coupled to
(B-2) using Method Ito provide compound I-85a. Compound 1-85 was prepared from
compound I-85a using
Method J. ESI-MS m/z: 441.2 [M+H] '.
205

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 61
CI
fIsILN CI 0 40
CI 0 0
CI 0 0
NCI 0 0 rsi 0 N N N .
B-2 N CI N
_
__________________________ . HN -I.
N
HNN NH2
NH2 NN
II NN
II
I-86a I-86b 1-90
e
N N
I / 0 0
0 Nil
______________ a.
N
HNNNH2
1-86 NN
[00810] Compound I-86a was prepared from 2-chloro-6-nitrobenzoic acid
according to MethodK.
Compound I-86a was then coupled to (B-2) using Method I to provide compound I-
86b. Compound I-86b was
converted to compound 1-90 using Method J. Compound 1-90 was coupled to 2-
methoxy-5-(4,4,5,5-tetramethyl-
1,3,2-dioxaborolan-2-yl)pyrimidine using the procedure described in Example 8
to provide compound 1-86. ESI-
MS m/z: 518.2 [M+H]'.
Example 62
\N-N
\
N
0 a
101 N
N
NH
1-87 N 1
N
I
H2N N
[00811] Compound 1-87 was prepared from 1-90 by Suzuki coupling with 1-
methy1-4-(4,4,5,5-tetramethyl-
1,3,2-dioxaborolan-2-y1)-1H-pyrazole in analogous fashion to compound 1-86.
ESI-MS m/z: 490.2 [M+H] '.
206

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 63
lil
I
0 0
0 !II
N'.
FIF1
1-88 NII N
H2N N
[00812] Compound 1-88 was prepared from 1-90 by Suzuki coupling with 2-
methylpyridin-4-ylboronic
acid in analogous fashion to compound 1-86. ESI-MS m/z: 501.2 [M+H] '.
Example 64
N,N
I
0 0
0 !ill
N
111-1
1-89 )N
N 1
I ,
H2N N=
[00813] Compound 1-90 was coupled to 4-(tributylstannyl)pyridazine using
the procedure described in
Example 8 to provide compound 1-89. ESI-MS m/z: 488.2 [M+H]'.
Example 65
1
N N
Y i
B N N
CI 0 0 0-, 0 I /
ifi ,rsii /
o al
N
HI;1 N NH2
iFIN N NH2
NN
NN
1-90 II
1-91
[00814] To a solution of (S)-2-(14(2-aminopyrido[3,2-d]pyrimidin-4-
yl)amino)ethyl)-5-chloro-3-
phenylquinazolin-4(3H)-one (30 mg, 0.067 mmol) and 2-methy1-5-(4,4,5,5-
tetramethy1-1,3,2-dioxaborolan-2-
yl)pyrimidine (22 mg, 0.1 mmol) in 1,4-dioxane-H20 (4:1, 2 mL) in a sealed
tube, 2-dicyclohexylphosphino-2',6'-
diisopropoxybiphenyl (15.6 mg, 0.034 mmol), palladium(II) acetate (3.8 mg,
0.017 mmol), and Na2CO3 (21 mg,
0.20 mmol) were added sequentially. The mixture was degassed and backfilled
with argon (three cycles), and then
stirred at 120 C for 1 h. The mixture was allowed to cool to RT, and then
partitioned between ethyl acetate and
207

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
water. The organic layer was washed with brine, dried over Na2SO4 and
filtered. The filtrate was concentrated in
vacuo and the residue was purified by ISCO column chromatography (silica gel
cartridge, 0-10% Me0H/DCM) to
afford compound 1-91, (S)-2-(14(2-aminopyrido[3,2-d]pyrimidin-4-
yl)amino)ethyl)-5-(2-methylpyrimidin-5-y1)-3-
phenylquinazolin-4(3H)-one (17 mg, 50% yield). ESI-MS m/z: 502.2 [M+H]
Example 66
CI 0
SN
HN NYNH2
N
1-92
[00815] Compound 1-92 was prepared from compound 1-52 using Method J. ESI-
MS m/z: 443.1 [M+H]
Example 67
OMe
OMe NN
NN0
so
H N
CI 0 CI 0 al
,N = OMe N 41r'
OMe HO-13,0H 110
OMe
HNLT H
N N N
HNj=yN,CI 2 HN N
1-52
I-93a NlaN 1-93
[1-.
[00816] Compound 1-93 was prepared in two steps from compound 1-52.
Compound 1-52 was converted
to compound I-93a using 4-methoxybenzylamine in Method J. Compound I-93a was
coupled with 2-methoxy-5-
(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)pyrimidine using the procedure
described in Example 8 to provide
compound 1-93. ESI-MS m/z: 637.4 [M+H]
Example 68
S
CI 0 0
110
S? 1.1
N H N N 2
SnBu3
HNLNrjNH2
1-56 1-94 c7,
[00817] Compound 1-56 (151 mg, 0.342 mmol), 2-Methyl-5-
(tributylstannyl)thiazole (159 mg, 0.410
mmol), PdC12(amphos)2 (24.19 mg, 0.034 mmol), PdC12(amphos)2 (24.19 mg, 0.034
mmol) were charged in a dry
208

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
vial. The vial was evacuated and filled with Ar. Then degassed DMF (1 ml) was
added and the mixture was stirred
at 100 C overnight. The reaction was cooled and the material was extracted
with Et0Ac and wash with water then
brine. The organic layer was dried, filtered and concentrated to dryness.
Purification on silica gel (12g) column and
eluting with DCM/methanol (0 to 5% of methanol) provided compound 1-94. ESI-MS
m/z: 506.2 [M+H]+.
Example 69
CI 0 NH 0
SNN
Methylamine
HN N CI DIPEA, NMP, 120 C HF1 N N
NN NN
1-52 1-95
[00818] A mixture of compound 1-52 (500 mg, 1.09 mmol, 1 equiv.), N,N-
Diisopropylethylamine (0.56
mL, 3.28 mmol, 3 equiv.), 1.5 mL methylamine (40% in water, 17.2 mmols, 15.8
equiv.) in 5 mL NMP was heated
in a sealed tube at 120 C for 45 h. 1.5 mL methylamine (40% in water, 17.2
mmols, 15.8 equiv.) and 1 mL NMP
were added and heated in a sealed tube at 120 C for another 18 h. The mixture
was cooled, diluted with 10 mL 1:1
mixture of saturated brine and 2.5 M sodium carbonate and 25 mL DCM. The
aqueous layer was extracted with
DCM (3x5 mL), the combined organic layers were washed with water (10 mL),
dried over sodium sulfate and the
solvents were evaporated in vacuo. The residue was chromatographed on silica
gel (4 g, ISCO) using acetone-DCM
as eluent. The product fractions were concentrated and the impurities were
crystallized off and the filtrates were
concentrated under reduced pressure to provide 106 mg of compound 1-95 as a
light yellow solid. ESI-MS m/z:
452.2 [M+H]+.
Example 70
CI 0 CI 0
SNN
Methylamine
HN N CI DIPEA, NMP, 120 C HN N N
nr
NN NN
1
1-52 -96
[00819] A mixture of compound 1-52 (522 mg, 1.13 mmol, 1 equiv.), N,N-
Diisopropylethylamine (0.58
mL, 3.39 mmol, 3 equiv.), 0.985 mL methylamine (40% in water, 11.2 mmols, 10
equiv.) in 5 mL NMP was heated
in a sealed tube at 100 C for 0.5 h. The clear solution was mixture was
cooled to room temp, diluted with 10 mL
water, added to crushed ice, the precipitated white solids were filtered under
suction, washed with water (3x10 mL)
and dried under high vacuum to give 516 mg compound 1-96 as a white solid. ESI-
MS m/z: 457.2 [M+H]+.
209

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 71
1)Me
N
CI 0 00 0 N
1.1
_______________________________________________ =
HN N N HN N N
T
NN
NN
1-96 I 1-97
II
[00820] A mixture of compound 1-96 (100 mg, 0.219 mmol, 1 equiv.), 2-
Methoxypyrimidine-5-boronic
acid (67.4 mg, 0.438 mmol, 2 equiv.), sodium carbonate (116 mg, 1.094 mmol, 5
equiv.), and Pd-AMPHOS catalyst
(31.0 mg, 0.044 mmol, 0.2 equiv.) in 2.9 mL degassed 4:1 dioxane-water was
sparged with argon for 2 min. The
mixture was sealed and heated at 100 C for lh, cooled, diluted with 10 mL
each of DCM and water. The organic
layer was collected, the aqueous layer was extracted with DCM (3x10 mL), the
combined DCM layers were washed
with water (10 mL), brine (10 mL), dried over sodium sulfate and the solvents
were removed in vacuo. The residue
was purified on silica gel (12 g, ISCO) using 0-10% methanol in 1:1 DCM-Et0Ac
as eluent to provide 97 mg of
compound 1-97 as a light yellow solid. ESI-MS m/z: 531.3 [M+H]
Example 72
AN H
CI 0 SI 0 40)
HN N N HN N N
" "
NN NN
1-96 1-98
[00821] A mixture of compound 1-96 (100 mg, 0.219 mmol, 1 equiv.), 2-
Methylpyrimidine-5-pinacol
boronate (96mg, 0.438 mmol, 2 equiv.), sodium carbonate (116 mg, 1.094 mmol, 5
equiv.), and Pd-AMPHOS
catalyst (31.0 mg, 0.044 mmol, 0.2 equiv.) in 2.9 mL degassed 4:1 dioxane-
water was sparged with argon for 2 min.
The mixture was sealed and heated at 100 C for lh, cooled, diluted with 10 mL
each of DCM and water. The
organic layer was collected, the aqueous layer was extracted with DCM (3x10
mL), the combined DCM layers were
washed with water (10 mL), brine (10 mL), dried over sodium sulfate and the
solvents were removed in vacuo. The
residue was purified on silica gel (12 g, ISCO) using 0-10% methanol in 1:1
DCM-Et0Ac to provide 110 mg of
compound 1-98 as a light yellow solid. ESI-MS m/z: 515.3 [M+H]+.
210

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 73
OMe
N ' N
1 / 0 N 0
,
al)a0H
N
1-99
[00822] Compound 1-99 was synthesized by coupling compound 1-100 and 2-
methoxy-5-(4,4,5,5-
tetramethy1-1,3,2-dioxaborolan-2-yl)pyrimidine using the procedure described
in Example 8. ESI-MS m/z: 517.3
[1\4+1-1]+.
Example 74
CI 0 0
CI CI 0 0
0 ; N
f
-IP-
0 ;
NOH
1-1 F11-12 ,
G-1 HNOH
I I
NN
1-100
[00823] Compound 1-100 was prepared from compound I-1 coupled to (G-1)
(see Barlin, Gordon B.; Tan,
Weng Lai, Australian Journal of Chemistry (1984), 37(12), 2469-77) using
Method I. ESI-MS m/z: 443.2 [1\4+1-1]+.
Example 75
CI 0 0 NH 0 0
40; N
Methylamine 10 /
1-11 N NH2 DIPEA, NMP, 120 C
1-1r1NNH2
II II
N 1-101 NN
1-56 N
[00824] A mixture of compound 1-56 (100 mg, 0.226 mmol, 1 equiv.), N,N-
Diisopropylethylamine (0.16
mL, 0.9 mmol, 4 equiv.), 1.5 mL methylamine (40% in water, excess) in 1 mL NMP
was heated in a sealed tube at
120 C for 18 h. Another 1.5 mL methylamine in water and 1 mL NMP were added,
the tube was sealed and the
mixture was heated at 120 C for 52h. The contents were cooled, added to 15 mL
water, the precipitate were filtered
under suction, washed with water (3x10 mL) and dried under high vacuum to
provide 71 mg of compound 1-101 as
a white solid. ESI-MS m/z: 438.3 [M+H]+.
211

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 76
CI 0 0 N 0 0
0 ; N
Dimethylamine _________________________________________ 10 /
- _
I-11 N NH2 DIPEA, NMP, 120 C
1-1r1NNH2
II II
NN 1-102 N N
1-56 II
[00825] A mixture of compound 1-56 (100 mg, 0.226 mmol, 1 equiv.), N,N-
Diisopropylethylamine (0.16
mL, 0.9 mmol, 4.0 equiv.), 1.13 mL dimethylamine (2.0 M in THF, 10 equiv.) in
1 mL NMP was heated in a sealed
tube at 120 C for 18 h. Another 2 mL dimethylamine in THF and 2 mL NMP were
added, the tube was sealed and
the mixture was heated at 120 C for 52h. The contents were cooled, diluted
with Et0Ac (20 mL), added to 30 mL
water, the organic layer was collected, the aqueous layer was extracted with
Et0Ac (3x10 mL), the combined
organic layers were washed with water (20 mL), brine (10 mL), dried over sod.
sulfate and the solvents were
evaporated in vacuo. The residue was chromatographed on silica gel (4 g, ISCO)
using DCM-Acetone to provide
38.8 mg of compound 1-102 as a white solid. ESI-MS m/z: 452.3 [M+H]+.
Example 77
a
NN
F
I CI 0
el
CI 0 SI F 1 \r CI CI 0 el F N
N B-2 N 0 / Me
I. / Me -I" / Me 1-IFINNH2
F1H2 FIFJ NY CI
II
NN
II
I-103a NN
1-103
I-103b
[00826] Amine I-103a (made by general method E) was coupled to (B-2) using
Method Ito provide
compound I-103a. Compound 1-103 was prepared from compound I-103a using Method
J. ESI-MS m/z: 461.2
[M+H]+.
Example 78
I 0
N 0
CI N
I
CI 0 1 CI 0 N 1 0,B / /
N
-74_6 0 N
N N '
A 3 110
1101 / Me / Me
________________________________________________________ lel / Me
_
NH2 FIF1 N =
1-104 HN N
NN
NN
1-105 1-106
212

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
[00827] Amine 1-104 (prepared according to method L) was coupled to (A-3)
using Method I to provide
compound I-105. Compound I-105 was coupled to 1-methy1-5-(4,4,5,5-tetramethyl-
1,3,2-dioxaborolan-2-
yl)pyridin-2(1H)-one in the Suzuki reaction performed in analogous fashion to
compound 1-21 in Example 8 to
provide compound 1-106. ESI-MS m/z: 467.2 [M+H] '.
Example 79
I 0
N
CI 0 I
N 0 _7402_,B1
/
0 0 0
N
401 / Me
/ Me
HN N z
:0 HN N
L11
N
N
1_2 1-107
[00828] Compound 1-2 was coupled to 1-methy1-5-(4,4,5,5-tetramethyl-1,3,2-
dioxaborolan-2-yl)pyridin-
2(1H)-one in the Suzuki reaction performed in analogous fashion to compound 1-
21 in Example 8 to provide
compound 1-107. ESI-MS m/z: 501.2 [M+H]+.
Example 80
CI
rkli -Isi\ CI 0 ei
CI 0 0 N`-'=-,-1
0 ;
0 ; A-2
..
Hi
z
NH2 N N
11-1 11-2 NJ(
[00829]
Amine II-1 was prepared using Method E and then coupled with (A-2) using
Method G to
provide compound 11-2. ESI-MS m/z: 416.0 [M+H] '.
Example 81
CI
Isil- \ CI 0 0
Cl 0 0 *N-L..---/
0
F ;
0 ; F A-2 .
HFJ
z
NH2 N N
11-3 11-4 r%J._I
213

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00830] Amine 11-3 was prepared using Method E and was then coupled with
(A-2) using Method G to
provide compound 11-4. ESI-MS m/z: 434.0 [M+H] '.
Example 82
/
N-N /
y N-N
/
V
CI 0 0 B,
0- 0 0 a
110 ; 2 I. = ;
_
FIR FIF1)n
11-2 NLT 11-5 NJ
[00831] Compound 11-5 was prepared from compound 11-2 according to the
following procedure:
[00832] A mixture of (S)-8-chloro-2-pheny1-3-(1-(pyrazolo[1,5-a]pyrimidin-
7-
ylamino)ethyl)isoquinolin-1(2H)-one 11-2 (66 mg, 0.158 mmol, 1.0 eq), 1-methy1-
4-(4,4,5,5-tetramethyl-1,3,2-
dioxaborolan-2-y1)-1H-pyrazole (66 mg, 0.316 mmol, 2.0 eq), PdC12(dppf)(13 mg,
0.016 mmol, 0.1 eq) and
Na2CO3(84 mg, 0.79 mmol, 5.0 eq) was suspended in a mixture of N,N-
dimethylacetamide (5 mL) and water (2
mL). The resulting mixture was degassed and back-filled with argon three times
and stirred at 80 C under argon
overnight. The mixture was concentrated in vacuo and the residue was purified
by flash column chromatography on
silica gel (1-5% Me0H-DCM) to afford the product, (S)-8-(1-methy1-1H-pyrazol-4-
y1)-2-phenyl-3-(1-(pyrazolo[1,5-
a]pyrimidin-7-ylamino)ethyl)isoquinolin-1(2H)-one 11-5 (43 mg, 60% yield). ESI-
MS m/z: 462.2 [M+H] '.
Example 83
CI
Ni -Isi CI 0 j\
J\ ;
Ni---z...--->
CI 0
0
0 ; A-2
..
Hi
z
NH2 N N
11-6 11-7 NJ(
[00833] Amine 11-6 was prepared using Method E and then coupled with (A-
2) using Method G to
provide compound 11-7. ESI-MS m/z: 380.2 [M+H] '.
Example 84
/
N¨N
/
V 0
NI\
1101
iih)n
N N
11-8 NLT
214

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00834] Compound 11-8 was prepared from compound 11-7 in analogous
fashion to compound II-5 in
Example 82. ESI-MS m/z: 426.2 [M+H] '.
Example 85
/
N-N
0 ; F
141
)n
N N
11-9 NLT
[00835] Compound 11-9 was prepared from compound 11-4 in analogous
fashion to compound 11-5 in
Example 82. ESI-MS m/z: 480.2 [M+H] '.
Example 86
OTs
0
N-...1
110 ;
do, N'
; B-6
_______________________________________ 3.. FIFI
-
FIFI2 NNI'N1
V.---7.--1
II-1 11-10
[00836] Intermediate imidazo[1,2-b]pyridazin-8-y14-methylbenzenesulfonate
(B-6) was prepared
according to Method B. Amine II-1 was then coupled to (B-6) using Method G to
provide compound II-10. ESI-
MS m/z: 416.2 [M+H]'.
Example 87
OTs
A
N.)
0 ;
CI 0 yx N'
110 ; B-6
. .
HN
_
17.1H2 NN-N
11-6 11-1 1
[00837] Amine 11-6 was prepared using Method E and then coupled with (B-
6) using Method G to
provide compound 11-1 1. ESI-MS m/z: 380.2 [M+H]'.
215

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 88
/
N-N Br
y, r......,,N N-N"
13, N-N
0 0 / CINA-7? /
- Z
CI 0 7
0 0
mA
B-3 NA
NA ___
_D.,.. so . _31...
0 /
0 /
z
_
k H2 =
NH2 HNCI
N
11-6 11-12 N r N-
\-,-.--/
11-13
[00838] Amine 11-6 was prepared using Method E. Amine 11-12 was coupled
with (B-3) using Method
G to provide compound 11-13. ESI-MS m/z: 460.2 [M+H] .
[00839] Amine 11-12 was prepared from 11-6 according to the following
procedure:
[00840] To a mixture of (S)-3-(1-aminoethyl)-8-chloro-2-
cyclopropylisoquinolin-1(2H)-one 11-6 (878
mg, 3.35 mmol, 1.0 eq) and 1-methy1-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-
2-y1)-1H-pyrazole (815 mg, 3.92
mmol, 1.2 eq) in anhydrous DMA (10 mL) in a sealed tube, PdC12(dppf) (219 mg,
0.27 mmol, 0.08eq) and aqueous
Na2CO3 solution (1 M, 10.0 mL, 10.0 mmol, 3.0 eq) were added and the resulting
mixture was stirred at 120 C for 3
h. The reaction mixture was allowed to cool to RT, quenched with water, and
then extracted with ethyl acetate (200
mL x 3). The combined organic layers were washed with brine, dried over Na2SO4
and filtered. The filtrate was
concentrated in vacuo and the residue was purified by ISCO (silica gel
cartridge, 0-8% Me0H-DCM) to afford the
product, (S)-3-(1-aminoethyl)-2-cyclopropy1-8-(1-methyl-1H-pyrazol-4-
yl)isoquinolin-1(2H)-one 11-12 as a
pink/magenta solid. ESI-MS m/z: 309.2 [M+H] '.
Example 89
OTs
CI 0 ei
N-N.)
CI 0 0 N F
N F B-6 0 7
... z
0 7
HI
F1H2 N
NN
N
11-3 -
-\_:--/-
11-14
[00841] Amine 11-3 was prepared using Method E and then coupled with (B-
6) using Method G to
provide compound 11-14. ESI-MS m/z: 434.0 [M+H]'.
216

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 90
&?/ Br
N-N
CI ,B =N40 /
CINl-N-) 0
0 0 0
B-3
z
F1H2 HNCI
F1H2
/L
N N
11-3 11-15
11-16
[00842] Compound 11-16 was prepared from compound 11-15 in analogous
fashion to compound 11-13
in Example 88. ESI-MS m/z: 514.0 [M+H]
Example 91
N-N
S NF
V 0 el
HN
Thq-N1
11-17
[00843] Compound 11-17 was prepared from compound 11-14 in analogous
fashion to compound 11-5
in Example 82. ESI-MS m/z: 480.2 [M+H]
Example 92
N-N
/
0 N
HN
NN -N
11-18
[00844] Compound 11-18 was prepared from compound II-10 in analogous
fashion to compound 11-5
in Example 82. ESI-MS m/z: 462.2 [M+H]
217

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Example 93
CI
4110
ci 0
NS A-2 S¨

H:J_
FIR )n
NH2 N N
11-19 11-20
[00845] Amine 11-19 was prepared using Method F and then coupled with (A-
2) using Method G to
provide compound 11-20. ESI-MS m/z: 422.0 [M+H]
Example 94
N-N
S
FIN
N N
11-21 tr
[00846] Compound 11-21 was prepared from compound 11-20 in analogous
fashion to compound 11-5
in Example 82. ESI-MS m/z: 468.2 [M+H]
Example 95
OTs
CI\ 11
CI\ 11
B-6
HN-
N- H2 NN
11-19 11-22
[00847] Amine 11-19 was coupled with (B-6) using Method G to provide
compound 11-22. ESI-MS
m/z: 422.0 [M+H]
218

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 96
N-N

)0L
S
HN
N. ,N-N
11-23
[00848] Compound 11-23 was prepared from compound 11-22 in analogous
fashion to compound 11-5
in Example 82. ESI-MS m/z: 468.2 [M+H]
Example 97
CI
NN N\ CI 0
CI 0
D-6
HFiN)
F1H2 N N
11-1 11-24 NLT
[00849] Amine II-1 was coupled with (D-6) using Method G to provide
compound 11-24. ESI-MS m/z:
417.0 [M+H]
Example 98
CI ,NH, CI 0
N 1.n-BuLl/THF
COOH (C0C1)2 NINO 2.HMPA
. 1101 H 0 ____
DCM DCM/pyridine
11-25 11-26Boc'NH I
CI
CI 0
CI 0
1% Ni%
Conc. HCI N. A-2
______________ a
Me0H z HN
NH2
NN
11-27 11-28 NL-#
219

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00850] Compound 11-28 was prepared in 4 steps according to the following
procedures:
[00851] Compound 11-26 was prepared from 11-25 in analogous fashion to (E-
2) in Method E except
that 2-(pyrrolidin- 1 -yl)ethanamine was used in place of aniline and pyridine
was used as a base in place of
triethylamine . Compound 11-26 was converted to 11-27 according to the
following procedure:
[00852] To a mixture of 2-chloro-6-methyl-N-(2-(pyrrolidin-1-
yl)ethyl)benzamide 11-26 (1.33 g, 5.0
mmol, 1.0 eq) and HMPA (0.87 mL, 5.0 mmol, 1.0 eq) in THF (12.5 mL) at -60 C
under argon, n-butyllithium
solution (2.5 M in hexanes, 5.0 mL, 12.5 mmol, 2.5 eq) was added dropwise
while keeping the temperature below -
60 C. The resulting mixture was stirred between -70 C and -60 C for 30 min.
To this mixture, (S)-tert-butyl 1-
(methoxy(methyl)amino)-1-oxopropan-2-ylcarbamate (1.39 g, 6.0 mmol, 1.2 eq)
was added quickly. The resulting
mixture was allowed to slowly warm to RT (over 1 h) and then stirred at RT for
an additional 2 h. The reaction
mixture was cooled to 0 C, quenched with water (5.0 mL), and then acidified
with aqueous HC1 (5 M) to adjust the
pH to 5-6. The mixture was concentrated in vacuo. The residue was suspended in
a mixture of Me0H (20 mL) and
H20 (5.0 mL), concentrated HC1 (10.0 mL) was added and the resulting mixture
was stirred at reflux for 2 h. The
mixture was cooled to -5 C and basified with saturated aqueous NH4OH to
adjust the pH to 8-9. The precipitate was
filtered off and the filtrate was extracted with DCM (20 mL x 3). The combined
organic layers were dried over
anhydrous Na2SO4 and filtered. The filtrate was concentrated in vacuo and the
residue was purified by flash column
chromatography on silica gel (1-10% Me0H-DCM) to afford the product, (S)-3-(1-
aminoethyl)-8-chloro-2-(2-
(pyrrolidin-1-y1)ethyl)isoquinolin-1(2H)-one II-27)as a yellow solid.
[00853] Compound 11-27 was then coupled with (A-2) using Method G to
provide compound 11-28.
ESI-MS m/z: 437.2 [M+H]
Example 99
OTs
CI 0
CI 0 NN
N INµp B-6
F1H2
11-27 11-29
[00854] Amine 11-27 was coupled with (B-6) using Method G to provide
compound 11-29. ESI-MS
m/z: 437.2 [M+H]
Example 100
CI
N
N C 1 0
C 1 0
D-6
H
11-3 F1H 2 11-30 N N
NJ
220

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00855] Amine 11-3 was coupled with (D-6) using Method G to provide
compound 11-30. ESI-MS m/z:
435.0 [M+H] '.
Example 101
OTs
ei
0 /N F
N
0 ; F C-4 3.
HR-
11-3 F1H2 11-31
N/ N-
)==-1
[00856] Amine 11-3 was coupled with (C-4) using Method G to provide
compound 11-31. ESI-MS m/z:
448.2 [M+H] '.
Example 102
OTs
[.._..,:.N CI 0 ei
CI 0 ei N-N,,)
0
C-4 0 ;
;FIFI
-
11-1 Fl H2 11-32 Isl'iNi
N\___ j
7----
[00857] Amine II-1 was coupled with (C-4) using Method G to provide
compound 11-32. ESI-MS m/z:
430.2 [M+H] '.
Example 103
i
N-N
/ "O N 40
0
HN
11-33 NN-N
7-j-
[00858] Compound 11-33 was prepared from compound 11-32 in analogous
fashion to compound 11-5
in Example 82. ESI-MS m/z: 476.2 [M+H] '.
221

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 104
.ci .ci
CI 0
CI 0 Si CI 0 1. 0 NH
_... _,..
N N
0 HN'Fmoc
NH 2 FIN'Fmoc
11-34 11-35 11-36
0
CI CI CI CI
____________ 0 N 0 N 0 N
_,..
3. _,..
z
FIN'Fmoc HN,Fmoc FIN'Fmoc
11-37 11-38 11-39
CI 0 OH
CI 0 CI ro
0
CI
,.) N 10 N _____________
_,.. _õ. a.
0 N
:
HN HN
-Fmoc 'Fmoc
HN
11-40 11-41 11A2 'Fmoc
CI ro
ro ,L N
CI 0 1µ1.) N)-"=--:-->
D-6 __ .N 3.
/ F11-1
NH2N
IµV Ni \--- -
II-43 11-44
[00859] Compound 11-44 was prepared according to the following
procedures:
[00860] Compound 11-34 was prepared according to Method E. To a stirred
mixture of 11-34 (10.0
mmol, 1.0 eq) in DCM (20 mL), saturated aqueous NaHCO3 solution (2.8 mL) was
added. The mixture was cooled
to 0 C and 9-fluorenylmethyl chloroformate (10.5 mmol, 1.05 eq) was added in
one portion. The resulting mixture
was allowed to warm to RT and stirred at RT for 1 h. The organic layers were
separated, washed with water (50 mL
x 2), dried over anhydrous MgSO4 and filtered. The filtrate was concentrated
in vacuo to afford the product 11-35.
[00861] Compound 11-35 (10.0 mmol, 1.0 eq) was then dissolved in TFA (25
mL) and the resulting
mixture was stirred at reflux for 1 h. The mixture was allowed to cool to RT
and concentrated in vacuo to remove
TFA. The resulting brown oil residue was partitioned between DCM (50 mL) and
saturated aqueous NaHCO3 (50
mL) solution. The organic layer was washed with water (20 mL x 2), dried over
anhydrous MgSO4 and filtered. The
filtrate was concentrated in vacuo to afford the product 11-36.
222

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00862] A mixture of compound 11-36 (45.0 mmol) and DMF (0.3 mL) in SOC12
(100 mL) was stirred
at reflux for 2 h. The mixture was allowed to cool to RT and concentrated in
vacuo. The residue was purified by
flash column chromatography on silica gel to afford the product 11-37.
[00863] To a stirred mixture of 11-37 (5.4 mmol, 1 eq), Pd(OAc)2 (363 mg,
1.62 mmol, 0.3 eq) and
PPh3 (846 mg, 3.24 mol, 0.6 eq) in THF (50 mL) at RT, tributyl(vinyl)fin (1.97
g, 5.94 mmol, 1.1 eq) was added and
the resulting mixture was stirred at reflux overnight. The mixture was allowed
to cool to RT and filtered through
silica gel (10 g). The filtrate was concentrated in vacuo and the residue was
purified by flash column
chromatography on silica gel (1-5% Me0H-DCM) to afford the product 11-38.
[00864] To a stirred mixture of 11-38 (7.6 mmol, 1 eq) in a mixture of
1,4-dioxane (40 mL) and H20
(40 mL) at RT, osmium tetraoxide (5 mg) was added and the resulting mixture
was stirred at RT for 30 min. To this
mixture, sodium periodate (3.3 g, 15.2 mmol, 2 eq) was added and the resulting
mixture was stirred at RT overnight.
The reaction mixture was filtered through silica gel (10 g). The filtrate was
extracted with DCM (3 x 60 mL),
washed with brine, dried over Na2SO4 and filtered. The filtrate was
concentrated in vacuo and the residue was
purified by flash column chromatography on silica gel (1-5% Me0H-DCM) to
afford the aldehyde 11-39.
[00865] To a solution of compound 11-39 (3.53 mmol, 1 eq) in DMF (40 mL)
at RT, oxone (6.5 g,
10.59 mmol, 3 eq) was slowly added and the resulting mixture was stirred at RT
overnight. The reaction mixture
was poured into water (50 mL) and neutralized with concentrated ammonium
hydroxide to adjust the pH value to 7-
8. The mixture was stirred at RT for 30 min and then extracted with ethyl
acetate (3 x 50 mL). The combined
organic layers were washed with brine, dried over Na2SO4 and filtered. The
filtrate was concentrated in vacuo and
the residue was purified by flash column chromatography on silica gel (1-5%
Me0H -DCM) to afford the carboxylic
acid 11-40.
[00866] To a solution of compound 11-40 (0.327 mol, 1 eq) and DMF (0.1
mL) in DCM (10 mL),
oxalyl chloride (83.1 mg, 0.654 mmol, 2 eq) was added and the resulting
mixture was stirred at RT for 1 h. The
mixture was concentrated in vacuo to afford the acid chloride 11-41 as a
yellow oil. The product obtained was used
directly in the next step without purification.
[00867] To a solution of morpholine (34 mg, 0.392 mmol, 1.3 eq) and
triethylamine (100 mg, 0.981
mol, 3 eq) in DCM (20 mL), acid chloride 11-41 was added at RT. The resulting
mixture was stirred at RT for 1. The
reaction mixture was poured into water (50 mL) and extracted with DCM (3 x 50
mL). The combined organic layer
was washed with brine, dried over Na2SO4 and filtered. The filtrate was
concentrated in vacuo and the residue was
purified by flash column chromatography on silica gel (1-5% Me0H-DCM) to
afford the amide 11-42.
[00868] To a stirred mixture of 11-42 (1.76 mmol, 1 eq) in Et0H (50 mL)
at RT, morpholine (3.05 g,
35.2 mmol, 20 eq) was added and the resulting mixture was stirred at reflux
for 4 h. The mixture was allowed to
cool to RT and then concentrated in vacuo. The residue was purified by flash
column chromatography on silica gel
(1-10% Me0H-DCM) to afford the amine 11-43.
[00869] Amine 11-43 was then coupled with (D-6) using Method G to provide
compound 11-44. ESI-
MS m/z: 438.2 [M+H]'.
223

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 105
/
N-N
y \
N-N
CI 0 A
2 0 NA
_________________________________________ a.
HN
/N1-14
r,-_/4
11-45 Nõ)
N-
[00870]
Compound 11-45 was prepared from compound II-11 in analogous fashion to
compound 11-5
in Example 82. ESI-MS m/z: 426.2 [M+H] '.
Example 106
OMe
OMe
OMe CI N ' N
N ' N
A I
). e
I N ' N CM 0 l
/
I ....)
0
CI 0 a - 0 a, N__ 0 N N H0 OH N A 2 /
________________________ 3.
IW ___________________________________________________ 3 Z
NH
. .
F.1H2 1;1E12 11
...L.----)
11-1 11-46 11-47 11
[00871]
Compound 11-46 was prepared from compound II-1 in analogous fashion to
compound 11-12 in
Example 88 except that 2-methoxypyrimidin-5-ylboronic acid was used in place
of 1-methy1-4-(4,4,5,5-tetramethyl-
1,3,2-dioxaborolan-2-y1)-1H-pyrazole. Compound 11-46 was then coupled to
compound A-2 to provide compound
11-47 according to Method G. ESI-MS m/z: 490.2 [M+H] '.
Example 107
OMe
OTs
OMe
r-Jkl 1 N
N N
0 0 IV'
1. N
________________________________________ I ,
NH
Fl H2
N
11-46
11-48 Isr
224

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00872] Compound 11-46 was coupled with B-6 to provide compound 11-48
according to Method G. ESI-
MS m/z: 490.2 [M+H]'.
Example 108
CI
0
NI N, CI 0
,õ. = ...:,.,. . . ..). . . .."--. .,.. j. . ;
CI 0
N0 H2N NZ-3 .
___________________________________________ 11. F1H
0 /
_ Nil-
I;IFI2
H2N N
11-1
11-49
[00873] Compound 11-49 was prepared by coupling compound Z-3 to compound
II-1 according to Method
G. ESI-MS m/z: 431.2 [M+H]+.
Example 109
re
1)Me
N N
; 0 y
CI 0 0 N N I
/ 0 0 0 H0 OH ;
i.
1;11i
111-1
14, \-ril
I-12N N
-... ,--)-...-z>
FI2N N
11-49 11-50
[00874] Compound 11-50 was prepared from compound 11-49 in analogous
fashion to compound 11-5 in
Example 82 except that 2-methoxypyrimidin-5-ylboronic acid was used in place
of 1-methy1-4-(4,4,5,5-tetramethyl-
1,3,2-dioxaborolan-2-y1)-1H-pyrazole. ESI-MS m/z: 505.2 [M+H] '.
Example 110
CI
is1C.¨=N
I 0F3 0 40)
Isl..--N N CF3 0 0
CF3 0 00
t
A-2 o) = ,
is ;
_
is ; FIR N
i
/yN ¨1.-
FIR N
N __________________________________________ iR H2

N NN
0 µ---NH
III-1 111-2 111-3
225

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00875] Amine III-1 was prepared according to Method Q. Compound III-1 was
then converted to
compound 111-2 using Method S. The THP protecting group was then removed
according to the following general
procedure:
[00876] To a mixture of compound 111-2 in ethanol (4 vol)/water (2 vol),
concentrated HC1 solution (2 vol)
was added and the resulting mixture was stirred at RT for 1 h. The resulting
mixture was diluted with cold water,
basified with saturated aqueous NaHCO3 to adjust the pH to 8-9 and then
extracted with DCM (3 x 20 vol). The
combined organic layers were washed with brine, dried over Na2SO4 and
filtered. The filtrate was concentrated in
vacuo. The residue purified by flash column chromatography on silica gel
eluting with a mixture solvent of Me0H
and DCM to afford compound 111-3. ESI-MS m/z: 451.0[M+H]+.
Example 111
CI
CF3 0
CF3 0
110
1.1 B-3
Hh N
h- H2 NN
III-1 111-4
[00877] Compound 111-4 was prepared by coupling amine III-1 and (B-3)
according to Method Q. ESI-
MS m/z: 462.0 [M+H]'.
Example 112
CI
CF3 0
I
CF3 0
1-4
HN N
k- H2
N T
111-1 111-5
[00878] Compound 111-5 was prepared by coupling amine III-1 and (1-4)
according to Method S. ESI-MS
m/z: 468.0 [M+H]
226

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 113
CI
1%11 -14\ C F3 0 el
CF3 0
0 N ;
0 J-2
,. _
z FIN
NH2 N N
NLT
111-1 111-6
[00879] Compound 111-6 was prepared by coupling amine III-1 and (J-2)
according to Method S. ESI-MS
m/z: 450.2 [M+H] .
Example 114
Br
r.......N CF3 0 IS CF3 0 40)
CF3 0 IS) CIN ' N -1 0 N
0 ;
; K-3
0
... /
HN C-a.1 _
HN-
N1-12
N'/N-N
NN-N
111-1 111-7 111-8
[00880] Compound 111-7 was prepared by coupling amine III-1 and (K-3)
according to Method S.
Compound 8 was then prepared according to the following procedure:
[00881] To a suspension of (S)-3-(1-(6-chloroimidazo[1,2-b]pyridazin-8-
ylamino)ethyl)-2-pheny1-8-
(trifluoromethyl)isoquinolin-1(2H)-one 111-7 (50 mg, 0.104 mmol, 1.0 eq) in
Me0H (20 mL), palladium on carbon
(10%, 17 mg) and triethylamine (0.1 mL, 0.72 mmol, 6.9 eq) were added. The
resulting mixture was degassed and
back-filled with hydrogen (three cycles), and then stirred at RT under
hydrogen overnight. The mixture was filtered
and the filtrate was concentrated in vacuo. The residue was purified by flash
column chromatography on silica gel
(5-10% ethyl acetate-petroether) to afford the product, (S)-3-(1-(imidazo[1,2-
b]pyridazin-8-ylamino)ethyl)-2-
pheny1-8-(trifluoromethyl)isoquinolin-1(2H)-one 111-8. ESI-MS m/z: 450.2 [M+H]
.
Example 115
CI
i%1---N1 CF3 0 0
CF3 0 0 N S 0 ; F
Si N F 1-4
a. -
HN N
, i
NH2 /-N
N T
t-S
111-9 111-10
227

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
[00882] Amine 111-9 was prepared according to Method Q. Compound III-10
was then prepared by
coupling compound 111-9 with (I-4) using Method S. ESI-MS m/z: 486.0 [M+H] .
Example 116
CI
141 --N1 CF3 0 0
CF3 0
N-L,-.-..--->
0
0
1.1 N F ; F J-2
. _
-
NH2 N N
%T
111-9 111-1 1
[00883] Compound III-11 was prepared by coupling amine 111-9 and (J-2)
according to Method S. ESI-
MS m/z: 468.2 [M+H] .
Example 117
CI
N N CF3 0 0
CF3 0 0
SI ; F
; F B-3
N. _
Hk N
h-1-12
NN
111-9 111-12
[00884] Compound 111-12 was prepared by coupling amine 111-9 and (B-3)
according to Method S. ESI-
MS m/z: 480.2 [M+H]'.
Example 118
CF3 0 0
N F
0 /
a
HN N
i
N
N
-"¨NH
111-13
[00885] Compound 111-13 was prepared in analogous fashion to compound 111-
3 in Example 75 except
that amine 111-9 was used in place of amine III-1. ESI-MS m/z: 469.0 [M+H]'.
228

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 119
Br
r.....-.N
CIN1 CF3 0 0
CF3 0 0
0 ; F
K-3
0 ; F 3.- -
HL CI
_
NH2
N' N-
III-9 III-1 4
[00886] Compound 111-14 was prepared by coupling amine 111-9 and (K-3)
according to Method S. ESI-
MS m/z: 502.0 [M+H] '.
Example 120
CF3 0 0 CF3 0 40 0 ; F N F
01 /
_
RH I, ,C
T Elk
Is1-14
NN
N\.--.-1--
111-14 111-15
[00887] Compound 111-15 was prepared from compound 111-14 using the
analogous procedure for
compound 111-8 in Example 79. ESI-MS m/z: 468.2 [M+H]+.
Example 121
CI
N---N1
I CF3 0 0
-....
o
NNN N N CF3 0 001
CF3 0 SI
A-2 0 , 40, ;
1
. ; I-IFIõN -
,NH2 HN N NH2
NrYN 1
NH2
---N
0 <YN
\--NH
111-1 111-16 111-17
[00888] Compound 111-16 was prepared by coupling amine III-1 and (A-2)
according to Method S. This
compound 111-16 was then converted to compound 111-17 according to the
analogous procedure for compound 111-3
in Example 75. ESI-MS m/z: 466.0 [M+H] '.
229

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 122
CI F
j.
CF3 0 0 N N
H
0 0 ; ; F-1
.
1-1F1 N
_
F1112 F_yN
111-1 111_18 NH
[00889] Compound 111-18 was prepared by coupling amine III-1 and (F-1)
according to Method S. ESI-
MS m/z: 468.2 [M+H]'.
Example 123
CF3 0
CI CN
N=

--'. CF3 0 0
\
0 N N
H
____________________________________________________ 1101 ;
1101 /N E-5
. _
i
1-1F1 N
_
F1112 ....saN
NC \
111-1 111-19
[00890] Compound 111-19 was prepared by coupling amine III-1 and (E-5)
according to Method S. ESI-
MS m/z: 475.0 [M+H]'.
Example 124
0
_....¨NH2
C F 3 0 SI N N
H 0 ;0
0 /N G-3
. _
1-1F1 N
z 0 I -I
RH2 ,\......../N
H21%_--14H
111-1 111-20
[00891] Compound 111-20 was prepared by coupling amine III-1 and (G-3)
according to Method S. ESI-
MS m/z: 493.2 [M+H]'.
230

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 125
CI
/L N
N))
CF3 0 0
____________________________________________ 0 ;0
41N)
h- H2 N N
NLT
III-1 III-21
[00892] Compound 111-21 was prepared by coupling amine III-1 and (L-6)
according to Method S. ESI-
MS m/z: 451.2 [M+H]'.
Example 126
CI
N, N CF3 0 0
1 I
C F3 0 0 CI'N
Si0 ____________________________________________ /N ; M-3
. _
141 NY CI
_
174 H2
NN
III-1 III-22
[00893] Compound 111-22 was prepared by coupling amine III-1 and (M-3)
according to Method S. ESI-
MS m/z: 496.2 [M+H]'.
Example 127
CF3 0 40) C F3 0 0
N N
lel / 1101 /
_,... _
_
EIFI NY CI EIFI N NH2
N N N N
111-22 111-23 L.
[00894] Compound 111-23 was prepared from 111-22 according to the
following procedure:
[00895] Compound 111-22 (0.11 mmol, 1.0 eq) was suspended in ammonium
hydroxide (10-35% solution,
20 mL) in a sealed tube and the resulting mixture was stirred at 140 C
overnight. The mixture was allowed to cool
to RT and then extracted with DCM (3 x 15 mL). The combined organic layers
were washed with brine, dried over
Na2504 and filtered. The filtrate was concentrated in vacuo and the residue
was purified by column chromatography
on silica gel (1-2% Me0H-DCM) to afford compound 111-23. ESI-MS m/z: 477.2
[M+H] '.
231

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 128
CI
N CF3 0
CF3 0 CI' N S
N CI
17.1H2
N7Y
111-24
111-1
[00896] Compound 111-24 was prepared by coupling amine III-1 and
commercially available 5,7-
dichlorothiazolo[5,4-d]pyrimidine according to Method S. ESI-MS m/z: 502.0
[M+H]
Example 129
CF3 0 CF3 0
HN- N CI N NH2
III-24 III-25
[00897] Compound 111-25 was prepared from compound 111-24 in analogous
fashion to compound 111-23
in Example 92. ESI-MS m/z: 473.2 [M+H]
Example 130
CI
CF3 0
CF3 0 N S
1101
I. /N 1-4
Hh N
1;1- H2 /N
N T
-s
111-1 111-26 y
[00898] Compound 111-26 was prepared by coupling amine III-1 and (I-4)
according to Method S. ESI-
MS m/z: 482.0 [M+H]'.
232

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 131
CI
CF3 0
CF3 0
1.1
1101 C-6
N
h112
NN-N
III-1 III-27
[00899] Compound 111-27 was prepared by coupling amine III-1 and (C-6)
according to Method S. ESI-
MS m/z: 451.2 [M+H]'.
Example 132
CI
CF3 0
CF3 0
/N D-4
FIR N
RI-12
N'N
N=1
III-1 III-28
[00900] Compound 111-28 was prepared by coupling amine III-1 and (D-4)
according to Method S. ESI-
MS m/z: 451.2 [M+H]'.
Example 133
Ts,
0
Nj
CF3 0 IS)
CF3 0 el
N-5
1.1
HN
F1H2 NN
111-1 111-29
[00901] Compound 111-29 was prepared by coupling amine III-1 and (N-5)
according to Method S. ESI-
MS m/z: 464.0 [M+H]'.
233

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 134
CF 3 0
40/ NF
N NH2
N
Nr
t¨N1-1
111-30
[00902] Compound 111-30 was prepared in analogous fashion to compound 111-
17 in Example 88 except
that amine 111-9 was used in place of amine III-1 as starting material. ESI-MS
m/z: 484.2 [M+H]
Example 135
CI
)N
C F3 0
C F3 0 N
1101 0-8
HN
h- H2 NN
III-1 III-31 L.
[00903] Compound 111-31 was prepared by coupling amine III-1 and (0-8)
according to Method S except
that N,N-diisopropylethylamine was used in place of triethylamine. ESI-MS m/z:
461.2 [M+H]
Example 136
CI F
N l<FF
CFO 40:1
H2N
CF3 0 SI
__________________________________________ 101
H-2
HNNNH2
rill2
III-1 III-32
[00904] Compound 111-32 was prepared by coupling amine III-1 and (11-2)
according to Method S. ESI-
MS m/z: 494.0 [M+H]
234

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 137
CI
NO2 CF3 0 IS CF3 0
CF3 0 INCI MeNaHS
0H-
1.1 NH
111-12 Et3N / Et0H NO2 NO2
I
-1=1SH
111-1 111-33 N CI 111-34
CF3 0 CF3 0
CH(OEt)3
SnC12.H20
Reflux
NH HN
Et0H NH2
zyN
111-35
-N SH 111-36
[00905] Compound 111-36 was prepared from amine III-1 according to the
following procedures:
[00906] To a mixture of (S)-3-(1-aminoethyl)-2-pheny1-8-
(trifluoromethyl)isoquinolin-1(2H)-one III-1
(1.0 g, 3.0 mmol, 1.0 eq) and 2,4-dichloro-3-nitropyridine (578 mg, 3.0 mmol,
1.0 eq) in Et0H (10 mL),
triethylamine (608 mg, 6.0 mmol, 2.0 eq) was added and the resulting mixture
was stirred at reflux overnight. The
mixture was allowed to cool to RT and then concentrated in vacuo. The
resultant residue was purified by flash
column chromatography on silica gel (1% Me0H-DCM) to afford the product, (S)-3-
(1-(2-chloro-3-nitropyridin-4-
ylamino) ethyl)-2-phenyl-8-(trifluoromethyl) isoquinolin-1 (2H)-one 111-33.
[00907] To a solution of (S)-3-(1-(2-chloro-3-nitropyridin-4-
ylamino)ethyl)-2-pheny1-8-(trifluoromethyl)
isoquinolin-1 (2H)-one 111-33 (500 mg, 1.0 mmol, leq) in Me0H (10 mL), sodium
hydrosulphide (69 g, 1.29 mmol,
1.2 eq) was added and the resulting mixture was stirred at reflux for 2 h. The
mixture was allowed to cool to RT and
then concentrated in vacuo. The residue was purified by flash column
chromatography on silica gel (2-3% Me0H-
DCM) to afford the product, (S)-3-(1-(2-mercapto-3-nitropyridin-4-ylamino)
ethyl)-2-pheny1-8-
(trifluoromethyl)isoquinolin-1(2H)-one 111-34.
[00908] A mixture of (S)-3-(1-(2-mercapto-3-nitropyridin-4-ylamino) ethyl)-
2-phenyl-8-(trifluoro
methyl)isoquinolin-1(2H)-one 111-34 (120 mg, 0.247 mmmol, 1.0 eq) and stannous
chloride dihydrate (297 mg, 1.32
mmol, 5.3 eq) in Et0H (5 mL) was stirred at refluxed for 2 h. The resulting
mixture was cooled to RT and
concentrated in vacuo. The residue was suspended in water (5 mL), neutralized
with saturated NaHCO3 solution to
adjust the pH to 7-8 and then extracted with ethyl acetate (5 mL x 3). The
combined organic layers were washed
with brine, dried over Na2SO4 and filtered. The filtrate was concentrated in
vacuo to afford the product, (S)-3-(1-(3-
amino-2-mercaptopyridin-4-ylamino) ethyl)-2-phenyl-8-
(trifluoromethyl)isoquinolin-1(2H)-one 111-35.
[00909] A mixture of (S)-3-(1-(3-amino-2-mercaptopyridin-4-ylamino)ethyl)-
2-pheny1-8-(trifluoro
methyl)isoquinolin-1(2H)-one 111-35 (80 mg, 0.175 mmol, 1.0 eq) in triethyl
orthoformate (180 mL) was stirred at
235

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
reflux for 2 h. The reaction mixture was allowed to cool to RT and then
concentrated in vacuo. The residue was
purified by flash column chromatography on silica gel (1-2% Me0H-DCM) to
afford the product, (S)-2-pheny1-3-
(1-(thiazolo[5,4-b]pyridin-7-ylamino)ethyl)-8-(trifluoromethyl)isoquinolin-
1(2H)-one 111-36. ESI-MS m/z: 467.0
[M+H]
Example 138
CI
NCN CF3 0
CF3 0N
1101 P-4
CN
h- H2 NfN
1
111-1 111-37
[00910] Compound 111-37 was prepared by coupling amine III-1 and (P-4)
according to Method S. ESI-
MS m/z: 486.2 [M+H]'.
Example 139
CI
N
F3C\ 110
F3C\
N S
N
trN 1-4
_________________________________________ >
HI;1 N
1;1H2
III-38 III-39 N
[00911] Amine 111-38 was prepared according to Method R. Compound 111-39
was then prepared by
coupling amine 111-38 and (1-4) according to Method S. ESI-MS m/z: 474.0 [M+H]
Example 140
F3C\
N
S
H N
N
III-40 N/y
[00912] Compound 111-40 was prepared from amine 111-38 in analogous
fashion to compound 111-3 in
Example 75. ESI-MS m/z: 457.0 [M+H]
236

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 141
F3C\
tN
Hh N
III-41 N
[00913] Compound 111-41 was prepared from amine 111-38 and (B-3) according
to Method S. ESI-MS m/z:
468.0 [M+H]'.
Example 142
CI N
I I CF3 0
CF3 0 NC N
NH2 1.1
________________________________________ 11.
N
NC
NH2
rN
NH2
III-1 111-42
[00914] Compound 111-42 was prepared by coupling amine III-1 and
commercially available 4-amino-6-
chloropyrimidine-5-carbonitrile according to Method S. ESI-MS m/z: 451.2
[M+H]'.
Example 143
CI
N
I I
ci CF3 0 el CF3 0
CF3 0 el M-3
NF
NH
NH
FJH2 N N "1
CI N 0 N
III-9 III-43a III-43
[00915] Amine 111-9 was prepared according to Method Q. Compound III-43a
was prepared from amine
111-9 and M-3 according to Method S. Compound III-43a was converted to 111-43
according to the following
procedure:
[00916] To a stirred solution of quinolinone III-43a (0.1 mmol, 1.0 eq) in
THF (10 mL) at RT, sodium
methoxide (0.2 mmol, 2 eq) was added and the resulting mixture was stirred at
reflux for 1-2 h. The mixture was
237

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
allowed to cool to RT and then extracted with DCM (15 mL x 3). The combined
organic layers were washed with
brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo
and the residue was purified by
column chromatography on silica gel (1-2% Me0H/DCM) to afford compound 111-43.
ESI-MS m/z: 510.2 [M+H]
Example 144
CI
N CN
CF3 0
0F3 0 H2N N
I01Hinge-1
N NH2
FIH2
NC
III-1
111-44
[00917] Compound 111-44 was prepared from amine III-1 and 2-amino-4-
chloropyrimidine-5-carbonitrile
(Hinge-1) according to Method S. ESI-MS m/z: 451.2 [M+H]
Example 145
CI
N'CN
CF3 0
CF3 0 H2N N
N
Hinge-1
/
N NH2
NH2
NC
111-9
111-45
[00918] Compound 111-45 was prepared from amine 111-9 and 2-amino-4-
chloropyrimidine-5-carbonitrile
(Hinge-1) according to Method S. ESI-MS m/z: 469.2 [M+H]
Example 146
CI
CF3 0
CF3 0 H2N
101 H-2
N NH2
FIH2
F3C
111-9
111-46
[00919] Compound 111-46 was prepared from amine 111-9 and 4-chloro-5-
(trifluoromethyl)pyrimidin-2-
amine (H-2) according to Method S. ESI-MS m/z: 512.0 [M+H]
238

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 147
C F3 0 40) C F3 0 0 C F3 0 0
0 ; F N F N F
z RH
NH NH _11,,õ.
-Dm.
N N N IN N N
CI ' N Bn0 N HO N
III-43a III-47a III-47
[00920] Compound III-43a was converted to compound III-47a in analogous
fashion to compound 111-43
in Example 143 except that sodium benzyloxide was used in place of sodium
methoxide. Compound 111-47 was
prepared from III-47a according to the following procedure:
[00921] To a solution of III-47a (280 mg, 0.478 mmol) in Me0H (30 mL) at
RT, Pd/C (150 mg, 10%) was
added and the resulting mixture was stirred under hydrogen overnight. The
mixture was filtered and the filtrate was
concentrated in vacuo to afford compound 111-47. ESI-MS m/z: 496.1 [M+H] '.
Example 148
C F3 0 401
N F C F3 0 Si
0 /
CI CIN F
111-9 N- H2
Nj.---, N N ----N
..... -a- .., jzz. ...i.õ NH
CI1 -/k1 N CI N N,
H PMB N-hl
III-48a --",
III-48b 1 .......
CI-N N
PMB
C F3 0 0 C F 3 0 ei C F3 0
N F N F N I. F
/;1H NH NH
III-48 Nk--N III-48dN-N III-48c
Nj\---N
..õ1õ.... ,--....
1
HO N N HO N N, Bn0 N N,
H PMB PMB
[00922] Compound 111-48 was prepared in five steps from 2,6-dichloro-9H-
purine according to the
following procedure: 2,6-dichloro-9H-purine (10 mmol, 1.0 equiv) was dissolved
in 30 mL N,N-
dimethylformamide. Sodium hydride (11 mmol, 1.1 equiv) was added portion-wise
at 0 C and stirred for 10
minutes, after which p-methoxylbenzyl chloride (11 mmol, 1.1 equiv) was added
and the reaction was allowed to
239

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
stir at room temperature overnight. The reaction was then transferred to a
separatory funnel with excess ethyl
acetate and water. The water layer was then extracted with ethyl acetate (3 x
200 mL), dried over Na2SO4, and
concentrated. The mixture was purified by silica gel chromatography, eluting
with 10% ethyl acetate/diethyl ether,
to provide III-48a.
[00923] III-48a was then couped to compound 111-9 using Method S to afford
compound III-48b.
Compound III-48b was then converted to compound III-48e in analogous fashion
to compound 111-43 in Example
143 except that sodium benzyloxide was used in place of sodium methoxide.
Compound III-48e was then
deprotected using analogous conditions as 111-47 in Example 147 to provide III-
48d. Compound III-48d was then
further deprotected to provide 111-48 according the the following procedure:
Compound III-48d was dissolved in
excess TFA and allowed to stir at reflux for 5h, after which it was
concentrated in vacuo. The residue was
resuspended in saturated NaHCO3 and transferred to a separatory funnel where
it was extracted with methylene
chloride (2 x 200 mL). The organic layers were combined, concentrated and
purified using flash silica gel
chromatography (7.5:1 methylene chloride/methanol) to provide 111-48. ESI-MS
m/z: 485.2 [M+H]
Example 149
CI N
F F
CF3 0 I 0
NC
1 NH2
01
111-9 illi2 HNN
111-49 I N
NC
NH2
[00924] Compound 111-49 was prepared from amine 111-9 and 4-amino-6-
chloropyrimidine-5-carbonitrile
according to Method S. ESI-MS m/z: 469.0 [M+H]
Example 150
CI CI 0 401
N
CI 0
110
NH
F4H2 NC I el
1-1 IV-1
[00925] 4-Chloro-7-fluoroquinazoline was coupled with amine I-1 using
Method Ito afford compound IV-
1. ESI-MS m/z: 445.2 [M+H]
240

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 151
ci CI 0 0 CI 0 SI
N N N F N F
CI 0 001 L L
¨N¨cl 0 0
0 ; F Z
NH
NH2 L L
¨lil ¨CI N NH2
IV-2a IV-2b IV-2
[00926] Amine IV-2a was prepared using Method E. It was then coupled with
2,4-dichloro-1,3,5-triazine
using Method I to provide compound IV-2b. Compound IV-2b was converted to
compound IV-2 according to the
following procedure: Compound IV-2b (71 mg, 0.16 mmol) was suspended in
ammonium hydroxide (30 mL) in a
sealed tube and the resulting mixture was stirred at 140 C overnight. The
mixture was allowed to cool to RT and
then extracted with DCM (15 mL x 3). The combined organic layers were washed
with brine, dried over Na2SO4
and filtered. The filtrate was concentrated in vacuo and the residue was
purified by column chromatography on
silica gel (1-2% Me0H/DCM) to afford compound IV-2 (20 mg, 29% yield). ESI-MS
m/z: 411.0 [M+H] '.
Example 152
Cl CI 0 0
0 N
F
' N N
CI 0 /
F -
N 0 ISI NH
________________________________________ )...
1101 F
- N '
FIFi2 I
NSF
1-1 IV-3
[00927] 4-Chloro-6,7-difluoroquinazolinewas coupled with amine I-1 using
Method Ito afford compound
IV-3. ESI-MS m/z: 463.2 [M+H] '.
Example 153
CI
C el
N I 0
' N"--
CI 0 0 N N
A-4 0 /
,
0 _N: NH
NH2

.)..---..-N
1-1 1V-4
241

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00928] Amine I-1 was prepared using Method E. It was then coupled with
compound (A-4) using
Method I to provide compound IV-4. ESI-MS m/z: 416.1 [M+H]
Example 154
CI
CI 0 el
I N
CI 0 101
B-4
NH
RI-12 I
1-1 1V-5
[00929] Compound B-4 was coupled with amine I-1 using Method Ito afford
compound IV-5. ESI-MS
m/z: 428.2 [M+H]
Example 155
CI õN
N CI 0
CI 0
C-3
HN,
Fl H2
1-1
IV-6
[00930] Compound C-3 was coupled with amine I-1 using Method Ito afford
compound IV-6. ESI-MS
m/z: 416.2 [M+H]
Example 156
N CI 0
CI 0 N Ns/
HN N N
F1H2
1-1 IV-7
[00931] Compound IV-7 was prepared from amine I-1 by coupling to 2-chloro-
7H-purine using Method I.
ESI-MS m/z: 417.2 [M+H]
242

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 157
0 H
CI 0 0
NH2 0 ;
C I 0 0
N
)
1-IFI
NH2
NH2
1-1 N
IV-8
[00932] Compound IV-8 was prepared from amine I-1 and commercially
available 2-
aminonicotinaldehyde according to the following procedure: To a stirred
mixture of compound I-1 (150 mg, 0.5
mmol) and 2-aminonicotinaldehyde (37 mg, 0.6 mmol) in Me0H (5 mL), acetic acid
(0.2 mL) and NaBH3CN (79
mg, 1.25 mmol) were added sequentially. The resulting mixture was stirred at
RT for 16 h and LC-MS showed
about 60% conversion. Additional amounts of 2-aminonicotinaldehyde (18 mg, 0.3
mmol) and NaBH3CN (40 mg,
0.62 mmol) were added and the resulting mixture was stirred for 16 h. The
reaction was quenched by aqueous
NaHCO3 solution, and then extracted with ethyl acetate. The combined organic
layer was washed with brine, dried
over Na2SO4 and filtered. The filtrate was concentrated in vacuo and the
residue was purified by ISCO (12 g silica
gel cartridge, 0-10% Me0H/DCM) to afford compound IV-8 (65 mg, 32% yield). ESI-
MS m/z: 405.0 [M+H] '.
Example 158
00
CI HjY0' 0
Pd/C, H2 c N CI N. CO2Et L1AIH4 OHDess Martine =
' _)... el = N .---c- _,... r .,N = N
\
N
Et3N, Me0H N Et0H THF /---N DCM
/1.'----N
NH2 NH2
0
. = pi ""\-
C I 0 lel
CI 0 40 0 )
NH2 N-N''''')..
C)=N
1-1 IV-9
[00933] To a stirred solution of 6-chloropyridazin-3-amine (12.9 g, 0.1
mol) and triethylamine (11.2 g,
0.11 mmol) in Me0H (20 mL), Pd/C (10%, 1.3 g) was added. The resulting mixture
was degassed and back-filled
with hydrogen (three cycles) and then stirred at RT under hydrogen atmosphere
for 18 h. The resulting mixture was
filtered and the filtrate was concentrated in vacuo to afford pyridazin-3-
amine, which was used in the next step
without further purification.
243

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00934] A mixture of pyridazin-3-amine (9.51 g, 0.1 mol) and 2- ethyl 2-
chloro-3-oxopropanoate (22.5 g,
0.15 mol) in Et0H (20 mL) was stirred at reflux overnight. The mixture was
allowed to cool to RT and
concentrated in vacuo to remove Et0H. The residue was partitioned between
water (100 mL) and ethyl acetate (100
mL). The organic layer was separated. The aqueous layer was extracted with
ethyl acetate (2 x 50 mL). The
combined organic layers were washed with brine, dried over Na2SO4 and
filtered. The filtrate was concentrated in
vacuo. The residue was slurried in isopropyl ether (50 mL) with stirring for
30 min, collected by filtration and dried
in vacuo to afford ethyl imidazo[1,2-b]pyridazine-3-carboxylate (7.8 g, 40.8%
yield).
[00935] To a solution of ethyl imidazo[1,2-b]pyridazine-3-carboxylate
(1.91 g, 10 mmol) in anhydrous
THF (20 mL), lithium aluminium hydride (1.34 g, 35 mmol) was added in portions
over 15 min. After stirring at RT
for 16 h, the mixture was quenched with ice-water (20 mL) and then extracted
with ethyl acetate (30 mL x 2). The
combined organic layers were washed with brine, dried over Na2SO4 then
filtered. The filtrate was concentrated in
vacuo to afford imidazo[1,2-b]pyridazin-3-ylmethanol (0.85 g, 57% yield).
[00936] A mixture of imidazo[1,2-b]pyridazin-3-ylmethanol (45 mg, 0.3
mmol) and Dess-Martin
periodinane (130 mg, 0.3 mmol) in anhydrous DCM (5 mL) were stirred at RT
overnight. The recation mixture was
quenched with water (5 mL). The organic layer was separated, dried over MgSO4
and filtered. The filtrate was
concentrated in vacuo to afford imidazo[1,2-b]pyridazine-3-carbaldehyde (40
mg, 90.7% yield).
[00937] To a stirred mixture of compound I-1 (60 mg, 0.2 mmol) and
imidazo[1,2-b]pyridazine-3-
carbaldehyde (30 mg, 0.2 mmol) in DCM (5 mL), acetic acid (24 mg, 0.4 mmol)
and triacetoxyborohydride (85 mg,
0.42 mmol) were added respectively. After stirring at RT for 2 h, the mixture
was quenched with water (10 mL).
The organic layer was separated, dried over MgSO4 and filtered. The filtrate
was concentrated in vacuo to afford
(S)-8-chloro-3-(1-(imidazo[1,2-b]pyridazin-3-ylmethylamino)ethyl)-2-
phenylisoquinolin-1(2H)-one (IV-9) (75 mg,
87.2% yield). ESI-MS m/z: 430.2 [M+H]
Example 159
CI N
C
CI NTh
NCF3I "
CI NH2
1-1/1N
'N
CF3
147.1H 2 NH2
IV-10a IV-10
[00938] Compound IV-10a was coupled with 2-chloro-5-
(trifluoromethyl)pyrimidin-4-amine using
Method Ito afford compound IV-10. ESI-MS m/z: 481.2 [M+H]
244

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 160
Ci CI BrCI CHO CI
CI
'OH CN
k 11,
N N N N N N m
N-
H
CI CN
CI 0
N
CI 0 Op
NH CN
NH2 ,N
N N
1-1
111-11
[00939] To a suspension of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (3.99 g,
26.0 mmol, 1.0 eq) in dry DCM
(150 mL) under argon, N-bromosuccinimide (6.02 g, 33.8 mmol, 1.3 eq) was added
and the resulting mixture was
stirred at RT for 3 h. The reaction mixture was diluted with Me0H (30 mL) and
then concentrated in vacuo to yield
a slight brown solid. The residue was triturated with H20 (150 mL). The solid
was collected by filtration and then
re-crystallized in Me0H to afford 5-bromo-4-chloro-7H-pyrrolo[2,3-d]
pyrimidine (4.0 g, 66% yield).
[00940] To a solution of 5-bromo-4-chloro-7H-pyrrolo[2,3-d]pyrimidine
(2.33 g, 10.0 mmol, 1.0 eq) in
anhydrous THF (100 mL) at -78 C under argon, n-BuLi solution (2.5 M in THF,
8.8 mL, 22.0 mmol, 2.2 eq) was
added dropwise (over 10 min). The resulting mixture was stirred at -78 C for
lh and then DMF (2.0 g, 11.0 mmol,
1.1 eq) was added dropwise (over 10 min). The mixture was stirred at -78 C
for an additional 30 min and then
stirred at RT overnight. The reaction mixture was quenched with H20 (50 mL)
and then concentrated in vacuo. The
residue was triturated with saturated aqueous NH4C1 solution. The solid was
collected by filtration, rinsed with
ethyl acetate, and dried in vacuo to afford 4-chloro-7H-pyrrolo[2,3-
d]pyrimidine-5-carbaldehyde (1.17 g, 65%
yield).
[00941] To a suspension of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine-5-
carbaldehyde (1.17 g, 6.47 mmol, 1.0
eq) and hydroxylamine hydrochloride (0.54 g, 7.77 mmol, 1.2 eq) in Et0H (25
mL), aqueous NaOH solution (0.31
g, 7.77 mmol, 1.2 eq) in H20 (4 mL) was added dropwise. The resulting mixture
was stirred at RT for 30 min and
then was diluted with a sufficient amount of Et0H to allow stirring for
additional 30 min. The solid was collected
by filtration, rinsed with H20 and dried in vacuo to afford 4-chloro-7H-
pyrrolo[2,3-d]pyrimidine-5-carbaldehyde
oxime (0.89 g, 70% yield) as a mixture of isomers. ESI-MS m/z: 194.8 [M-H].
[00942] To a suspension of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine-5-
carbaldehyde oxime (865 mg, 4.40
mmol, 1.0 eq) in DCM (20 mL), thionyl chloride (3.1 mL, 43.7 mmol, 10.0 eq)
was added and the resulting mixture
was stirred at RT overnight. The reaction mixture was concentrated in vacuo.
The residue was suspended in water
(60 mL) and saturated aqueous NaHCO3 was added to adjust the pH to 4. The
solid was collected by filtration,
rinsed with water followed by ethyl acetate to afford the first batch of
product. The filtrate was then extracted with
ethyl acetate (50 mL x 3). The combined organic layer was washed with brine,
dried over Na2SO4 and filtered. The
245

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
filtrate was concentrated in vacuo and the residue was combined with the above-
obtained solid. The crude product
was then re-crystallized in ethyl acetate/hexanes (1:1) and dried in vacuo to
afford 4-chloro-7H-pyrrolo[2,3-
d]pyrimidine-5-carbonitrile (763 mg, 97% yield). ESI-MS m/z: 178.8 [M+H] 176.8
[M-H].
[00943] 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile was coupled
with amine I-1 using Method I
to afford compound IV-11. ESI-MS m/z: 4420.2 [M-H].
Example 161
0
NH2CH2CH(OMe)2 CCI3CN/THF TFA
Et0H
-70 C to -50 C -30 C to RT H conc.H2SO4
4h overnight Ref lux
NH
0
.AcOH OH CI
H2NOSO3H ___________________ CV0---\ NH2 N POCI3 N
'
K2CO3/H20 NH 2 Et0H Reflux
NN
90 C Reflux/overnight
CI 0
CI 0
011
NH
N
NIA2 LN N
1-1
1V-12
[00944] To a stirred solution of trichloroacetonitrile (28.8 g, 200 mmol,
1 eq) in anhydrous THF (70 mL)
at -60 C under an argon atmosphere, 2,2-dimethoxyethanamine (21.8 mL, 200
mmol, 1.0 eq) was added dropwwase
over 5 min. The resulting mixture was allowed to warm to RT and stirred at RT
for 4 h. The mixture was
concentrated in vacuo and the residue was added in portions to a stirred
solution of trifluoroacetic acid (100 mL) at -
30 C under argon. The resulting mixture was then stirred from -30 C to RT
overnight. The reaction mixture was
concentrated in vacuo to afford 2-(trichloromethyl)-1H-imidazole. The crude
product was used in the next step
without further purification.
[00945] The above-obtained residue was dissolved in Et0H (300 mL). To this
solution, conc. H2SO4
(98%, 30 mL, 522 mmol, 2.76 eq) was added dropwise while keeping the reaction
temperature below 25 C. The
resulting mixture was stirred at reflux for 7 h and then stirred at RT
overnight. The mixture was concentrated in
vacuo to remove Et0H. The resulting suspension was diluted with ice-water (200
mL) and neutralized with
concentrated ammonium hydroxide to adjust the pH to 5-6 while keeping the
temperature below 5 C. The solid
was collected by filtration, rinsed with water (10 mL x 3), and dried in vacuo
to afford the first batch of crude
product (10 g). The filtrate was extracted with ethyl acetate (200 mL x 2).
The combined organic layer was washed
with brine, dried over Na2SO4 and filtered. The filtrate was concentrated in
vacuo. The resulting residue was
246

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
combined with the first batch crude product and then re-crystallized in
isopropyl ether to afford ethyl 1H-imidazole-
2-carboxylate (18 g, 64.3% yield).
[00946] To a stirred solution of hydroxylamine-o-sulfonic acid (26.64 g,
235.8 mmol, 3.0 eq) in H20 (17
mL) at 0 C, ethyl 1H-imidazole-2-carboxylate (11.0 g, 78.6 mmol, 1.0 eq) was
added and the resulting mixture was
stirred at 90 C for 30 min. The mixture was cooled to RT and K2CO3 (3.6 g,
26.2 mmol, 1.0 eq) was added in
portions. The resulting mixture was stirred at RT overnight, filtered and
rinsed with H20 (10 mL x 3). The filtrate
was extracted with ethyl acetate (50 mL x 5). The combined organic layer was
washed with brine, dried over
Na2SO4 and filtered. The filtrate was evaporated in vacuo and the residue was
purified by flash column
chromatography on silica gel (1% Me0H in DCM) to afford ethyl 1-amino-1H-
imidazole-2-carboxylate (800 mg,
6.5 % yield). ESI-MS m/z: 156.1 [M+H]
[00947] A mixture of ethyl 1-amino-1H-imidazole-2-carboxylate (800 mg,
5.16 mmol, 1.0 eq) and
formamidine acetate (2.68 g, 25.78 mmol, 5.0 eq) in Et0H (100 mL) was stirred
at reflux overnight. The resulting
mixture was cooled to RT. The solid was collected by filtration, rinsed with
Et0H (3 x 2 mL) and petroleum ether
(2 mLx 3), and then dried in vacuo to afford imidazo[1,2-f][1,2,4]triazin-4-ol
(400 mg, 50.2% yield).
[00948] Imidazo[1,2-f][1,2,4]triazin-4-ol (400 mg, 2.94 mmol, 1.0 eq) was
dissolved in POC13(10 mL,
109.2 mmol, 37.1 eq) and the resulting mixture was stirred at reflux for 2 h.
The mixture was concentrated in vacuo
to remove POC13. The residue was poured into ice water (30 mL) and neutralized
with saturated aqueous NaHCO3
solution to adjust the pH to 6-7 while keeping the temperature below 5 C. The
mixture was extracted with ethyl
acetate (30 mL x 4). The combined organic layer was washed with brine, dried
over Na2SO4 and filtered. The
filtrate was concentrated in vacuo and the residue was purified by flash
column chromatography on silica gel (16%
ethyl acetate in petro ether) to afford 4-chloroimidazo[1,2-f][1,2,4]triazine
(300 mg, 66.0 % yield).
[00949] 4-Chloroimidazo[1,2-f][1,2,4]triazine was coupled with amine I-1
using Method Ito afford
compound IV-12. ESI-MS m/z: 417.2 [M+H]+.
Example 162
CI
Nr'1\
CI 0 el
CI 0
N
HF1 N
1,1H2 Ii
1-19 N
IV-13
[00950] 4-Chloroimidazo[1,2-f][1,2,4]triazine was coupled with amine 1-19
using Method Ito afford
compound IV-13. ESI-MS m/z: 435.0 [M+H]+.
247

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 163
CI
N A
NN CI 0
CI 0 401
H 1;1 N
NI- H2
rN,N
1-19
IV-14
[00951] 4-Chloroimidazo[5,14][1,2,4]triazine was coupled with amine 1-19
using Method I to afford
compound IV-14. ESI-MS m/z: 435.0 [M+H]+.
Example 164
CI 0SN40)
Me
N CI
NN
IV-19
CI
[00952] Compound IV-19 was isolated as a byproduct of the coupling
reaction to provide compound IV-
20. ESI-MS m/z: 499.12 [M+H]
Example 165
CI CI 0
CI 0 el
Me
N N CI
-
HN- N I I CI
.7.
H 2
NN
II
1-1
IV-20
[00953] Amine I-1 (300 mg, 1.0 equiv) was dissoved in dioxane (5 mL). 2,4-
Dichloropteridine (1.5 equiv)
and diisopropylethyl amine were dissolved in 5 mL dioxance and added slowly to
amine I-1. The reaction was
heated to 60 C for 72h after which point there was no starting material by
LC/MS analysis. The reaction was
transferred to a separatory funnel with excess dichloromethane. The organic
layer was washed with saturated
sodium bicarbonate (2x), brine (1x) and water (1x), dried over sodium sulfate
and concentrated to provide crude
material which was purified using flash silica gel chromatography (gradient of
50-100% ethyl acetate/hexanes) to
provide cmpound IV-20. ESI-MS m/z: 463.13 [M+H]
248

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 166
CI 0 CI 0 ei
Me Me
HNCI
T HN CI
N ON
NH2
IV-23
IV-21
[00954] A 35 mL heavy-walled tube with PTFE stopper and PTFE-jacketed-
silicone o-ring seal was
charged with isoquinolinone IV-23 (0.23 mmol) and 1,4-dioxane (10 mL). The
suspension was treated with
ammonium hydroxide solution (10 mL), and the tube sealed tightly and placed in
a 110 C bath overnight. After
cooling, LC/MS showed a mixture of products and < 10% remaining starting
material. The reaction mixture was
dilluted with 2 volumes of brine after which a solid is formed that was
collected via vacuum filtration to provide
amide IV-21. ESI-MS m/z: 453.17 [M+H]
Example 167
CI 0 el CI 0
Me Me
FINT N CI FINNNH2
Nyµc
NN
I Ii
IV-20 IV-22
[00955] A 35 mL heavy-walled tube with PTFE stopper and PTFE-jacketed-
silicone o-ring seal was
charged with isoquinolinone IV-20 (243 mg, 0.52 mmol) and 1,4-dioxane (10 mL).
The suspension was treated
with ammonium hydroxide solution (10 mL), and the tube sealed tightly and
placed in a 140 C bath overnight.
After cooling, LC/MS showed complete conversion and the reaction mixture was
transferred to a larger flask using
1-2 mLs of Me0H, then diluted with 2 volumes of brine. After stirring about
lh, the resulting precipitate was
collected by filtration, washing with water. The product cake was dried in
vacuo to give compound IV-22 as a tan
powder. ESI-MS m/z: 444.20 [M+H]
Example 168
CI
CI 0 40)
CI 0 el
Me
Me H
if
NH
2
1-1
IV-23
249

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00956] Compound I-1 was reacted with 4,6-dichloronicotinonitrile
according to Method I to provide
compound IV-23. ESI-MS m/z: 435.11 [M+H]
Example 169
CI 0 CI 0
Me Me
HN CIRH H, ,N 2
ON ON
IV-21 NH2 IV-24 NH2
[00957] A 35 mL heavy-walled tube with PTFE stopper and PTFE-jacketed-
silicone o-ring seal was
charged with isoquinolinone IV-21 (0.14 mmol) and 1,4-dioxane (4 mL). The
suspension was treated with
ammonium hydroxide solution (30%, 5 mL), and the tube sealed tightly and
placed in a 150 C bath for 3d. The
mixture was then recharged with 5 mL of ammonium hydroxide (30%, 5mL) and
heated for 24 additional hours at
170 C. The reaction mixture transferred to a separtory funnel with excess
methylene chloride. The organic layer
was washed with brine (1x) and water (1x), dried over Na2SO4 and concentrated
to provide material which is 85% of
compound IV-24 and 15% of starting material which was used directly for
biochemical profiling. ESI-MS m/z:
434.18 [M+H]
Example 170
O NH2 Me0
II CI 0
EtOLN Me0
I
CI 0 ei
CI 0 el
B,
HO' OH N
NH
NH
NH2 NN
y, NN
NH2 I II
1-1
CO2Et NH2
CO2Et
IV-25a IV-25
[00958] Compound IV-25a was prepared from compound I-1 by coupling to
ethyl 4-amino-2-
chloropyrimidine-5-carboxylate using Method I. Compound IV-25a was then
converted to compound IV-25
according to the following procedure: The mixture of compound IV-25a (300 mg,
0.65 mmol) and 2-
methoxypyridin-4-yl)boronic acid (149 mg, 0.98 mmol) in 1,4-dioxane:water
(3:1, 12 mL) was degassed and
backfilled with argon (three cycles). To this mixture, 2-dicyclohexylphosphino-
2',6'-diisopropoxybiphenyl (121
mg, 0.26 mmol), palladium(II) acetate (29 mg, 0.13 mmol), and Na2CO3 (207 mg,
1.95 mmol) were added
sequentially. The resulting mixture was degassed and backfilled with argon
(three cycles), and then stirred at 120 C
for 3 h. The mixture was allowed to cool to RT, and then partitioned between
ethyl acetate and water. The organic
layer was washed with brine, dried over Na2SO4 and filtered. The filtrate was
concentrated in vacuo and the residue
250

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
was purified by ISCO column chromatography (silica gel cartridge, 0-10%
Me0H/DCM) to afford compound IV-25
(160 mg, 46% yield). ESI-MS m/z: 537.2 [M+H]
Example 171
0 CI
CI 0 CI 0
0, 0
,LNS -CI Me Me
HN N
Me CI NYCI
NH2 11
1-1 N H2N.µ" N
IV-26a IV-26b
CI 0 CI 0
CI 0 CI
Cl>1 JCI<
CI 0 0 CI So Me (110 Me
11
N 0 FITNr NH2
N
IV-26c IV-26
[00959] Compound I-1 was reacted with 2,4-dichloro-5-nitropyrimidine
according to Method Ito provide
compound IV-26a. Compound IV-26a was then converted to compound IV-26 in 3
steps according to the
following procedures: Compound IV-26a (450 mg, 1.0 equiv) was dissoved in 30
mL of methanol and purged with
N2. Palladium on carbon (1.0 equiv) was added and the reaction was flushed
with H2. The reaction was kept under
an atmosphere of H2 using a ballon and stirred at room temperature for 12h
after which it was filtered through celite
and washed with excess methanol. The solution was concentrated to provide
amine IV-26b which was used directly
in the next step. Compound IV-2611 (68 mg, 1.0 equiv) was dissolved in
tetrahydrofuran (5 mL). Triphosgene was
added (1.2 equiv), follwed by triethylamine (3.0 equiv) and the reaction was
heated to 50 C for 16h. The reaction
was cooled and transferred to a separatory funnel with excess ethyl acetate.
The organic layer was washed with
brine (1x), dried over Na2SO4 and concentrated under vacuum to provide crude
material that was purified using
CombiFlash silica gel chromatography (60% ethyl acetate/hexanes) to provide
compound IV-26c. Compound IV-
26c was then added to a sealed tube (0.08 mmol) and dissolved in 1 mL dioxane.
Ammonium hydoxide (30%, 4
mL) was added and the reaction was sealed and heated to 160 C for 24h after
which it was recharged with 3 mL
additional ammonium hydroxide and heated for 24 additional hours. The reaction
was cooled, concentrated and
purified via HPLC purificaiton to provide compound IV-26. ESI-MS m/z: 433.1
[M+H]
Example 172
CI CI 0 ei
CI 0 ss)'.."2
110 Me
Me
F1H2
S--N
1-1 IV-27
251

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00960] A solution of dichlorothiadiazole (250 mg, 1.62 mmol, 1.1 eq.) in
THF (4 mL) was treated
dropwise with a solution of amine I-1 (440 mg, 1.47 mmol, 1 eq.) and
triethylamine (250 uL, 1.6 mmol, 1.1 eq.) in
THF (6 mL). The reaction was monitored by LC/MS and allowed to proceed for 5d,
at which point most of the
material appeared to be converted and the reaction mixture was diluted with
DCM, 2-3g of silica gel added, and
concentrated in vacuo. Purification of this material by flash chromatography
(gradient of 30-100% ethyl
acetate/hexanes) afforded compound IV-27 as a white solid (540 mg, 88% yield).
ESI-MS m/z: 417.11 [M+H] '.
Table 3. In Vitro IC50 data for selected compounds.
IC50 (nM) Greater than 10 1.IM Greater than
11.IM to 11.IM to 100 nM Less than 100 nM
101.IM
252

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
IC50 (nM) Greater than 10 1.IM Greater than 1
1.IM to 1 1.IM to 100 nM Less than 100 nM
1.IM
PI3K 6 1-53, 1-54, 1-55, I- 1-30, 1-46, 1-52, I- 1-6, 1-27,
1-35, 1-37, 1-2, 1-4, 1-14, 1-16,
62, 1-71, 1-72, 1-77, 68, 1-99, I-100, I- 1-39, 1-93, 1-95,
I- 1-18, 1-20, 1-21, I-
111-26, 111-29, III- 102, 98, I-101, 1-106, 23, 1-24, 1-
25, 1-29,
37, 111-43, 111-47, 11-13, 11-28, 11-29, 11-16, 11-21, 11-
23, 1-33, 1-36, 1-38, I-
111-48, 11-31, 11-32, 11-33, 11-44, 40, 1-
41, 1-42, 1-43,
IV-1, IV-3, IV-4, 111-22, 111-28, 111-14, 111-40, 1-44, 1-45,
1-47, I-
IV-5, IV-6, IV-8, IV-7, IV-9, IV-14, IV-2, IV-21, IV-22
48, 1-49, I-50, I-51,
IV-10, IV-25, IV- IV-19, IV-20, IV-23 1-57, 1-58, 1-59,
I-
26, IV-27 63, 1-65, 1-70, 1-
73,
1-74, 1-75, 1-76, I-
78, 1-79, 1-80, 1-81,
1-82, 1-83, 1-84, I-
85, 1-86, 1-87, 1-88,
1-89, 1-90, 1-91, I-
92, 1-94, 1-96, 1-97,
1-103,
11-2, 11-4, 11-5, 11-7,
11-8, 11-9, II-10, II-
11, 11-14, 11-17, II-
18, 11-20, 11-22, II-
24, 11-30, 11-45, II-
47, 11-48, 11-49, II-
50,
111-3, 111-4, 111-5,
111-6, 111-8, III-10,
III-11, 111-12, III-
13, 111-15, 111-17,
111-18, 111-19, III-
20, 111-21, 111-23,
111-24, 111-25, III-
27, 111-30, 111-31,
111-32, 111-36, III-
39, 111-41, 111-42,
111-44, 111-45, III-
46, 111-49,
IV-11, IV-12, IV-
13, IV-24
253

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
IC50 (nM) Greater than 10 1.IM Greater than 1
1.IM to 1 1.IM to 100 nM Less than 100 nM
101.IM
PI3K y 1-68, 1-71, 1-72, I- 1-6, 1-27, 1-46, 1-53, 1-33, 1-
37, 1-39, I- 1-2, 1-4, 1-14, 1-16,
77, 1-99, 1-54, 1-55, I-100, I- 44, 1-62, 1-65, 1-
73, 1-18, 1-20, 1-21, I-
II-28, 11-29, 106, 1-93, 1-97, 1-98, I- 23, 1-24,
1-25, 1-29,
111-28, 111-47, III- 11-8, 11-13, 11-31, II- 102, 1-30, 1-35, 1-36,
I-
48, 32, 11-33, 11-44, 11-7, 11-9, II-11,
II- 38, 1-40, 1-41, 1-42,
IV-1, IV-3, IV-4, 111-14, 111-26, III- 16, 11-21, 11-23,
II- 1-43, 1-45, 1-47, I-
IV-5, IV-6, IV-7, 29, 111-37, 111-40, 30, 11-45, 11-47,
II- 48, 1-49, I-50, I-51,
IV-8, IV-9, IV-10, 111-41, 111-43, 48, 11-50, 1-52, 1-57, 1-58,
I-
IV-14, IV-19, IV- IV-2, IV-20 111-22, IV-21 59, 1-63, 1-70, 1-
74,
23, IV-25, IV-26, 1-75, 1-76, 1-78,
I-
IV-27 79, 1-80, 1-81, 1-
82,
1-83, 1-84, 1-85, I-
86, 1-87, 1-88, 1-89,
1-90, 1-91, 1-92, I-
94, 1-95, 1-96, I-
101, 1-103,
11-2, 11-4, 11-5, II-
10, 11-14, 11-17, II-
18, 11-20, 11-22, II-
24, 11-49,
111-3, 111-4, 111-5,
111-6, 111-8, III-10,
III-11, 111-12, III-
13, 111-15, 111-17,
111-18, 111-19, III-
20, 111-21, 111-23,
111-24, 111-25, III-
27, 111-30, 111-31,
111-32, 111-36, III-
39, 111-42, 111-44,
111-45, 111-46, III-
49,
IV-11, IV-12, IV-
13, IV-22, IV-24
254

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
IC50 (nM) Greater than 10 1.IM
Greater than 1 1.IM to 1 1.IM to 100 nM Less than 100 nM
101.IM
PI3K cc 1-27, 1-30, 1-33, I- 1-2, 1-4, 1-6, 1-18, I- 1-14, 1-
16, 1-20, I-
35, 1-37, 1-39, 1-41, 21, 1-24, 1-25, 1-29, 23, 1-40, 1-45, 1-58,
1-42, 1-46, 1-52, I- 1-36, 1-38, 1-43, I- 1-63, 1-75, 1-76, I-
53, 1-54, 1-55, 1-57, 44, 1-47, 1-48, 1-49, 81, 1-82, 1-83, 1-85,
1-59, 1-62, 1-65, I- I-50, I-51, 1-70, I- 1-88, 1-90, 1-92, I-
68, 1-71, 1-72, 1-77, 73, 1-74, 1-79, 1-80, 94, 1-103,
1-78, 1-93, 1-95, I- 1-84, 1-86, 1-87, I- II-10,
97, 1-98, 1-99, 1-100, 89, 1-91, 1-96, 1-106, 111-5, 111-17, 111-40,
I-101, 1-102, 11-2, 11-4, 11-5, 11-7, IV-24
11-13, 11-16, 11-21, 11-8, 11-9, II-1 1, II-
11-23, 11-28, 11-29, 14, 11-17, 11-18, 11-
11-30, 11-31, 11-32, 20, 11-22, 11-24, 11-
11-33, 11-44, 11-47, 45, 11-48,
11-49, 11-50, 111-3, 111-4, 111-6,
III-11, 111-14, III- 111-8, III-10, 111-12,
19, 111-21, 111-22, 111-13, 111-15, III-
111-24, 111-26, III- 18, 111-20, 111-23,
28, 111-29, 111-31, 111-25, 111-27, III-
111-32, 111-37, III- 30, 111-36, 111-39,
41, 111-42, 111-43, 111-44, 111-45, III-
111-46, 111-47, III- 49,
48, IV-12, IV-13
IV-1, IV-2, IV-3,
IV-4, IV-5, IV-6,
IV-7, IV-8, IV-9,
IV-10, Iv-11, IV-
14, IV-19, IV-20,
IV-21, IV-22, IV-
23, IV-25, IV-26,
IV-27
255

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
IC50 (nM) Greater than 10 1.IM Greater than 1 1.IM
to 1 1.IM to 100 nM Less than 100 nM
101.IM
PI3K 13 1-6, 1-21, 1-25, 1-27, 1-4, 1-18, 1-24, 1-40, 1-2, 1-
14, 1-16, 1-20, 1-23, 1-92, 1-103,
1-30, 1-35, 1-36, I- I-50, I-51, 1-58, I- 1-29, 1-33, 1-49, I-
II-10,
37, 1-38, 1-39, 1-41, 65, 1-63, 1-70, 1-74, 83, 1-90, 111-5,
111-17,
1-42, 1-43, 1-44, I- 1-75, 1-76, 1-79, I- 11-2, 11-7, II-11,
II- IV-24
45, 1-46, 1-47, 1-48, 81, 1-82, 1-85, 1-88, 14, 11-20, 11-22, II-
1-52, 1-53, 1-54, I- 1-93, 1-94, 1-96, I- 24,
55, 1-57, 1-59, 1-62, 106, 111-3, 111-4, 111-6,
1-68, 1-71, 1-72, I- 11-4, 11-5, 11-8, II- 111-8, III-10, 111-19,
73, 1-77, 1-78, 1-80, 18, 11-28, 11-29, II- 111-20, 111-23, 111-
1-84, 1-86, 1-87, I- 30, 11-44, 11-45, II- 27, 111-30, 111-36,
89, 1-91, 1-95, 1-97, 49, 111-39, 111-40, 111-
1-98, 1-99, I-100, I- III-11, 111-12, III- 44, 111-45, 111-49,
101, 1-102, 13, 111-15, 111-18, IV-12
11-9, 11-13, 11-16, II- 111-21, 111-24, III-
17, 11-21, 11-23, II- 25, 111-31, 111-32,
31, 11-32, 11-33, II- 111-46,
47, 11-48, 11-50, IV-11, IV-13, IV-22
111-14, 111-22, III-
26, 111-28, 111-29,
111-37, 111-41, III-
42, 111-43, 111-47,
111-48,
IV-1, IV-2, IV-3,
IV-4, IV-5, IV-6,
IV-7, IV-8, IV-9,
IV-10, IV-14, IV-
19, IV-20, IV-21,
IV-23, IV-25, IV-
26, IV-27
256

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
IC 50 (nM) Greater than 10 1.IM Greater than 1
1.IM to 1 1.IM to 100 nM Less than 100 nM
1.IM
B cell proliferation 1-99, I-100 1-6, 1-27, 1-52, 1-95, 1-2, 1-
4, 1-14, 1-16,
EC50 (nM)
1-98, 1-18, 1-20, 1-
21, I-
111-46 23, 1-24, 1-25,
1-33,
1-38, 1-40, 1-41, I-
42, 1-43, 1-48, 1-49,
I-50, I-51, 1-57, I-
58, 1-59, 1-63, 1-70,
1-73, 1-74, 1-75, I-
76, 1-78, 1-79, 1-80,
1-81, 1-82, 1-83, I-
84, 1-85, 1-86, 1-87,
1-88, 1-89, 1-90, I-
91, 1-96, 1-97, I-
101,
11-2, 11-4, 11-5, 11-7,
11-9, II-10, II-11,
11-14, 11-17, 11-18,
11-30, 11-45, 11-49,
II-50,
111-3, 111-4, 111-5,
111-6, 111-8, III-10,
III-11, 111-12, III-
13, 111-15, 111-17,
111-27, 111-30, III-
31, 111-36, 111-39,
111-41, 111-42, III-
44, 111-45, 111-49,
IV-2, IV-11, IV-22,
IV-24
257

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Table 4. Structures of the Compounds for the IC50 results described in Table 3
above.
Structure
c i 00 0 0 0 40 CI 0 0
F CI 0
40 ; N
.I
N
0
01N 0N F 0 /
' 1 I;IH E NH
NH F NH F NH N
Ni'l
N N N rsi
, N
N N Compound 1-6 NI
Compound 1-16
Compound 1-2 Compound 1-4 Compound 1-14
o 401 oi o 0 \
\ CI 0
A \
N-N
N-N N
F
0 ; 10/ ; F N
0 0
1.1 N\
0
NA
NH
NH
0 N NH 0 / /
N rsi i
I N N
NH NH
N 1 1
N N rµi N N
rµi
I I
Iµl
Compound 1-18 Compound 1-20 Compound 1-23
N
Compound 1-21 Compound 1-24
\N-NS CI 0
0 01 40
0
\ N
N 0 0 ei NI ------)HN
sS i
N F S NH
. / NH NH NH N l'i
t I
NH Nr%1 N N-- , N', N
1 N 1 /
N I'l N NI
Compound 1-33
L 1 Compound 1-27 Compound 1-29 Compound 1-30
N
Compound 1-25
\N-N \ \ \ \
N-N N-N N-N N-N
\ \ \ \ \
N N N N N
0 0 0 0 0 0
NA
N
0 N lei N = F * N SI F
I. / 0 /
a i t
NH NH NH NH NH
N 1 NI NI NI l%V 1 , I'l N I'l N
1, I'l
1
CI N H2N N CI N H2N N CI N
Compound L35 Compound 1-36 Compound 1-37
Compound 1-38 Compound 1-39
\N-N Me OMe NH2
N
\ N 'N N 'N , -`14
N I I I
0
I / / 0
NA 0 0
/
N 0 6
N .... 0 N 0
N 411
H
H H H 101 ....
. is .... 0 .... Si ....
A N&NII N&N1 6,N % N9
NH N N N
I I I
N NI 1
,L I
H2N N Compound 1-41 Compound 1-42
Compound 1-43 Compound 1-44
Compound 1-40
258

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
OMe
/
N¨N/
N¨N OMe
N CI 0
/ 40
I / N
0 40 , 0 - 0 N 0 1
N 0 0 . ;
H N H
010 ..... H
411 .... 1110 .... N
N i
NSN N511 N.ki
&N
N H
40 .... NH
N N I
lµl
I N511
/ I 1
N
/
Compound 1-47 I
Compound 1-45 Compound 1-46 N
Compound 1-48 Compound 1-
49
N-N CI o 0 CI 0 0 CI 0 0
\
110
N F \ N
H N N
F
0 00 IP ,..- N N CI
0 0
a
IFI N
N
Sri
E E
I;
0 / I NH NH
F1H
xN; NCci N
NC, IN1
I
I I
N
risl
N 1,1
Compound 1-50 1 Compound 1-52 Compound 1-53 Compound 1-
54
Isl-
Compound 1-51
\N-N OMe N ,, OMe N,
HO 0 gib
0
I
\ 1,1"As.'N I0 0 I , N 111111
N I 0 al
110 ..-- N N
0 0 o 0 so
_NH
7
-
0 ....!" Tillii NH2
NU
N so ...: NH i:,N2IN.,,NH2
NyNH2
0
0 / /
a
&N 0-- I
NC t..---
F1H N
I
risl
1
Isl
Compound 1-55 Compound 1-57 Compound 1-58 Compound 1-59 Compound 1-
62
,...L8H2
si 0 0
N N Cl 0 0
NH 0
I {N *
0 0 I I
---..--
l%1 N el F N
N HN N 10I /
NH
5¨ HN N
N&N14 NN NN NH
N
N r%I
I
I
1 0 N
Compound 1-63 Compound 1-65 Compound 1-68 N
Compound 1-70 Compound 1-
71
o o NN N \ N
I I I
o 0
N N N N 0 /
I I 0
I.
/ 0 ; 7
0 N
0 0 N,,r,NH2 N
* lel
6
,,
*
N 4 N I. F
/
/ =

(401 /
a a HNNNH2
HA N
1;1H NH NN
Lli
N
N, Isi NN;
I
0 N N
Compound 1-72 Compound 1-73 Compound 1-74 Compound 1-75 Compound 1-
76
259

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
o 0 N
N"N
N"N
I
I I /
N N N N /
0 40) 0I
I. 0 0
I
40 N
0 0 0 40
F
* N N
N N F 0 /
0 / i
101 / RH RH
FIN3,NT,NH2
E
NH
i N
NH
I
N I
I
I NINI H2N N H2N N
HO N I
H2N N
Compound 1-79 Compound 1-80
Compound 1-81
Compound 1-77
Compound 1-78
N \ 0
1 0 0
N 1.1 N ."-- N N
I I I N F N N
o 0 o al 0 0 0 / I
0
=N
Si N ''''11111.
* N F i
NH
HFLiN:IN,NH2 N
'
HN N NH2 i
.c.i._:- r,
101 N 1:1.1
6; N NH
I 11D- N N NH2 NH
--....... ,...--- r,
NC;
N NH2 I
H2N N
Compound 1-82 Compound 1-83
Compound 1-84 Compound 1-85
Compound 1-86
\N-N N N
, , N CI 0 0
J
, 1 1 N. N
I
0 a 0 0 0 0
0 SI
N N N N
NH
N
0 0
N N . . Ni- 110
a N 1%1 N .
RH NH AH
I i
NH
N, Ni N r%1 N r%1 H2N N NL
I I I c,..-.;-- r,
N H2N N H2N N N NH2
H2N
Compound 1-90
Compound 1-87 Compound 1-88
Compound 1-89 Compound 1-91
CI o 0
NZ: ,H
N 0 0 CI 0 a
Si ; .1.'killiir i
o irk
s,
0 0
40 ," 40
," .1-Liir
141 N NH2 NH ION 0 Me 40 ," N "
N . Y
N N N
NaIN u,
HN N,r,NH 2 H
3,-1, ' 6::
1 N I
6N I
1
Compound 1-95
Compound 1-92 Compound 1-93
Compound 1-94 Compound 1-96
260

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
o' 1 OMe CI 0 0 HN OH I NH 0 0
N ' N
NN I N 'N N
I / I
1101
110 0 ,
N
0 a 0 a
N .'killir / 0 a
i
0 ; Nitillir HN , ,.., OH
110 /N ''''Illlir
/
. I ,N
HNN5NIN NH2
N N N
N I
N; I
H-N TI /
H" 3 '*-
i
-..., N
N
la N
I sr:
Compound I-100
Compound I-101
Compound 1-98
Compound 1-97 Compound 1-99
/ F
--'N 0 a CI 0 0 0
IW

N re N"I'Lllir is ,,,N ri N' 1
I
0 N I /0
I 40
N,..., N
I N
HRJN&NINH2 N
NI-12 40 .... Me 40 .... Me
I
HN N HR N
I
Compound 1-102 Compound 1-103 N",%N Na, N
I
Compound 1-106 Compound 1-107
\N-N CI 0 \
CI 0 0 C I 0 0
\ A N-N
0
lel
N N F N 0 ; N\ 0
/ 0 40
NA
IZ1' 1-1 NH I. ; .
FJH
0 /
Ths1.-N AH Ni "'INJ L
*--NI\
NN N)"--z---.---.) - F1H
lil 1---- --I---> N--.1--->
Compound 11-2
Compound 11-4 NI Compound 11-7
Compound 11-5 'N
Compound 11-8
\N-N CI 0 \
0 CI 0 40
\ CI 0 A \ N-N
N N N
F
0 ei 0
0
0 0
N F
; ; 0NA
* A' 1-1 NHH
/ NH
NH r.,..õN ____.N
NH
rõ,õ.N
r..,-..N NI-N.)
N Compound II
-10
Compound 11-10 II- N,1k1-.1 CI
11 Compound 11-
14
Compound 11-9
Compound 11-13
\N-N \N-N \
N-N CI 0
\
N N N N
0 0 0 ei 0 0
e\--1
N 0
=N F . N F 0 N -) s .
KIH NH
a i
4H 4H NH Ni --14
x
eNA
N''.--/
CI ri,N-...1
N...) Compound 11-20 Compound
11-21
Compound 11-16 Compound 11-17
Compound 11-18
261

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
N -N CI0 CI 0
CI 0
0 ; \ ) 0 N CI
---- 0 0 el io N......,,o 40 N...-...õ_õ.0
s NH NH
E
NH NH
NH
ni cr.õõN
Ni-z----->
1,_...õN
N N N---NI'N.-.1
Compound 11-28 Compound 11-
29
Compound 11-22 Compound 11-23 Compound 11-24
ci 0 40 c i o 0 c i o el \
N-N 0
\
1.1N F 0 N F . N N
CI 0 N)
/ 0 a
NHNHNH
E
0 ,..: -tip- 0 N
N i
N N---N
NH
NH
N
Compound 11-31 Compound 11-32 ,N,N..) N NJ'
Compound 11-30 N))
Compound 11-33
Compound 11-44
\N-N OMe OMe OMe
CI 0 0
\
N N ' N N N N N
0 N
1 1
NA / 0
I. 1 / o
lei 0 /
. / o
lel
101 N N NH N
,
NH 0 / 101 / N-N 0 /
- ,
NH NH N NH
H2N..N.---0- \
N'N....)
/1-- \NI
Compound 11-45
N N-N-,) Compound 11-49 H2N.õ----:.N..--0
Compound 11-47 Compound 11-48 Compound 11-50
F F F FF
F F F F F F CF3 0 el F
0 el 0 0 0 0
N
=N N lel 0 N
N lei
0 / 110 / 0 /
E i E NH E
NH NH NHNH
N1-="N
NCN N r'' NN
1 1 1
N S lµi
W..)
N N N Compound 111-6
H Compound III-4 Compound 111-5
Compound 111-8
Compound 111-3
CF3 0 0 CF3 0 0 CF3 0 40 CF3 0 140 CF3 0 0
0N F 0 N F 110 ; F
0 N F 0 ; F
E
NH NH NH NH RH
N---N INN N Ni
1
N.--S N_> N -*---z..- L 1 j__
CIN"N.)
. N N
H
Compound III-10 Compound III-11 Compound 111-12 Compound
111-13 Compound 111-14
262

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
F F F F
CF3 0 0 F F F F F F F F0
0 0 0 0 0 0
N F N 1.
lel / N N
0
N
111
. 0 /
NH . .
Nnii) -A
NH
NH2
. ,
NH NH F CN
V , \
L I
m
,N.-.1 N.---44 Isdl
11
NN
N------
N N N -
H
Compound 111-20
Compound 111-15 H2N N N N N
H
H H
Compound 111-19
Compound 111-17 Compound 111-18
F F F F F
F F F F F F F F F F
0 0 0 40 0 0 0
0 0
N N N 1.I N I. N Si
/ * /
a a A
NH NH NH RH NH
N N
N lk
I N
' ir - NN N rµi NJ
NI---=---..--.> I I il
CI H2N N CIN S H2N N S
Compound 111-21 Compound 111-22 Compound 111-23 Compound
111-24 Compound 111-25
F F F F F F
F F F F F F F F F
0 SI 0 0 0 0 o o
N N N 0 ; Si N
el F
NH i NH
NH RH
NH
N --'N N
1 r.="N\ Ni-r.'::.-NN
N.-"N
Ikl-N...)¨
j.....
N---S N
H2N N N
Compound 111-26 Compound 111-29
Compound 111-27 Compound 111-28 Compound 111-
30
F F F F F F
0 0
F F F F F F F F
0 0 00 F---..pc el
=N N N 5 NS / 1
S.---/
/ 0 / 101 / 1101 /
a
NH
i 1
RH F NH NH RH
NCN NII
N
F-N N
* I N
1=1 NH2 )--.
N S S
Compound 111-31 Compound 111-32 Compound 111-36 Compound
111-37 Compound 111-39
F
F F F--..... 0 F F F F
F F CF3 0 0 F F
40 jLON 40 0 0
N H
N F 0
;
NH NH 110
.
0 /
, / IkkfN
A
NH '
HF1 N NH2
NN NN N i'l
----N NI'l NH2 ' Ni )Uri
1 ,
H
Compound 111-41 Compound 111-42
I
Compound 111-40 Compound 111-43 Compound
111-44
263

CA 02842187 2014-01-16
WO 2013/012915
PCT/US2012/047186
Structure
F F
F F CF3 0 ei CF3 0 40) CF3 0 0 F F
014 "W
0
N F 10 N F 0 N F 0
,6
.. F
0 /
N el F
. i
HN L N NH2 NH NH NH T1 N H isl-
N
N -
---LCF3 N .---=N
N -
I I j_
HO N
NC N
H2N N HO N N
H
NH2
Compound 111-45 Compound 111-46 Compound 111-47
Compound 111-48 Compound 111-
49
CI 0 a 0, 0 0 ci o a ci 0 0 0, 0 00
1101 N 0 lµl F
0 N
0 ;
0
A A
HN N HN N E
F1H N N
NH
I NH
1),1 i, N
N --- N N
N NH2 F
F Compound IV-2 F Compound IV-4 Compound IV-
5
Compound IV-1 Compound IV-3
(
a o 0 ci o al ci o a 0, 0 0 c,
N'Th
0 ; .11'killr
110 ; .....'11P" 10 N ....iir N
0 / HN
N
FIN,N.,.,r........\ HN N N .
a .
HN HN ?
i,,
UN CF3
&
Compound IV-6 H NH2 NH2
Compound IV-7 I . N cN-N---:. Compound
IV-10
j= N
Compound IV-8
Compound IV-9
CI o 0 CI 0 0 ci 0 0 ci 0 el a o 0
110 N 0 ; so N F so N F N
.1'LliPr
Me
i
FIN N CI
?I'l ION a
NH NH FIN
);):
N--
Is N i-:----N\ N j-:----- \N
I ,N N -:=-N \
N
N-N¨_, N-N_.,j
N N - N-.,
H a
Compound IV-11 Compound IV-12 Compound IV-13 Compound IV-
14 Compound IV-19
CI o 0 ci o 0 CI o 40 0, 0 a CI o 0
10N
/ N .1.killir N
.1.111P.
10N ..., Me olo .... Me . So ... Me 40 ..... Me
,
HN
41 N NH2
HN N CI HN,.....rTrCI 1-11NH2
Xril CI
VI
N' 0.y...-.\õõN o_N
1 I L.- N
N'''
NH2
N
NH2 Compound IV-22 Compound IV-23
Compound IV-20 Compound IV-21 Compound IV-
24
264

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Structure
o N
CI 0 40 ci 0
0
Me
Me 4111112-F.
N N NH2 HAN,CI
HN N 0
S-N
Compound IV-26 Compound IV-27
NH2 ch
Compound IV-25
Biolodcal Activity Assessment
[00961]
A P13-Kinase HTRFO assay kit (cat No. 33-016) purchased from Millipore
Corporation was
used to screen compounds provided herein. This assay used specific, high
affinity binding of the GRP1 pleckstrin
homology (PH) domain to PIP3, the product of a Class lA or 1B PI3 Kinase
acting on its physiological substrate
PIP2. During the detection phase of the assay, a complex was generated between
the GST-tagged PH domain and
biotinylated short chain PIP3. The biotinylated PIP3 and the GST-tagged PH
domain recruited fluorophores
(Streptavidin-Allophycocyanin and Europium-labeled anti-GST respectively) to
form the fluorescence resonance
energy transfer (FRET) architecture, generating a stable time-resolved FRET
signal. The FRET complex was
disrupted in a competitive manner by non-biotinylated PIP3, a product formed
in the PI3 Kinase assay.
[00962]
PI3 Kinase a, 13, 7 and 6 activity was assayed using the PI3 Kinase HTRFO
assay kit
(catalogue No. 33-016) purchased from Millipore Corporation. Purified
recombinant PI3Ka (catalogue No. 14-602-
K), PI3K13 (catalogue No. 14-603-K), PI3K7 (catalogue No. 14-558-K) and PI3K6
(catalogue No. 14-604-K) were
obtained from Millipore Corporation. Purified recombinant PI3K enzyme was used
to catalyze the phosphorylation
of phosphatidylinositol 4,5-bisphosphate (PIP2 at 10 1.IM) to
phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the
presence of 101.IM ATP. The assay was carried out in 384-well format and
detected using a Perkin Elmer EnVision
Xcite Multilabel Reader. Emission ratios were converted into percent
inhibitions and imported into GraphPad Prism
software. The concentration necessary to achieve inhibition of enzyme activity
by 50% (IC50) was calculated using
concentrations ranging from 20 1.IM to 0.1 nM (12-point curve). IC50 values
were determined using a nonlinear
regression model available in GraphPad Prism 5.
Example 173: Chemical Stability
[00963]
The chemical stability of one or more subject compounds is determined
according to standard
procedures known in the art. The following details an exemplary procedure for
ascertaining chemical stability of a
subject compound. The default buffer used for the chemical stability assay is
phosphate-buffered saline (PBS) at
pH 7.4; other suitable buffers can be used. A subject compound is added from a
100 1.IM stock solution to an
aliquot of PBS (in duplicate) to give a final assay volume of 400 1.IL,
containing 5 1.IM test compound and 1%
DMSO (for half-life determination a total sample volume of 700 1.11 is
prepared). Reactions are incubated, with
shaking, for 24 hours at 37 C; for half-life determination samples are
incubated for 0, 2, 4, 6, and 24 hours.
Reactions are stopped by adding immediately 100 1.11 of the incubation mixture
to 100 1.11 of acetonitrile and
265

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
vortexing for 5 minutes. The samples are then stored at -20 C until analysis
by HPLC-MS/MS. Where desired, a
control compound or a reference compound such as chlorambucil (5 M) is tested
simultaneously with a subject
compound of interest, as this compound is largely hydrolyzed over the course
of 24 hours. Samples are analyzed via
(RP)HPLC-MS/MS using selected reaction monitoring (SRM). The HPLC conditions
consist of a binary LC pump
with autosampler, a mixed-mode, C12, 2 x 20 mm column, and a gradient program.
Peak areas corresponding to the
analytes are recorded by HPLC-MS/MS. The ratio of the parent compound
remaining after 24 hours relative to the
amount remaining at time zero, expressed as percent, is reported as chemical
stability. In case of half-life
determination, the half-life is estimated from the slope of the initial linear
range of the logarithmic curve of
compound remaining (%) vs. time, assuming first order kinetics.
Example 174: Expression and Inhibition Assays of p110a/p85a, p11013/p85a,
p1106/p85a, and pllOy:
[00964]
Class I P13-Ks can be either purchased (p110a/p85a, p11013/p85a, p1106/p85a
from Upstate,
and pl 107 from Sigma) or expressed as previously described (Knight et al.,
2004). IC50 values are measured using
either a standard TLC assay for lipid kinase activity (described below) or a
high-throughput membrane capture
assay. Kinase reactions are performed by preparing a reaction mixture
containing kinase, inhibitor (2% DMSO final
concentration), buffer (25 mM HEPES, pH 7.4, 10 mM MgC12), and freshly
sonicated phosphatidylinositol (100
g/m1). Reactions are initiated by the addition of ATP containing 10 Ci of 7-
32P-ATP to a final concentration of
or 100 M and allowed to proceed for 5 minutes at room temperature. For TLC
analysis, reactions are then
terminated by the addition of 105 1 1N HC1 followed by 160 1 CHC13:Me0H
(1:1). The biphasic mixture is
vortexed, briefly centrifuged, and the organic phase is transferred to a new
tube using a gel loading pipette tip
pre coated with CHC13. This extract is spotted on TLC plates and developed for
3 ¨ 4 hours in a 65:35 solution of n-
propano1:1M acetic acid. The TLC plates are then dried, exposed to a
phosphorimager screen (Storm, Amersham),
and quantitated. For each compound, kinase activity is measured at 10 ¨ 12
inhibitor concentrations representing
two-fold dilutions from the highest concentration tested (typically, 200 M).
For compounds showing significant
activity, IC50 determinations are repeated two to four times, and the reported
value is the average of these
independent measurements.
[00965]
Other commercial kits or systems for assaying P13-K activities are available.
The
commercially available kits or systems can be used to screen for inhibitors
and/or agonists of P13 -Ks including, but
not limited to, PI 3-Kinase a, 0, 6, and y. An exemplary system is PI 3-Kinase
(human) HTRFTm Assay from
Upstate. The assay can be carried out according to the procedures suggested by
the manufacturer. Briefly, the assay
is a time resolved FRET assay that indirectly measures PIP3 product formed by
the activity of a P13-K. The kinase
reaction is performed in a microtiter plate (e.g., a 384 well microtiter
plate). The total reaction volume is
approximately 20 IA per well. In the first step, each well receives 2 IA of
test compound in 20% dimethylsulphoxide
resulting in a 2% DMSO final concentration. Next, approximately 14.5 IA of a
kinase/PIP2 mixture (diluted in 1X
reaction buffer) is added per well for a final concentration of 0.25-0.3 g/m1
kinase and 10 M PIP2. The plate is
sealed and incubated for 15 minutes at room temperature. To start the
reaction, 3.5 IA of ATP (diluted in lx
reaction buffer) is added per well for a final concentration of 10 M ATP. The
plate is sealed and incubated for 1
266

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
hour at room temperature. The reaction is stopped by adding 5 IA of Stop
Solution per well and then 5 IA of
Detection Mix is added per well. The plate is sealed, incubated for 1 hour at
room temperature, and then read on an
appropriate plate reader. Data is analyzed and IC50s are generated using
GraphPad Prism 5.
Example 175: B Cell Activation and Proliferation Assay
[00966] The ability of one or more subject compounds to inhibit B cell
activitation and proliferation is
determined according to standard procedures known in the art. For example, an
in vitro cellular proliferation assay
is established that measures the metabolic activity of live cells. The assay
is performed in a 96 well microtiter plate
using Alamar Blue reduction. Balb/c splenic B cells are purified over a Ficoll-
PaqueTM PLUS gradient followed by
magnetic cell separation using a MACS B cell Isolation Kit (Miletenyi). Cells
are plated in 90 IA at 50,000 cells/well
in B Cell Media (RPMI + 10% FBS + Penn/Strep + 50 1.1M bME + 5 mM HEPES). A
compound provided herein is
diluted in B Cell Media and added in a 10 Ill volume. Plates are incubated for
30 min at 37 C and 5% CO2 (0.2%
DMSO final concentration). A 50 jil B cell stimulation cocktail is then added
containing either 10 jig/ml LPS or 5
jig/ml F(ab')2 Donkey anti-mouse IgM plus 2ng/m1 recombinant mouse IL4 in B
Cell Media. Plates are incubated
for 72 hours at 37 C and 5% CO2. A volume of 15 1.11_, of Alamar Blue reagent
is added to each well and plates are
incubated for 5 hours at 37 C and 5% CO2. Alamar Blue fluoresce is read at
560Ex/590Em, and IC50 or EC50
values are calculated using GraphPad Prism 5.
Example 176: Tumor Cell Line Proliferation Assay
[00967] The ability of one or more subject compounds to inhibit tumor
cell line proliferation can be
determined according to standard procedures known in the art. For instance, an
in vitro cellular proliferation assay
can be performed to measure the metabolic activity of live cells. The assay is
performed in a 96 well microtiter plate
using Alamar Blue reduction. Human tumor cell lines are obtained from ATCC
(e.g., MCF7, U-87 MG, MDA-MB-
468, PC-3), grown to confluency in T75 flasks, trypsinized with 0.25% trypsin,
washed one time with Tumor Cell
Media (DMEM + 10%FBS), and plated in 90 Ill at 5,000 cells/well in Tumor Cell
Media. A compound provided
herein is diluted in Tumor Cell Media and added in a 10 Ill volume. Plates are
incubated for 72 hours at 37 C and
5% CO2. A volume of 101.11_, of Alamar Blue reagent is added to each well and
plates are incubated for 3 hours at 37
C and 5% CO2. Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 values
are calculated using GraphPad
Prism 5.
Example 177: Antitumor Activity in vivo
[00968] The compounds described herein can be evaluated in a panel of
human and murine tumor
models.
Paclitaxel-refractory Tumor Models
1. Clinically-derived Ovarian Carcinoma Model.
267

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[00969] This tumor model is established from a tumor biopsy of an
ovarian cancer patient. Tumor
biopsy is taken from the patient. The compounds described herein are
administered to nude mice bearing staged
tumors using an every 2 days x 5 schedule.
2. A2780Tax Human Ovarian Carcinoma Xenograft (Mutated Tubulin).
[00970] A2780Tax is a paclitaxel-resistant human ovarian carcinoma
model. It is derived from the
sensitive parent A2780 line by co-incubation of cells with paclitaxel and
verapamil, an MDR-reversal agent. Its
resistance mechanism has been shown to be non-MDR related and is attributed to
a mutation in the gene encoding
the beta-tubulin protein. The compounds described herein can be administered
to mice bearing staged tumors on an
every 2 days x 5 schedule.
3. HCT116/VM46 Human Colon Carcinoma Xenograft (Multi-Drug Resistant).
[00971] HCT116NM46 is an MDR-resistant colon carcinoma developed from
the sensitive HCT116
parent line. In vivo, grown in nude mice, HCT116NM46 has consistently
demonstrated high resistance to paclitaxel.
The compounds described herein can be administered to mice bearing staged
tumors on an every 2 days x 5
schedule.
4. M5076 Murine Sarcoma Model
[00972] M5076 is a mouse fibrosarcoma that is inherently refractory to
paclitaxel in vivo. The
compounds described herein can be administered to mice bearing staged tumors
on an every 2 days x 5 schedule.
[00973] One or more compounds as provided herein can be used in
combination other therapeutic
agents in vivo in the multidrug resistant human colon carcinoma xenografts
HCTNM46 or any other model known
in the art including those described herein.
Example 178: Microsome stability assay
[00974] The stability of one or more subject compounds is determined
according to standard
procedures known in the art. For example, stability of one or more subject
compounds is established by an in vitro
assay. For example, an in vitro microsome stability assay is established that
measures stability of one or more
subject compounds when reacting with mouse, rat or human microsomes from
liver. The microsome reaction with
compounds is performed in 1.5 mL Eppendorf tube. Each tube contains 0.1 1.11
of 10.0 mg/ml NADPH; 75 1.11 of
20.0 mg/ml mouse, rat or human liver microsome; 0.4 1.IL of 0.2 M phosphate
buffer, and 425 1.11 of ddH20.
Negative control (without NADPH) tube contains 75 1.11 of 20.0 mg/ml mouse,
rat or human liver microsome; 0.4
1.11 of 0.2 M phosphate buffer, and 525 1.11 of ddH20. The reaction is started
by adding 1.0 1.11 of 10.0 mM tested
compound. The reaction tubes are incubated at 37 C. 100 1.11 sample is
collected into new Eppendorf tube
containing 300 1.11 cold methanol at 0, 5, 10, 15, 30 and 60 minutes of
reaction. Samples are centrifuged at 15,000
rpm to remove protein. Supernatant of centrifuged sample is transferred to new
tube. Concentration of stable
compound after reaction with microsome in the supernatant is measured by
Liquid Chromatography/Mass
Spectrometry (LC-MS).
268

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 179: Plasma stability assay
[00975]
The stability of one or more subject compounds in plasma is determined
according to
standard procedures known in the art. See, e.g., Rapid Commun. Mass Spectrom.,
10: 1019-1026. The following
procedure is an HPLC-MS/MS assay using human plasma; other species including
monkey, dog, rat, and mouse are
also available. Frozen, heparinized human plasma is thawed in a cold water
bath and spun for 10 minutes at 2000
rpm at 4 C prior to use. A subject compound is added from a 400 1.IM stock
solution to an aliquot of pre-warmed
plasma to give a final assay volume of 400 1.IL (or 800 1.IL for half-life
determination), containing 5 1.IM test
compound and 0.5 % DMSO. Reactions are incubated, with shaking, for 0 minutes
and 60 minutes at 37 C, or for
0, 15, 30, 45 and 60 minutes at 37 C for half life determination. Reactions
are stopped by transferring 50 1.IL of the
incubation mixture to 200 1.IL of ice-cold acetonitrile and mixed by shaking
for 5 minutes. The samples are
centrifuged at 6000 x g for 15 minutes at 4 C and 120 1.IL of supernatant
removed into clean tubes. The samples are
then evaporated to dryness and submitted for analysis by HPLC-MS/MS.
[00976]
In one embodiment, one or more control or reference compounds (5 1.IM) are
tested
simultaneously with the test compounds: one compound, propoxycaine, with low
plasma stability and another
compound, propantheline, with intermediate plasma stability.
[00977]
Samples are reconstituted in acetonitrile/methanol/water (1/1/2, v/v/v) and
analyzed via
(RP)HPLC-MS/MS using selected reaction monitoring (SRM). The HPLC conditions
consist of a binary LC pump
with autosampler, a mixed-mode, C12, 2 x 20 mm column, and a gradient program.
Peak areas corresponding to the
analytes are recorded by HPLC-MS/MS. The ratio of the parent compound
remaining after 60 minutes relative to the
amount remaining at time zero, expressed as percent, is reported as plasma
stability. In case of half-life
determination, the half-life is estimated from the slope of the initial linear
range of the logarithmic curve of
compound remaining (%) vs. time, assuming first order kinetics.
Example 180: Kinase Signaling in Blood
[00978]
PI3K/ Akt /mTor signaling is measured in blood cells using the phosflow method
(Methods
Enzymol. (2007) 434:131-54). This method is by nature a single cell assay so
that cellular heterogeneity can be
detected rather than population averages. This allows concurrent dinstinction
of signaling states in different
populations defined by other markers. Phosflow is also highly quantitative. To
test the effects of one or more
compounds provided herein, unfractionated splenocytes, or peripheral blood
mononuclear cells are stimulated with
anti-CD3 to initiate T-cell receptor signaling. The cells are then fixed and
stained for surface markers and
intracellular phosphoproteins. Inhibitors provided herein inhibit anti-CD3
mediated phosphorylation of Akt -S473
and S6, whereas rapamycin inhibits S6 phosphorylation and enhances Akt
phosphorylation under the conditions
tested.
[00979]
Similarly, aliquots of whole blood are incubated for 15 minutes with vehicle
(e.g., 0.1%
DMSO) or kinase inhibitors at various concentrations, before addition of
stimuli to crosslink the T cell receptor
(TCR) (anti-CD3 with secondary antibody) or the B cell receptor (BCR) using
anti-kappa light chain antibody
(Fab'2 fragments). After approximately 5 and 15 minutes, samples are fixed
(e.g., with cold 4% paraformaldehyde)
269

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
and used for phosflow. Surface staining is used to distinguish T and B cells
using antibodies directed to cell surface
markers that are known to the art. The level of phosphorylation of kinase
substrates such as Akt and S6 are then
measured by incubating the fixed cells with labeled antibodies specific to the
phosphorylated isoforms of these
proteins. The population of cells are then analyzed by flow cytometry.
Example 181: Colony Formation Assay
[00980] Murine bone marrow cells freshly transformed with a p190 BCR-Abl
retrovirus (herein
referred to as p190 transduced cells) are plated in the presence of various
drug combinations in M3630
methylcellulose media for about 7 days with recombinant human IL-7 in about
30% serum, and the number of
colonies formed is counted by visual examination under a microscope.
[00981] Alternatively, human peripheral blood mononuclear cells are
obtained from Philadelphia
chromosome positive (Ph+) and negative (Ph-) patients upon initial diagnosis
or relapse. Live cells are isolated and
enriched for CD19+ CD34+ B cell progenitors. After overnight liquid culture,
cells are plated in methocult GF+
H4435, Stem Cell Technologies) supplemented with cytokines (IL-3, IL-6, IL-7,
G-CSF, GM-CSF, CF, F1t3 ligand,
and erythropoietin) and various concentrations of known chemotherapeutic
agents in combination with either
compounds of the present disclosure. Colonies are counted by microscopy 12-14
days later. This method can be
used to test for evidence of additive or synergistic activity.
Example 182: In Vivo Effect of Kinase Inhibitors on Leukemic Cells
[00982] Female recipient mice are lethally irradiated from a 7 source in
two doses about 4 hr apart,
with approximately 5Gy each. About 1 hr after the second radiation dose, mice
are injected i.v. with about 1 x106
leukemic cells (e.g., Ph+ human or murine cells, or p190 transduced bone
marrow cells). These cells are
administered together with a radioprotective dose of about 5x106 normal bone
marrow cells from 3-5 week old
donor mice. Recipients are given antibiotics in the water and monitored daily.
Mice who become sick after about 14
days are euthanized and lymphoid organs are harvested for analysis. Kinase
inhibitor treatment begins about 10 days
after leukemic cell injection and continues daily until the mice become sick
or a maximum of approximately 35 days
post-transplant. Inhibitors are given by oral lavage.
[00983] Peripheral blood cells are collected approximately on day 10
(pre-treatment) and upon
euthanization (post treatment), contacted with labeled anti-hCD4 antibodies
and counted by flow cytometry. This
method can be used to demonstrate that the synergistic effect of one or more
compounds provided herein in
combination with known chemotherapeutic agents can reduce leukemic blood cell
counts as compared to treatment
with known chemotherapeutic agents (e.g., Gleevec) alone under the conditions
tested.
Example 183: Treatment of Lupus Disease Model Mice
[00984] Mice lacking the inhibitory receptor Fc7RIIb that opposes PI3K
signaling in B cells develop
lupus with high penetrance. Fc7RIIb knockout mice (R2KO, Jackson Labs) are
considered a valid model of the
270

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
human disease as some lupus patients show decreased expression or function of
Fc7RIIb (S. Bolland and J.V.
Ravtech 2000. Immunity 12:277-285).
[00985]
The R2K0 mice develop lupus-like disease with anti-nuclear antibodies,
glomerulonephritis
and proteinurea within about 4-6 months of age. For these experiments, the
rapamycin analogue RAD001 (available
from LC Laboratories) is used as a benchmark compound, and administered
orally. This compound has been shown
to ameliorate lupus symptoms in the B6.Slelz.S1e3z model (T. Wu et al. J. Clin
Invest. 117:2186-2196).
[00986]
The NZB/W Fl mice spontaniously develop a systemic autoimmune disease with
that is a
model of lupus. The mice are treated starting at 20 weeks of age for a
profilactic model and at 23 weeks of age for a
therapeutic model. Blood and urine samples are obtained at approximately
throughout the testing period, and tested
for antinuclear antibodies (in dilutions of serum) or protein concentration
(in urine). Serum is also tested for anti-
ssDNA and anti-dsDNA antibodies by ELISA.. Glomerulonephritis is assessed in
kidney sections stained with H&E
at the end of the study, or survival can be an endpoint. For example, tThe
proteozome inhibitor Bortezimib is
effective at blocking disease in the NZB/W model in both the profilactic and
therapeuctic model with reductions in
auto-antibody production, kidney damage, and improvements in survival (Nature
Medicine 14, 748 - 755 (2008)).
[00987]
Lupus disease model mice such as R2KO, BXSB or MLR/lpr are treated at about 2
months
old, approximately for about two months. Mice are given doses of: vehicle,
RAD001 at about 10 mg/kg, or
compounds provided herein at approximately 1 mg/kg to about 500 mg/kg. Blood
and urine samples are obtained at
approximately throughout the testing period, and tested for antinuclear
antibodies (in dilutions of serum) or protein
concentration (in urine). Serum is also tested for anti-ssDNA and anti-dsDNA
antibodies by ELISA. Animals are
euthanized at day 60 and tissues harvested for measuring spleen weight and
kidney disease. Glomerulonephritis is
assessed in kidney sections stained with H&E. Other animals are studied for
about two months after cessation of
treatment, using the same endpoints.
[00988]
This established art model can be employed to demonstrate that the kinase
inhibitors provided
herein can suppress or delay the onset of lupus symptoms in lupus disease
model mice.
Example 184: Murine Bone Marrow Transplant Assay
[00989]
Female recipient mice are lethally irradiated from a 7 ray source. About lhr
after the radiation
dose, mice are injected with about 1 x106 leukemic cells from early passage
p190 transduced cultures (e.g., as
described in Cancer Genet Cytogenet. 2005 Aug;161(1):51-6) . These cells are
administered together with a
radioprotective dose of approximately 5x106 normal bone marrow cells from 3-
5wk old donor mice. Recipients are
given antibiotics in the water and monitored daily. Mice who become sick after
about 14 days are euthanized and
lymphoid organs harvested for flow cytometry and/or magnetic enrichment.
Treatment begins on approximately day
and continues daily until mice become sick, or after a maximum of about 35
days post-transplant. Drugs are
given by oral gavage (p.o.). In a pilot experiment a dose of chemotherapeutic
that is not curative but delays leukemia
onset by about one week or less is identified; controls are vehicle-treated or
treated with chemotherapeutic agent,
previously shown to delay but not cure leukemogenesis in this model (e.g.,
imatinib at about 70mg/kg twice daily).
For the first phase p190 cells that express eGFP are used, and postmortem
analysis is limited to enumeration of the
271

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
percentage of leukemic cells in bone marrow, spleen and lymph node (LN) by
flow cytometry. In the second phase,
p190 cells that express a tailless form of human CD4 are used and the
postmortem analysis includes magnetic
sorting of hCD4+ cells from spleen followed by immunoblot analysis of key
signaling endpoints: p Akt -T308 and
S473; pS6 and p4EBP-1. As controls for immunoblot detection, sorted cells are
incubated in the presence or absence
of kinase inhibitors of the present disclosure inhibitors before lysis.
Optionally, "phosflow" is used to detect p Akt -
S473 and pS6-S235/236 in hCD4-gated cells without prior sorting. These
signaling studies are particularly useful if,
for example, drug-treated mice have not developed clinical leukemia at the 35
day time point. Kaplan-Meier plots of
survival are generated and statistical analysis done according to methods
known in the art. Results from p190 cells
are analyzed separated as well as cumulatively.
[00990] Samples of peripheral blood (100-2001.11) are obtained weekly
from all mice, starting on day
immediately prior to commencing treatment. Plasma is used for measuring drug
concentrations, and cells are
analyzed for leukemia markers (eGFP or hCD4) and signaling biomarkers as
described herein.
[00991] This general assay known in the art can be used to demonstrate
that effective therapeutic
doses of the compounds provided herein can be used for inhibiting the
proliferation of leukemic cells.
Example 185: Matrigel Plug Angiogenesis Assay
[00992] Matrigel containing test compounds are injected subcutaneously
or intraocularly, where it
solidifies to form a plug. The plug is recovered after 7-21 days in the animal
and examined histologically to
determine the extent to which blood vessels have entered it. Angiogenesis is
measured by quantification of the
vessels in histologic sections. Alternatively, fluorescence measurement of
plasma volume is performed using
fluorescein isothiocyanate (FITC)-labeled dextran 150. The results are
expected to indicate one or more compounds
provided herein that inhibit angiogenesis and are thus expected to be useful
in treating ocular disorders related to
aberrant angiogenesis and/or vascular permeability.
Example 186: Corneal Angiogenesis Assay
[00993] A pocket is made in the cornea, and a plug containing an
angiogenesis inducing formulation
(e.g., VEGF, FGF, or tumor cells), when introduced into this pocket, elicits
the ingrowth of new vessels from the
peripheral limbal vasculature. Slow-release materials such as ELVAX (ethylene
vinyl copolymer) or Hydron are
used to introduce angiogenesis inducing substances into the corneal pocket.
Alternatively, a sponge material is used.
[00994] The effect of putative inhibitors on the locally induced (e.g.,
sponge implant) angiogenic
reaction in the cornea (e.g., by FGF, VEGF, or tumor cells). The test compound
is administered orally, systemically,
or directly to the eye. Systemic administration is by bolus injection or, more
effectively, by use of a sustained-
release method such as implantation of osmotic pumps loaded with the test
inhibitor. Administration to the eye is by
any of the methods described herein including, but not limited to eye drops,
topical administration of a cream,
emulsion, or gel, intravitreal injection.
[00995] The vascular response is monitored by direct observation
throughout the course of the
experiment using a stereomicroscope in mice. Definitive visualization of the
corneal vasculature is achieved by
272

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
administration of fluorochrome-labeled high-molecular weight dextran.
Quantification is performed by measuring
the area of vessel penetration, the progress of vessels toward the angiogenic
stimulus over time, or in the case of
fluorescence, histogram analysis or pixel counts above a specific (background)
threshold.
[00996] The results can indicate one or more compounds provided herein
inhibit angiogenesis and
thus can be useful in treating ocular disorders related to aberrant
angiogenesis and/or vascular permeability.
Example 187: Microtiter-plate Angiogenesis Assay
[00997] The assay plate is prepared by placing a collagen plug in the
bottom of each well with 5-10
cell spheroids per collagen plug each spheroid containing 400-500 cells. Each
collagen plug is covered with 1100 Ill
of storage medium per well and stored for future use (1-3 days at 37 C, 5%
CO2). The plate is sealed with sealing.
Test compounds are dissolved in 200 Ill assay medium with at least one well
including a VEGF positive control and
at least one well without VEGF or test compound as a negative control. The
assay plate is removed from the
incubator and storage medium is carefully pipeted away. Assay medium
containing the test compounds are pipeted
onto the collagen plug. The plug is placed in a humidified incubator for (37
C, 5% CO2) 24-48 hours. Angiogenesis
is quantified by counting the number of sprouts, measuring average sprout
length, or determining cumulative sprout
length. The assay can be preserved for later analysis by removing the assay
medium, adding lml of 10%
paraformaldehyde in Hanks BSS per well, and storing at 4 C. The results are
expected to identify compounds that
inhibit angiogenesis in various cell types tested, including cells of ocular
origin.
Example 188: Combination use of PI3K-6 inhibitors and agents that inhibit IgE
production or activity
[00998] The compounds as provided herein can present synergistic or
additive efficacy when
administered in combination with agents that inhibit IgE production or
activity. Agents that inhibit IgE production
include, for example, one or more of TEI-9874, 2-(4-(6-cyclohexyloxy-2-
naphtyloxy)phenylacetamide)benzoic acid,
rapamycin, rapamycin analogs (i.e., rapalogs), TORC1 inhibitors, TORC2
inhibitors, and any other compounds that
inhibit mTORC1 and mTORC2. Agents that inhibit IgE activity include, for
example, anti-IgE antibodies such as
Omalizumab and TNX-901.
[00999] One or more of the subject compounds capable of inhibiting PI3K-
6 can be efficacious in
treatment of autoimmune and inflammatory disorders (AIID), for example,
rheumatoid arthritis. If any of the
compounds causes an undesired level of IgE production, one can choose to
administer it in combination with an
agent that inhibits IgE production or IgE activity. Additionally, the
administration of PI3K-6 or PI3K-43/7 inhibitors
as provided herein in combination with inhibitors of mTOR can also exhibit
synergy through enhanced inhibition of
the PI3K pathway. Various in vivo and in vitro models can be used to establish
the effect of such combination
treatment on AIID including, but not limited to (a) in vitro B-cell antibody
production assay, (b) in vivo TNP assay,
and (c) rodent collagen induced arthritis model.
(a) B-cell Assay
[001000] Mice are euthanized, and the spleens are removed and dispersed
through a nylon mesh to
generate a single-cell suspension. The splenocytes are washed (following
removal of erythrocytes by osmotic shock)
273

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
and incubated with anti-CD43 and anti-Mac-1 antibody-conjugated microbeads
(Miltenyi Biotec). The bead-bound
cells are separated from unbound cells using a magnetic cell sorter. The
magnetized column retains the unwanted
cells and the resting B cells are collected in the flow-through. Purified B-
cells are stimulated with
lipopolysaccharide or an anti-CD40 antibody and interleukin 4. Stimulated B-
cells are treated with vehicle alone or
with PI3K-6 inhibitors as provided herein with and without mTOR inhibitors
such as rapamycin, rapalogs, or
mTORC1/C2 inhibitors. The results are expected to show that in the presence of
mTOR inhibitors (e.g., rapamycin)
alone, there is little to no substantial effect on IgG and IgE response.
However, in the presence of PI3K-6 and
mTOR inhibitors, the B-cells are expected to exhibit a decreased IgG response
as compared to the B-cells treated
with vehicle alone, and the B-cells are expected to exhibit a decreased IgE
response as compared to the response
from B-cells treated with PI3K-6 inhibitors alone.
(b) TNP Assay
[001001]
Mice are immunized with TNP-Ficoll or TNP-KHL and treated with: vehicle, a
PI3K-6
inhibitor, an mTOR inhibitor, for example rapamycin, or a PI3K-6 inhibitor in
combination with an mTOR inhibitor
such as rapamycin. Antigen-specific serum IgE is measured by ELISA using TNP-
BSA coated plates and isotype
specific labeled antibodies. It is expected that mice treated with an mTOR
inhibitor alone exhibit little or no
substantial effect on antigen specific IgG3 response and no statistically
significant elevation in IgE response as
compared to the vehicle control. It is also expected that mice treated with
both PI3K-6 inhibitor and mTOR
inhibitor exhibit a reduction in antigen specific IgG3 response as compared to
the mice treated with vehicle alone.
Additionally, the mice treated with both PI3K-6 inhibitor and mTOR inhibitor
exhibit a decrease in IgE response as
compared to the mice treated with PI3K-6 inhibitor alone.
(c) Rat Collagen Induced Arthritis Model
[001002]
Female Lewis rats are anesthetized and given collagen injections prepared and
administered
as described previously on day 0. On day 6, animals are anesthetized and given
a second collagen injection. Caliper
measurements of normal (pre-disease) right and left ankle joints are performed
on day 9. On days 10-11, arthritis
typically occurs and rats are randomized into treatment groups. Randomization
is performed after ankle joint
swelling is obviously established and there is good evidence of bilateral
disease.
[001003]
After an animal is selected for enrollment in the study, treatment is
initiated. Animals are
given vehicle, PI3K-6 inhibitor, or PI3K-6 inhibitor in combination with
rapamycin. Dosing is administered on days
1-6. Rats are weighed on days 1-7 following establishment of arthritis and
caliper measurements of ankles taken
every day. Final body weights are taken on day 7 and animals are euthanized.
[001004]
The combination treatment using a compound as provided herein and rapamycin
can provide
greater efficacy than treatment with PI3K-6 inhibitor alone.
[001005]
Certain compounds provided herein (e.g., Compounds 1-21, 1-86, 1-87, 1-91, and
111-13) were
tested in the rat Collagen Induced Arthritis Model using procedures
substantially similar to those described above,
and all of the tested compounds demonstrated EC50 values of less than 50
mg/kg.
274

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
Example 189: Delayed Type Hypersensitivity Model
[001006] DTH was induced by sensitizing 60 BALB/c male mice on day 0 and
day 1 with a solution of
0.05% 2,4 dinitrofluorobenzene (DNFB) in a 4:1 acetone/olive oil mixture. Mice
were gently restrained while
201.IL of solution was applied to the hind foot pads of each mouse. The hind
foot pads of the mice were used as
they represent an anatomical site that can be easily isolated and immobilized
without anesthesia. On day 5, mice
were administered a single dose of vehicle, a compound provided herein at 10,
3, 1, or 0.3 mg/kg, or dexamethasone
at a dose of 5 mg/kg by oral gavage. Thirty minutes later mice were
anaesthetized, and a solution of 0.25% DNFB
in a 4:1 acetone/olive oil solution was applied to the left inner and outer
ear surface. This application resulted in the
induction of swelling to the left ear and under these conditions, all animals
responded to this treatment with ear
swelling. A vehicle control solution of 4:1 acetone/olive oil was applied to
the right inner and outer ear. Twenty
four hours later, mice were anaesthetized, and measurements of the left and
right ear were taken using a digital
micrometer. The difference between the two ears was recorded as the amount of
swelling induced by the challenge
of DNFB. Drug treatment groups were compared to vehicle control to generate
the percent reduction in ear
swelling. Dexamethasone is routinely used as a positive control as it has
broad anti-inflammatory activity.
Example 190: Peptidoglycan-Polysaccharide rat Arthritic Model
(a) Systemic arthritis model
[001007] All injections are performed under anesthesia. 60 female Lewis
rats (150-170) are
anesthetized by inhalation isoflurane using a small animal anesthesia machine.
The animals are placed in the
induction chamber until anesthetized by delivery of 4-5% isoflurane in 02 and
then held in that state using a nose
cone on the procedure table. Maintenance level of isoflurane is at 1-2%.
Animals are injected intraperitoneally (i.p.)
with a single injection of purified PG-PS 10S Group A, D58 strain
(concentration 251.tg/g of bodyweight) suspended
in sterile 0.85% saline. Each animal receives a total volume of 500
microliters administered in the lower left
quadrant of the abdomen using a 1 milliliter syringe with a 23 gauge needle.
Placement of the needle is critical to
avoid injecting the PG-PS 10S into either the stomach or caecum. Animals are
under continuous observation until
fully recovered from anesthesia and moving about the cage. An acute response
of a sharp increase in ankle
measurement, typically 20% above baseline measurement can peak in 3-5 days
post injection. Treatment with test
compounds can be PO, SC, IV or IP. Rats are dosed no more than two times in a
24 hour time span. Treatment can
begin on day 0 or any day after that through day 30. The animals are weighed
on days 0, 1, 2, 3, 4, 5, 6, 7 and
beginning again on day 12 ¨ 30 or until the study is terminated. Paw/ankle
diameter is measured with a digital
caliper on the left and right side on day 0 prior to injection and again on
day 1, 2, 3, 4, 5, 6 and 7. On day 12,
measurements begin again and continue on through day 30. At this time, animals
can be anesthetized with
isoflurane, as described above, and terminal blood samples can be obtained by
tail vein draws for the evaluation of
the compound blood levels, clinical chemistry or hematology parameters.
Animals are them euthanized with carbon
dioxide overdose. A thoracotomy can be conducted as a means of death
verification.
(b) Monoarticular arthritis model
275

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[001008]
All injections are performed under anesthesia. 60 female Lewis rats (150-170)
are
anesthetized by inhalation isoflurane using a small animal anesthesia machine.
The animals are placed in the
induction chamber until anesthetized by delivery of 4-5% isoflurane in 02 and
then held in that state using a nose
cone on the procedure table. Maintenance level of isoflurane is at 1-2%.
Animals are injected intra-articular (i.a.)
with a single injection of purified PG-PS 100P Group A, D58 strain
(concentration 50Oug/mL) suspended in sterile
0.85% saline. Each rat receives a total volume of 10 microliters administered
into the tibiotalar joint space using a 1
milliliter syringe with a 27 gauge needle. Animals are under continuous
observation until fully recovered from
anesthesia and moving about the cage. Animals that respond 2-3 days later with
a sharp increase in ankle
measurement, typically 20% above baseline measurement on the initial i.a.
injection, are included in the study. On
day 14, all responders are anesthetized again using the procedure previously
described. Animals receive an
intravenous (I.V.) injection of PG-PS (concentration 250uL/mL). Each rat
receives a total volume of 400 microliters
administered slowly into the lateral tail vein using a 1 milliliter syringe
with a 27 gauge needle. Baseline ankle
measurements are measured prior to IV injection and continue through the
course of inflammation or out to day 10.
Treatment with test compounds will be PO, SC, IV or IP. Rats are dosed no more
than two times in a 24 hour time
span. Treatment can begin on day 0 or any day after that through day 24. The
animals are weighed on days 0, 1, 2,
3, 4, 5, and beginning again on day 14 ¨ 24 or until the study is terminated.
Paw/ankle diameter is measured with a
digital caliper on the left and right side on day 0 prior to injection and
again on day 1, 2, 3, 4, 5, and beginning again
on day 14 ¨ 24 or until the study is terminated. At this time, animals can be
anesthetized with isoflurane, as
described above, and terminal blood samples can be obtained by tail vein draws
for the evaluation of the compound
blood levels, clinical chemistry or hematology parameters. Animals are them
euthanized with carbon dioxide
overdose. A thoracotomy can be conducted as a means of death verification.
Example 191: Mice Models for Asthma
[001009]
Efficacy of a compound provided herein in treating, preventing and/or managing
asthma can be
assessed using an conventional animal models including various mice models
described in, for example, Nials et al.,
Dis Model Mech. 1(4-5): 213-220 (2008).
(a) Acute Allergen Challenge Models
[001010]
Several models are known in the art and any of such models can be used.
Although various
allergens can be used to induce asthma-like conditions, the principle is
consistent throughout the methods. Briefly,
asthma-like conditions are induced through multiple systemic administration of
the allergen (e.g., ova, house dust
mite extracts and cockroach extracts) in the presence of an adjuvant such as
aluminum hydroxide. Alternatively, an
adjuvant-free system can be used, but it usually requires a higher number of
exposures to achieve suitable
sensitization. Once induced, animals exhibit many key features of clinical
asthma such as: elevated levels of IgE;
airway inflammation; goblet cell hyperplasia; epithelial hypertrophy; AHR ro
specific stimuli; and early and late
phase bronchoconstriction. Potential efficacy of a compound thus can be
assessed by determining whether one or
more of these clinical features are reversed or mitigated.
(b) Chronic Allergen Challenge Models
276

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[001011] Chronic allergen challenge models aim to reproduce more of the
features of the clinical
asthma, such as airway remodeling and persistent AHR, than acute challenge
models. While allergens similar to
those used in acute allergen challenge models can be used, in chronic allergen
challenge models, animals are
subjected to repeated exposure of the airways to low levels of allergen for a
period of up to 12 weeks. Once
induced, animals exhibit key features of human asthma such as: allergen-
dependent sensitization; a Th2-dependent
allergic inflammation characterized by eosinophillic influx into the airway
mucosa; AHR; and airway remodeling as
evidenced by goblet cell hyperplasia, epithelial hypertrophy, subepithelial or
peribronchiolar fibrosis. Potential
efficacy of a compound thus can be assessed by determining whether one or more
of these clinical features are
reversed or mitigated.
Example 192: Models for Psoriasis
[001012] Efficacy of a compound provided herein in treating, preventing
and/or managing psoriasis can
be assessed using an conventional animal models including various animal
models described in, for example,
Boehncke et al., Clinics in Dermatology, 25: 596-605 (2007).
[001013] As an example, the mouse model based on adoptive transfer of CD4
'CD45RBhi T cells
described in Hong et al., J. Immunol., 162: 7480-7491 (1999) can be made.
Briefly, female BALB/cBY (donor) and
C.B.-17/Prkdc scid/scid (recipient) mice are housed in a specific pathogen-
free environment and are used between 6
and 8 weeks of age. CD4 T cells are enriched from BALB/cBy splenocytes using a
mouse CD4 enrichment kit.
The cells are then labeled with PE-conjugated anti-CD4, FITC-conjugated anti-
CD45RB, and APC-conjugated anti-
CD25 antibodies. Cells are sorted using a cell sorter. CD4 tD45RBhiCD25 cells
are collected. Cells are
resuspended in saline and 4x108 cells/mouse are injected i.p. into C.B.-
17/Prkdc scid/scid mice. Mice may be dosed
with LPS, cytokines, or antibodies as necessary. Mice are monitored for
external signs of skin lesions twice each
week. After the termination, ear, back skin, lymph nodes and spleen may be
collected for further ex vivo studies.
Example 193: Models for Scleroderma
[001014] A compound's efficacy in treating scleroderma can be tested using
animal models. An
exemplary animal model is a mouse model for scleroderma induced by repeated
local injections of bleomycin
("BLM") described, for example, in Yamamoto et al., J Invest Dermatol 112: 456-
462 (1999), the entirety of which
is incorporated herein by reference. This mouse model provides dermal
sclerosis that closely resembles systemic
sclerosis both histologically and biochemically. The sclerotic changes
observed in the model include, but are not
limited to: thickened and homogenous collagen bundles and cellular filtrates;
gradual increase in number of mast
cells; degranulation of mast cells; elevated histamine release; increase in
hydroxyproline in skin; presence of anti-
nuclear antibody in serum; and strong expression of transforming growth factor
3-2 mRNA. Therefore, efficacy of a
compound in treating scleroderma can be assessed by monitoring the lessening
of one or more of these changes.
[001015] Briefly, the following exemplary procedures can be used to
generate the mouse model for
scleroderma: Specific pathogen-free, female BALB/C mice and C3H mice of 6
weeks old, weighing about 20 g, are
purchased and maintained with food and water ad libitum. BLM is dissolved in
PBS at differing concentrations and
277

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
sterilized with filtration. Aliquots of each concentration of BLM or PBS are
injected subcutaneously into the shaved
back of the mice daily for 1-4 weeks with a needle. Alternatively, mice are
injected every other day.
[001016] Histolopathological and biochemical changes induced can be
assessed using any methods
commonly practiced in the field. For example, histopathological changes can be
assessed using a standard avidine-
biotin peroxidase technique with anti-L3T4 monoclonal antibody, anti-Lyt2
monoclonal antibody, anti-mouse pan-
tissue-fixed macrophage antibody, anti-stem cell factor monoclonal antibody,
anti-transforming growth factor-I3
polyclonal antibody, and anti-decorin antibody. Cytokine expression of
cellular infiltrates can be assessed by using
several anti-cytokine antibodies. Hydroxyproline level can be assessed by
hydrolyzing skin pieces with
hydrochloric acid, neutralizing with sodium hydroxide, and colorimetrically
assessing the hydrolates at 560 nm with
p-dimethylaminobenzaldehyde. Pepsin-resistant collagen can be assessed by
treating collagen sample extracted
from biopsied tissues and analyzing by polyacrylamide stacking gel
electrophoresis. Mast cells can be identified by
toluidine blue, and cells containing matachromatic granules can be counted
under high magnification of a light
microscope. Serum levels of various cytokines can be assessed by enzyme-linked
immunosorbent assay, and
mRNA levels of the cytokines can be assessed by reverse-transcriptase
polymerase chain reaction. Autoantibodies
in serum can be detected using 3T3 fibroblasts as the substrate for the
screening.
Example 194: Models for Myositis
[001017] A compound's efficacy in treating myositis (e.g.,
dermatomyositis) can be tested using animal
models known in the art. One such example is the familial canine
dermatomyositis model described in Hargis et al.,
AJP 120(2): 323-325 (1985). Another example is the rabbit myosin induced mouse
model described in Phyanagi et
al., Arthritis & Rheumatism, 60(10): 3118-3127 (2009).
[001018] Briefly, 5-week old male SJL/J mice are used. Purified myosin
from rabbit skeletal muscle
(6.6 mg/ml) is emulsified with an equal amount of Freund's complete adjuvant
and 3.3 mg/ml Mycobacterium
butyricum. The mice are immunized repeatedly with emulsified rabbit myosin.
Once myositis is induced,
inflammatory cell filtration and necrotic muscle fiber should be evident in
the model. In the muscles of animals,
CD4 T cells are mainly located in the perimysum and CD8 T cells are mainly
located in the endomysium and
surround non-necrotic muscle fibers. TNFa, IFN7 and perforin are up-regulated
and intercellular adhesion molecule
1 is increased in the muscles.
[001019] To assess the efficacy of a compound, following administration of
the compound through
adequate route at specified dose, the mice are killed and muscle tissues are
harvested. The muscle tissue is
immediately frozen in chilled isopentane precooled in liquid nitrogen, and
then cryostat sections are prepared. The
sections are stained with hematoxylin and eosin for counting of number of
infiltrated cells. Three sections from
each mouse are prepared and photomicrographs are obtained. For
immunohistochemical tests, cryostat sections of
muscle are dried and fixed in cold acetone at -20 C. The slides are rehydrated
in PBS, and then endogeneous
peroxide activity is blocked by incubation in 1% hydrogen peroxide. The
sections are incubated overnight with rat
anti-mouse CD4 monoclonal antibody, rat anti-mouse CD8 monoclonal antibody,
rat anti-mouse F4/80 monoclonal
antibody or normal rat IgG in antibody diluent. The samples are washed with
PBS and incubated with biotin-
278

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
conjugated rabbit anti-rat IgG pretreated with 5% normal mouse serum. After
washing with PBS, the samples are
incubated with streptavidin-horseradish peroxidase. After washing PBS,
diaminobenzidine is used for visualization.
Example 195: Models for Sjogren Syndrome
[001020] A compound's efficacy in treating Sjogren's syndrome can be
tested using animal models
known in the art, for example, those described in Chiorini et al., Journal of
Autoimmunity 33: 190-196 (2009).
Examples include: mouse model spontaneously developed in first filial
generation of NZB mice crossed to NZW
mice (see, e.g., Jonsson et al., Clin Immunol Immunopathol 42: 93-101 (1987);
mouse model induced by i.p.
injection of incomplete Freund's adjuvant (id.; Deshmukh et al., J Oral Pathol
Med 38: 42-27 (2009)); NOD mouse
models wherein Sjogren's phenotype is developed by specific genotypes (see,
e.g., Cha et al., Arthritis Rheum 46:
1390-1398 (2002); Kong et al., Clin Exp Rheumatol 16: 675-681 (1998); Podolin
et al., J Exp Med 178: 793-803
(1993); and Rasooly et al., Clin Immunol Immunopathol 81: 287-292 (1996));
mouse model developed in
spontaneous lpr mutation; mouse model developed in Id3 knock-out mice (see,
e.g., Li et al., Immunity 21: 551-560
(2004)); mouse model developed in PI3K knock-out mice (see, e.g., Oak et al.,
Proc Natl Acad Sci USA 103: 16882-
16887 (2006)); mouse model developed in BAFF over-expressing transgenic mice
(see, e.g., Groom et al., J Clin
Invest 109: 59-68 (2002)); mouse model induced by injection of Ro antigen into
BALB/c mice (see, e.g., Oh-Hora
et al., Nat. Immunol 9: 432-443 (2008)); mouse model induced by injection of
carbonic anhydrase II (see, e.g.,
Nishimori et al., J Immunol 154: 4865-4873 (1995); mouse model developed in IL-
14 over-expressing transgenic
mice (see, e.g., Shen et al., J Immunol 177: 5676-5686 (2006)); and mouse
model developed in IL-12 expressing
transgenic mice (see, e.g., McGrath-Morrow et al., Am J Physiol Lung Cell Mol
Physiol 291: L837-846 (2006)).
Example 196: Models for Immune Complex Mediated Disease
[001021] The Arthus reaction is a type 3 immune response to immune
complexes, and thus, can be a
mechanistic model supporting therapeutic hypothesis for immune complex
mediated diseases such as rheumatoid
arthritis, lupus and other autoimmune diseases. For example, PI3K7 and 6
deficient mice can be used as
experimental models of the Arthus reaction and provide assessment of
therapeutic potential of a compound as to the
treatment of immune complex mediated diseases. The Arthus reaction can be
induced using the following
exemplary procedures as described in Konrad et al., Journal of Biological
Chemistry (2008 283(48): 33296-33303.
[001022] PI3K7- and PI3K6-deficient mice are maintained under dry barrier
conditions. Mice are
anesthetized with ketamine and xylazine, and the trachea is cannulated.
Appropriate amount of protein G-purified
anti-OVA IgG Ab is applied, and appropriate amount of OVA antigen is given
intravenously. For PI3K blocking
experiments, wortmanin is given intratrache ally together with the application
of anti-OVA igG. Mice are killed at 2-
4 hours after initiation of inflammation, and desired follow up assessments
can be performed using methods known
in the art.
Example 197: Isoform-Selective Cellular Assays
(a) PI3K6 Selective Assay
279

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
[001023] A compound's ability in selectively inhibiting PI3K6 can be
assessed using RAJI cells, i.e., B
lymphocyte cells derived from lymphoma patients. Briefly, serum-starved RAJI
cells are stimulated with anti-
human IgM, thereby causing signaling through the B-cell receptors, as
described in, for example, He et al.,
Leukemia Research (2009) 33: 798-802. B-cell receptor signaling is important
for the activation, differentiation,
and survival of B cells and certain B-cell derived cancers. Reduction of
phospho-AKT is indicative of compounds
that may inhibit B-cell proliferation and function in certain diseases. By
monitoring the reduction of phospho-AKT
in stimulated RAJI cells (using for example, phospho-AKT antibodies), a
compound's potential efficacy in
selectively inhibiting PI3K6 can be assessed.
[001024] Certain compounds provided herein (e.g., Compounds 1-21, 1-86, 1-
87, 1-91, 1-106, and 111-13)
were tested in RAJI cell model using procedures as described above. It was
found that IC50 values for phospho-
AKT are as follows: Compounds 1-21, 1-86, 1-87, 1-91, 1-106, and 111-13 in the
range of less than 100 nM.
(b) PI3Ky Selective Assay
[001025] A compound's ability in selectively inhibiting PI3Ky can be
assessed using RAW264.7
macrophages. Briefly, serum-starved PAW264.7 cells are stimulated with a known
GPCR agonist C5a. (See, e.g.,
Camps et al., Nature Medicine (2005) 11(9): 936- 943). Cells can be treated
with test compounds prior to,
simultaneously with, or subsequent to the stimulation by C5a. RAW 264.7 cells
respond to the complement
component fragment C5a through activation of the C5a receptor, and the C5a
receptor activates macrophages and
induces cell migration. Test compounds' ability to inhibit C5a-mediated AKT
phosphorylation is indicative of
selective inhibition of PI3Ky. Thus, by monitoring the reduction of phospho-
AKT in stimulated RAW 264.7 cells
(using for example, phospho-AKT antibodies), a compound's potential efficacy
in selectively inhibiting PI3Ky can
be assessed.
[001026] Certain compounds provided herein (e.g., Compounds 1-21, 1-86, 1-
87, 1-91, 1-106, and 111-13)
were tested in RAW 264.7 cell model using procedures as described above. It
was found that IC50 values for
phospho-AKT are as follows: Compounds 1-86, 1-87, and 1-91 in the range of
less than 100 nM; Compounds 1-21, I-
106, and 111-13 in the range of between 100 nM and 11.IM.
(c) PI3Ka Selective Assay
[001027] A compound's ability in selectively inhibiting PI3Ka can be
assessed using SKOV-3 cells, i. e. ,
human ovarian carcinoma cell line. Briefly, SKOV-3 cells, in which mutant
PI3Ka is constitutively active, can be
treated with test compounds. Test compounds' ability to inhibit AKT
phosphorylation in SKOV-3 cells, therefore,
is indicative of selective inhibition of PI3Ka. Thus, by monitoring the
reduction of phospho-AKT in SKOV-3 cells
(using for example, phospho-AKT antibodies), a compound's potential efficacy
in selectively inhibiting PI3Ka can
be assessed.
[001028] Certain compounds provided herein (e.g., Compounds 1-21, 1-86, 1-
87, 1-91, and 111-13) were
tested in SKOV-3 cell model using procedures as described above. It was found
that IC50 values for phospho-AKT
280

CA 02842187 2014-01-16
WO 2013/012915 PCT/US2012/047186
are as follows: Compound 1-87 in the range of between 100 nM and 1 1.IM;
Compounds 1-21, 1-86, 1-91, and 111-13
in the range of between 1 1.IM and 101.IM.
(d)PI3KP Selective Assay
[001029] A compound's ability in selectively inhibiting PI3KI3 can be
assessed using 786-0 cells, i.e.,
human kidney carcinoma cell line. Briefly, 786-0 cells, in which PI3KI3 is
constitutively active, can be treated with
test compounds. Test compounds' ability to inhibit AKT phosphorylation in 786-
0 cells, therefore, is indicative of
selective inhibition of PI3KI3. Thus, by monitoring the reduction of phospho-
AKT in 786-0 cells (using for
example, phospho-AKT antibodies), a compound's potential efficacy in
selectively inhibiting PI3KI3 can be
assessed.
[001030] Certain compounds provided herein (e.g., Compounds 1-21, 1-86, 1-
87, 1-91, and 111-13) were
tested in 786-0 cell model using procedures as described above. It was found
that IC50 values for phospho-AKT are
as follows: Compounds 1-21, 1-86, 1-87, 1-91, and 111-13 in the range of
between 11.IM and 10 1.IM.
281

Representative Drawing

Sorry, the representative drawing for patent document number 2842187 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2012-07-18
(87) PCT Publication Date 2013-01-24
(85) National Entry 2014-01-16
Dead Application 2018-07-18

Abandonment History

Abandonment Date Reason Reinstatement Date
2017-07-18 FAILURE TO REQUEST EXAMINATION
2017-07-18 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2014-01-16
Maintenance Fee - Application - New Act 2 2014-07-18 $100.00 2014-07-17
Maintenance Fee - Application - New Act 3 2015-07-20 $100.00 2015-07-07
Maintenance Fee - Application - New Act 4 2016-07-18 $100.00 2016-07-11
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
INFINITY PHARMACEUTICALS INC.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2014-01-16 1 68
Claims 2014-01-16 29 930
Description 2014-01-16 281 13,971
Cover Page 2014-02-28 2 34
Prosecution-Amendment 2014-04-14 1 29
PCT 2014-01-16 19 725
Assignment 2014-01-16 4 146