Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2021/050555
Kind Code:
A1
Abstract:
The present invention relates to compositions and methods for the treatment of disorders that lead to accumulation of oxalate, such as primary hyperoxaluria 1.

Inventors:
ZHANG YING (US)
LEE ESTHER (US)
MCRINER ANDREW (US)
HUPP CHRISTOPHER (US)
Application Number:
PCT/US2020/049956
Publication Date:
March 18, 2021
Filing Date:
September 09, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
X CHEM INC (US)
International Classes:
C07D261/18; C07D267/14; C07D403/12
Domestic Patent References:
WO2019165159A12019-08-29
Foreign References:
US20080039442A12008-02-14
US6228985B12001-05-08
US20060276466A12006-12-07
Attorney, Agent or Firm:
DECAMP, James, D. et al. (US)
Download PDF:
Claims:
What is claimed is:

Claims

1 . A compound having the structure of Formula I: wherein

X1 is N or CR1;

X2 is N or CR2;

X3 is N or CR3;

X4 is N or CR4;

X5 is N or CR5;

X6 is N or CR6;

X7 is N or CR7;

X8 is N or CR8;

X9 is N or CR9;

Y is O or NRY; each of R1, R2, R3, R4, and R5 is, independently, H, hydroxyl, halogen, optionally substituted Ci- C6 alkyl, or optionally substituted C1-C6 heteroalkyl;

RY is H or optionally substituted C1-C6 alkyl;

R10 is H, optionally substituted C1-C6 alkyl, or optionally substituted C3-C7 cycloalkyl; each of R11 and R12 is, independently, H or optionally substituted C1-C6 alkyl; each of R6, R7, R13, and R14 is, independently, H, hydroxyl, halogen, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R6 and R13, R13 and R14, or R7 and R14, together with the carbon atoms to which each is attached, combine to form an optionally substituted 5-membered aryl, optionally substituted 5-membered heteroaryl, optionally substituted 6-membered aryl, or optionally substituted 6-membered heteroaryl; and each of R8, R9, R15, and R16 is, independently, H, halogen, hydroxyl, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R8 and R15 or R9 and R16, together with the carbon atoms to which each is attached, combine to form an optionally substituted 5- or 6-membered aryl or heteroaryl, or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1 , wherein Y is O.

3. The compound of claim 1 , wherein Y is NRY.

4. The compound of claim 3, wherein RY is H or C1-C3 alkyl.

5. The compound of claim 4, wherein RY is H or

6. The compound of claim 5, wherein RY is H.

7. The compound of any one of claims 1 to 6, wherein X1 is N.

8. The compound of any one of claims 1 to 6, wherein X1 is CR1.

9. The compound of claim 8, wherein R1 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

10. The compound of claim 9, wherein R1 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

V/CH3

11. The compound of claim 10, wherein R1 is H, hydroxyl, F, Cl, \ ,

12. The compound of claim 11 , wherein R1 is H.

13. The compound of any one of claims 1 to 12, wherein X2 is N.

14. The compound of any one of claims 1 to 12, wherein X2 is CR2.

15. The compound of claim 14, wherein R2 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

16. The compound of claim 15, wherein R2 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

17. The compound of claim 16, wherein R2 is H, hydroxyl, F, Cl,

18. The compound of claim 17, wherein R2 is H.

19. The compound of any one of claims 1 to 18, wherein X3 is N.

20. The compound of any one of claims 1 to 18, wherein X3 is CR3.

21 . The compound of claim 20, wherein R3 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

22. The compound of claim 21 , wherein R3 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

23. The compound of claim 22, wherein R3 is H, hydroxyl, F, Cl, ,

24. The compound of claim 23, wherein R3 is H.

25. The compound of any one of claims 1 to 24, wherein X4 is N.

26. The compound of any one of claims 1 to 24, wherein X4 is CR4.

27. The compound of claim 26, wherein R4 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

28. The compound of claim 27, wherein R4 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

30. The compound of claim 29, wherein R4 is H.

31 . The compound of any one of claims 1 to 30, wherein X5 is N.

32. The compound of any one of claims 1 to 30, wherein X5 is CR5.

33. The compound of claim 32, wherein R5 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

34. The compound of claim 33, wherein R5 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

CH3

35. The compound of claim 34, wherein R5 is H, hydroxyl, F, Cl, Y ^CH3

36. The compound of claim 35, wherein R5 is H.

37. The compound of any one of claims 1 to 36, wherein X2 is CR2; X3 is CR3; X4 is CR4; X5 is

38. The compound of any one of claims 1 to 37, wherein R10 is optionally substituted C1-C6 alkyl or optionally substituted C3-C7 cycloalkyl.

39. The compound of any one of claims 1 to 37, wherein R10 is H or optionally substituted Ci-

C6 alkyl.

40. The compound of any one of claims 1 to 37, wherein R10 is H.

41. The compound of any one of claims 1 to 40, wherein R1 1 is H or optionally substituted Ci-

C3 alkyl.

42. The compound of claim 41 , wherein R1 1 is H or C1-C3 alkyl.

VCH3

43. The compound of claim 42, wherein R1 1 is H or '

44. The compound of claim 43, wherein R1 1 is H.

45. The compound of any one of claims 1 to 44, wherein R12 is H or optionally substituted Ci-

C3 alkyl.

46. The compound of claim 45, wherein R12 is H or C1-C3 alkyl.

YCH3

47. The compound of claim 46, wherein R12 is H or '

48. The compound of claim 47, wherein R12 is H.

49. The compound of any one of claims 1 to 48, wherein R1 1 is H, and R12 is H.

50. The compound of any one of claims 1 to 49, wherein X6 is N.

51. The compound of any one of claims 1 to 49, wherein X6 is CR6.

52. The compound of claim 51 , wherein R6 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

53. The compound of claim 52, wherein R6 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

54. The compound of claim 53, wherein R6 is H, hydroxyl, F, Cl,

55. The compound of claim 54, wherein R6 is H.

56. The compound of any one of claims 1 to 55, wherein X7 is N.

57. The compound of any one of claims 1 to 55, wherein X7 is CR7.

58. The compound of claim 57, wherein R7 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

59. The compound of claim 58, wherein R7 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

CH3

60. The compound of claim 59, wherein R7 is H, hydroxyl, F, Cl, Y

61 . The compound of claim 60, wherein R7 is H.

62. The compound of any one of claims 1 to 61 , wherein R13 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

63. The compound of claim 62, wherein R13 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

64. The compound of claim 63, wherein R13 is H, hydroxyl, F, Cl, ,

65. The compound of claim 64, wherein R13 is H, F, Cl,

66. The compound of any one of claims 1 to 65, wherein R14 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

67. The compound of claim 66, wherein R14 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

68. The compound of claim 67, wherein R14 is H, hydro .xyl, F, yCH3

69. The compound of any one of claims 1 to 68, wherein R14 is H, F, Cl, \ , or

70. The compound of any one of claims 1 to 69, wherein X8 is N.

71 . The compound of any one of claims 1 to 69, wherein X8 is CR8.

72. The compound of claim 71 , wherein R8 is H, hydroxyl, halogen, optionally substituted Ci-

C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

73. The compound of claim 72, wherein R8 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

74. The compound of claim 73, wherein R8 is H, hydroxyl, F, Cl, yCH3 yCF3 y<

75. The compound of claim 74, wherein R8 is H, F, Cl, ' , ' , or ' 3.

76. The compound of any one of claims 1 to 75, wherein X9 is N.

77. The compound of any one of claims 1 to 75, wherein X9 is CR9.

78. The compound of claim 77, wherein R9 is H, hydroxyl, halogen, optionally substituted Ci- C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

79. The compound of claim 78, wherein R9 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

CH 0. The compound of claim 79, wherein R9 is H, hydroxyl, F, Cl, Y 3

8

81 . The compound of claim 80, wherein R9 is H, F, Cl,

82. The compound of any one of claims 1 to 81 , wherein R15 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

83. The compound of claim 82, wherein R15 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl.

CH3

84. The compound of claim 83, wherein R15 is H, hydroxyl, F, Cl, Y V""CH3

85. The compound of claim 84, wherein R15 is H, F, Cl,

86. The compound of any one of claims 1 to 85, wherein R16 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl.

87. The compound of claim 86, wherein R16 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR°1, wherein R°1 is C1-C4 alkyl or Ci-C4 fluoroalkyl. yCH3 ^

88. The compound of claim 87, wherein R16 is H, hydroxyl, F, Cl, ' , ' 3 yCH3

89. The compound of claim 88, wherein R16 is H, F, Cl, '

90. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula la:

Formula la, or a pharmaceutically acceptable salt thereof.

91 . The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula lb: or a pharmaceutically acceptable salt thereof.

92. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula lc:

Formula lc, or a pharmaceutically acceptable salt thereof.

93. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula Id: or a pharmaceutically acceptable salt thereof.

94. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula le: or a pharmaceutically acceptable salt thereof.

95. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula If:

Formula If, or a pharmaceutically acceptable salt thereof.

96. The compound of any one of claims 1 to 89, wherein the compound of Formula I has the structure of Formula Ig:

Formula Ig, or a pharmaceutically acceptable salt thereof.

97. The compound of any one of claims 1 to 96, wherein the compound has the structure of any one of compounds 1-26 in Table 1.

98. A pharmaceutical composition comprising the compound of any one of claims 1 to 97 and a pharmaceutically acceptable excipient.

99. A method of reducing the level of hydroxyacid oxidase 1 (HA01) in a cell, wherein the method comprises contacting the cell with an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

100. A method of inhibiting the activity of HA01 in a cell, wherein the method comprises contacting the cell with an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

101 . The method of claim 99 or 100, wherein the cell is a liver cell.

102. The method of claim 101 , wherein the liver cell is a human liver cell.

103. A method of treating a disorder that leads to accumulation of oxalate in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

104. A method of treating an HA01 -associated disorder in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

105. The method of claim 104, wherein the disorder is primary hyperoxaluria 1 .

106. The method of claim 104, wherein the disorder is primary hyperoxaluria 2.

107. A method of treating primary hyperoxaluria in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

108. The method of claim 107, wherein the primary hyperoxaluria is primary hyperoxaluria 1.

109. The method of claim 107, wherein the primary hyperoxaluria is primary hyperoxaluria 2.

110. A method of reducing the level of oxalate in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

111. A method of reducing the level of glyoxylate in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

112. A method of reducing the level of glycine in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

113. A method of inhibiting accumulation of oxalate in a subject in need thereof, wherein the method comprises administering to the subject an effective amount of a compound of any one of claims 1 to 97, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 98.

114. The method of any one of claims 103 to 113, wherein the subject is human.

115. The method of claim 114, wherein the subject is less than 18 years old.

Description:
COMPOUNDS AND USES THEREOF

Background of the Invention

Oxalate is a metabolic end-product endogenously produced by the liver and filtered by the kidney to be excreted in urine. Oxalate tends to precipitate as insoluble calcium oxalate crystals, which is the main component of kidney stones and bladder stones. Disorders, e.g., primary hyperoxalurias (PHs), can result in the accumulation of oxalate which lead to, e.g., kidney stone disease (KSD), end stage renal disease (ESRD), and renal failure. Although there are some symptomatic treatments, such as surgical removal of kidney stones, at present, there are limited therapies for disorders that lead to oxalate accumulation, e.g., primary hyperoxalurias (PHs). Thus, there is a need to develop new approaches for the treatment of disorders that lead to oxalate accumulation (e.g., primary hyperoxalurias 1) and the symptoms that accompany such disorders (e.g., end stage renal disease).

Summary of the Invention

Primary hyperoxalurias (PHs) are rare autosomal recessive disorders affecting the glyoxylate or hydroxyproline pathways and that result in an overproduction of oxalate. The most common type of PH is primary hyperoxaluria type 1 (PH1), which is caused by mutations in the liver-specific peroxisomal enzyme alanine-glyoxylate aminotransferase (AGT). AGT catalyzes the detoxification (transamination) of glyoxylate in the liver. Glyoxylate is produced via oxidation of glycolic acid (glycolate) by hydroxyacid oxidase 1 (HA01), also known as glycolate oxidase (GO). Loss of AGT function results in accumulation of glyoxylate, which is then oxidized by lactate dehydrogenase (LDH) to form oxalate. The calcium salt of oxalate is insoluble and tends to precipitate as insoluble crystals in tissues. This potentially can result in kidney stones, kidney damage, kidney failure, hematuria, urinary tract infections, end stage renal disease (ESRD), systemic oxalosis, or injury to other organs. In most cases, PH1 eventually leads to ESRD, a life-threatening condition that prevents kidneys from effectively filtering fluids and waste products from the body. Inhibition of HA01 activity may reduce the production of glyoxylate thus reducing levels of toxic oxalate crystals in the kidneys of PH1 patients.

The present disclosure features compounds and methods useful for treating HA01 -associated disorders, for affecting the level and/or activity of HA01 , and/or for treating disorders (e.g., primary hyperoxalurias (e.g., primary hyperoxaluria 1)) that lead to oxalate accumulation and the symptoms (e.g., end stage renal disease) that accompany such disorders.

In an aspect, the disclosure features a compound having the structure of Formula I:

Formula I where X 1 is N or CR 1 ; X 2 is N or CR 2 ; X 3 is N or CR 3 ; X 4 is N or CR 4 ; X 5 is N or CR 5 ; X 6 is N or CR 6 ; X 7 is N or CR 7 ; X 8 is N or CR 8 ; X 9 is N or CR 9 ; Y is O or NR Y ; each of R 1 , R 2 , R 3 , R 4 , and R 5 is, independently, H, halogen, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R Y is H or optionally substituted C1-C6 alkyl; R 10 is H, optionally substituted C1-C6 alkyl, or optionally substituted C3- C7 cycloalkyl; each of R 11 and R 12 is, independently, H or optionally substituted C1-C6 alkyl; each of R 6 ,

R 7 , R 13 , and R 14 is, independently, H, halogen, hydroxyl, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R 6 and R 13 , R 13 and R 14 , or R 7 and R 14 , together with the carbon atoms to which each is attached, combine to form an optionally substituted 5- or 6-membered aryl or heteroaryl; and each of R 8 , R 9 , R 15 , and R 16 is, independently, H, halogen, hydroxyl, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R 8 and R 15 or R 9 and R 16 , together with the carbon atoms to which each is attached, combine to form an optionally substituted 5-membered aryl, optionally substituted 5-membered heteroaryl, optionally substituted 6-membered aryl, or optionally substituted 6- membered heteroaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, Y is O. In some embodiments, Y is NR Y . yCH 3

In some embodiments, R Y is H or C1-C3 alkyl. In some embodiments, R Y is H or ' . In some embodiments, R Y is H.

In some embodiments, X 1 is N. In some embodiments, X 1 is CR 1 . In some embodiments, R 1 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 1 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

V / CH3

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 1 is H, hydroxyl, F, Cl, \ ,

In some embodiments, X 2 is N. In some embodiments, X 2 is CR 2 . In some embodiments, R 2 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 2 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is ^CH 3

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 2 is H, hydroxyl, F, Cl, \ ,

In some embodiments, X 3 is N. In some embodiments, X 3 is CR 3 . In some embodiments, R 3 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 3 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is C 1 -C 4 alkyl or C 1 -C 4 fluoroalkyl. In some embodiments, R 3 is H, hydroxyl, F, Cl, V CHs ^CH 3 ,

In some embodiments, X 4 is N. In some embodiments, X 4 is CR 4 . In some embodiments, R 4 is H, hydroxyl, halogen, optionally substituted C 1 -C 4 alkyl, or optionally substituted C 1 -C 4 heteroalkyl. In some embodiments, R 4 is H, hydroxyl, halogen, C 1 -C 4 alkyl, C 1 -C 4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

C 1 -C 4 alkyl or C 1 -C 4 fluoroalkyl. In some embodiments, R 4 is H, hydroxyl, F, Cl, , , ,

In some embodiments, X 5 is N. In some embodiments, X 5 is CR 5 . In some embodiments, R 5 is H, hydroxyl, halogen, optionally substituted C 1 -C 4 alkyl, or optionally substituted C 1 -C 4 heteroalkyl. In some embodiments, R 5 is H, hydroxyl, halogen, C 1 -C 4 alkyl, C 1 -C 4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

C 1 -C 4 alkyl or C 1 -C 4 fluoroalkyl. In some embodiments, R 5 is H, hydroxyl, F, Cl, or . in some embodiments, R 5 is H.

In some embodiments, X 2 is CR 2 ; X 3 is CR 3 ; X 4 is CR 4 ; X 5 is CR 5 ; R 2 is H; R 3 is H; R 4 is F, Cl,

In some embodiments, R 10 is optionally substituted Ci-Ce alkyl or optionally substituted C3-C7 cycloalkyl. In some embodiments, R 10 is H or optionally substituted Ci-Ce alkyl. In some embodiments, R 10 is optionally substituted C 1 -C3 alkyl or optionally substituted C3-C6 cycloalkyl. In some embodiments, R 10 is H or optionally substituted C 1 -C3 alkyl. In some embodiments, R 10 is H. In some embodiments, R 10 is ^^3 in some embodiments , R 10 is . In some embodiments, R 10 is H or ^ h 3 . In some embodiments, R 10 is H 3 or t— ^ . In some embodiments, R 10 is H. In some embodiments, R 10 is ( -' H 3 . in some embodiments, R 10 is .

In some embodiments, R 11 is H or optionally substituted C1-C3 alkyl. In some embodiments, R 11

Y CH 3 is H or C1-C3 alkyl. In some embodiments, R 11 is H or \ . In some embodiments, R 11 is H.

In some embodiments, R 12 is H or optionally substituted C1-C3 alkyl. In some embodiments, R 12 is

V CH 3

H or C1-C3 alkyl. In some embodiments, R 12 is H or ' . In some embodiments, R 12 is H.

In some embodiments, R 11 is H, and R 12 is H.

In some embodiments, X 6 is N. In some embodiments, X 6 is CR 6 . In some embodiments, R 6 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 6 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is yCH 3 \y \CH

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 6 is H, hydroxyl, F, Cl, ' , \ 3 ,

In some embodiments, X 7 is N. In some embodiments, X 7 is CR 7 . In some embodiments, R 7 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 7 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 7 is H, hydroxyl, F, Cl, V CHs ^CH 3 ,

In some embodiments, R 13 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 13 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or -OR° 1 , wherein R° 1 is C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 13 is H, hydroxyl, In some embodiments, R 14 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 14 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or -OR° 1 , wherein R° 1 is C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 14 is H, hydroxyl In some embodiments, R 14 is H, F, Cl,

Y CH 3 Y cp3 or Y°"CH 3

In some embodiments, X 8 is N. In some embodiments, X 8 is CR 8 . In some embodiments, R 8 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 8 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 8 is H, hydroxyl, F, Cl, Y^CH 3

In some embodiments, X 9 is N. In some embodiments, X 9 is CR 9 . In some embodiments, R 9 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 9 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or-OR° 1 , wherein R° 1 is

C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 9 is H, hydroxyl, F, Cl, , ,

In some embodiments, R 15 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 15 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or -OR° 1 , wherein R° 1 is C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 15 is H, hydroxyl In some embodiments, R 16 is H, hydroxyl, halogen, optionally substituted C1-C4 alkyl, or optionally substituted C1-C4 heteroalkyl. In some embodiments, R 16 is H, hydroxyl, halogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or -OR° 1 , wherein R° 1 is C1-C4 alkyl or C1-C4 fluoroalkyl. In some embodiments, R 16 is H, hydroxyl

In some embodiments, the compound of Formula I has the structure of Formula la:

Formula la, or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of Formula I has the structure of Formula lb:

Formula lb, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula I has the structure of Formula lc:

Formula lc, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula I has the structure of Formula Id: or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of Formula I has the structure of Formula le:

Formula le, or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of Formula I has the structure of Formula If:

Formula If, or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of Formula I has the structure of Formula Ig:

Formula Ig, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound has the structure of any one of compounds 1-26 in Table , or a pharmaceutically acceptable salt thereof.

In an aspect, the disclosure features a compound having the structure of any one of compounds-26 in Table 1 , or a pharmaceutically acceptable salt thereof.

Table 1. Compounds 1-26 of the Disclosure

In another aspect, the disclosure features a pharmaceutical composition including any of the foregoing compounds, or pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable excipient. In an aspect, the disclosure features a method of reducing the level of HA01 in a cell, where the method includes contacting the cell with an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure features a method of inhibiting the activity of HA01 in a cell, where the method includes contacting the cell with an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In some embodiments, the cell is a liver cell. In some embodiments, the liver cell is a human liver cell.

In yet another aspect, the disclosure features a method of treating a disorder that leads to accumulation of oxalate in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure features a method of treating an HA01 -associated disorder in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In some embodiments, the disorder is primary hyperoxaluria 1 . In some embodiments, the disorder is primary hyperoxaluria 2.

In another aspect, the disclosure features a method of treating primary hyperoxaluria in a subject in need thereof (e.g., human in need thereof), where the method includes administering to the subject an effective amount of a compound of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In some embodiments, the primary hyperoxaluria is primary hyperoxaluria 1 . In some embodiments, the primary hyperoxaluria is primary hyperoxaluria 2.

In an aspect, the disclosure features a method of reducing the level of oxalate in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure features a method of reducing the level of glyoxylate in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In yet another aspect, the disclosure features a method of reducing the level of glycine in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In an aspect, the disclosure features a method of inhibiting the accumulation of oxalate in a subject in need thereof (e.g., human in need thereof), where the method comprises administering to the subject an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or any of the foregoing pharmaceutical compositions.

In some embodiments of any of the foregoing aspect, the subject is a human child, e.g., a human that is less than 18 years old (e.g., less than 15 years old, less than 10 years old, less than 9 years old, less than 8 years old, less than 7 years old, less than 6 years old, less than 5 years old, less than 4 years old, less than 3 years old, less than 2 years old, less than 1 years old). In some embodiments of any of the foregoing aspect, the subject is a human adult, e.g., a human that is at least 18 years old (e.g., at least 19 years old, at least 20 years old, at least 30 years old, at least 40 years old, at least 50 years old).

Chemical Terms

For any of the following chemical definitions, a number following an atomic symbol indicates that total number of atoms of that element that are present in a particular chemical moiety. As will be understood, other atoms, such as hydrogen atoms, or substituent groups, as described herein, may be present, as necessary, to satisfy the valences of the atoms. For example, an unsubstituted C2 alkyl group has the formula -CH2CH3. When used with the groups defined herein, a reference to the number of carbon atoms includes the divalent carbon in acetal and ketal groups but does not include the carbonyl carbon in acyl, ester, carbonate, or carbamate groups. A reference to the number of oxygen, nitrogen, or sulfur atoms in a heteroaryl group only includes those atoms that form a part of a heterocyclic ring.

The term “acyl,” as used herein, represents a hydrogen or an alkyl group that is attached to a parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group), acetyl, trifluoroacetyl, propionyl, and butanoyl. Exemplary unsubstituted acyl groups include from 1 to 6, from 1 to 11 , or from 1 to 21 carbons.

The term “alkyl,” as used herein, refers to a branched or straight-chain monovalent saturated aliphatic hydrocarbon radical of 1 to 20 carbon atoms (e.g., 1 to 16 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms). An alkylene is a divalent alkyl group.

The term “alkenyl,” as used herein, alone or in combination with other groups, refers to a straight chain or branched hydrocarbon residue having a carbon-carbon double bond and having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms). An alkenylene is a divalent alkenyl group.

The term “alkynyl,” as used herein, alone or in combination with other groups, refers to a straight chain or branched hydrocarbon residue having a carbon-carbon triple bond and having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms). An alkynylene is a divalent alkynyl group.

The term “amino,” as used herein, represents -N(R N1 )2, wherein each R N1 is, independently, H, OH, NO2, N(R N2 )2, S0 2 0R N2 , S0 2 R N2 , SOR N2 , an /V-protecting group, alkyl, alkoxy, aryl, arylalkyl, cycloalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), wherein each of these recited R N1 groups can be optionally substituted; or two R N1 combine to form an alkylene or heteroalkylene, and wherein each R N2 is, independently, H, alkyl, or aryl. The amino groups of the compounds described herein can be an unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(R N1 )2).

The term “aryl,” as used herein, refers to a monocyclic or polycyclic (e.g., bicyclic or tricyclic) radical of 6 to 12 carbon atoms having at least one aromatic ring. Examples of such groups include, but are not limited to, phenyl, naphthyl, 1 ,2,3,4-tetrahydronaphthyl, 1 ,2-dihydronaphthyl, indanyl, and 1 H- indenyl.

The term “arylalkyl,” as used herein, represents an alkyl group substituted with an aryl group. Exemplary unsubstituted arylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-C6 alkyl C6-C10 aryl, C1-C10 alkyl C6-C10 aryl, or C1-C20 alkyl C6-C10 aryl), such as, benzyl and phenethyl.

The term “azido,” as used herein, represents a -N3 group.

The term “cyano,” as used herein, represents a -CN group.

The term “carbocyclyl,” as used herein, refers to a monocarbocyclic or polycarbocyclic (e.g., bicyclic or tricyclic) radical of 3 to 12 carbon atoms in which no ring is aromatic. Carbocyclyl structures include cycloalkyl groups and unsaturated carbocyclyl radicals (e.g., cycloalkenyl and cycloalkynyl). Examples of a carbocyclyl group include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, norbornyl, adamantyl and cyclooctynyl.

The term “cycloalkyl,” as used herein, refers to a saturated, non-aromatic, monovalent monocarbocyclic or polycarbocyclic radical of 3 to 12 carbon atoms. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and adamantyl.

The terms “halo” or “halogen,” as used herein, means a fluorine (fluoro), chlorine (chloro), bromine (bromo), or iodine (iodo) radical.

The term “haloalkyl,” as used herein, refers to an alkyl group that is substituted with one or more halogen atoms. Examples of haloalkyl include fluoroalkyl (e.g., difluoromethyl, trifluoroethyl, and perfluoroalkyl (e.g., trifluoromethyl and pentafluoroethyl)), chloroalkyl, bromoalkyl, and iodoalkyl.

The term “heteroalkyl,” as used herein, refers to an alkyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. Examples of heteroalkyl groups are an “alkoxy” which, as used herein, refers alkyl-O- (e.g., methoxy and ethoxy). A heteroalkylene is a divalent heteroalkyl group.

The term “heteroalkenyl,” as used herein, refers to an alkenyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur.

Examples of heteroalkenyl groups are an “alkenoxy” which, as used herein, refers alkenyl-O- A heteroalkenylene is a divalent heteroalkenyl group.

The term “heteroalkynyl,” as used herein, refers to an alkynyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur.

Examples of heteroalkynyl groups are an “alkynoxy” which, as used herein, refers alkynyl-O- A heteroalkynylene is a divalent heteroalkynyl group.

The term “heteroaryl,” as used herein, refers to a monocyclic or polycyclic radical of 5 to 12 atoms having at least one aromatic ring containing at least 1 (e.g., 1 , 2, 3, 4, or 5) ring atoms selected from nitrogen, oxygen, and sulfur, with the remaining ring atoms being carbon. One or two ring carbon atoms of the heteroaryl group may be replaced with a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazoyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, oxaxolyl, and thiazolyl.

The term “heteroarylalkyl,” as used herein, represents an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroarylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-C6 alkyl C2-C9 heteroaryl, C1-C10 alkyl C2-C9 heteroaryl, or C1-C20 alkyl C2-C9 heteroaryl).

The term “heterocyclyl,” as used herein, refers a monocyclic or polycyclic radical having 3 to 12 atoms having at least one ring containing at least 1 (e.g., 1 , 2, 3, 4, or 5) ring atoms selected from nitrogen, oxygen, or sulfur, where no ring is aromatic. Heterocyclyl structures include heterocycloalkyl groups and unsaturated hetercyclyl radicals (e.g., heterocycloalkenyl and heterocycloalkynyl). Examples of heterocyclyl groups are morpholinyl, thiomorpholinyl, furyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl, and 1 ,3-dioxanyl.

The term “heterocyclylalkyl,” as used herein, refers to a saturated, non-aromatic, monovalent monocyclic or polycyclic radical of 3 to 12 atoms having at least one ring containing at least 1 (e.g., 1 , 2,

3, 4, or 5) ring atoms selected from nitrogen, oxygen, or sulfur, where no ring is aromatic.

The term “hydroxyalkyl,” as used herein, represents alkyl group substituted with an -OH group.

The term “hydroxyl,” as used herein, represents an -OH group.

The term “/V-protecting group,” as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used /V-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3rd Edition (John Wiley & Sons, New York, 1999). /V-protecting groups include, but are not limited to, acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4- bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L, or D, L-amino acids such as alanine, leucine, and phenylalanine; sulfonyl-containing groups such as benzenesulfonyl, and p-toluenesulfonyl; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p- bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2,4- 20 dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, a,a-dimethyl-3,5- dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl, arylalkyl groups such as benzyl, triphenylmethyl, and benzyloxymethyl, and silyl groups, such as trimethylsilyl. Preferred /V-protecting groups are alloc, formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t- butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).

The term “nitro,” as used herein, represents an -NO2 group.

The term “thiol,” as used herein, represents an -SH group.

Herein a phrase of the form “optionally substituted X” (e.g., optionally substituted alkyl) is intended to be equivalent to “X, wherein X is optionally substituted” (e.g., “alkyl, wherein said alkyl is optionally substituted”). It is not intended to mean that the feature “X” (e.g. alkyl) perse is optional.

The functional groups described herein (e.g., alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl (e.g., heterocycloalkyl)) may be substituted or unsubstituted. When substituted, there will generally be 1 to 6 (e.g., 1 to 5, 1 to 4, 1 to 3, or 1 to 2) substituents present, unless otherwise specified. Examples of substituents include: alkyl (e.g., unsubstituted and substituted, where the substituents include any group described herein, e.g., aryl, halo, hydroxyl), aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (e.g., substituted and unsubstituted cycloalkyl), halogen (e.g., fluoro), hydroxyl, heteroalkyl (e.g., substituted and unsubstituted methoxy, ethoxy, or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g., NH2 or mono- or dialkyl amino), azido, cyano, nitro, or thiol. Aryl, carbocyclyl (e.g., cycloalkyl), heteroaryl, and heterocyclyl groups may also be substituted with alkyl (unsubstituted and substituted such as arylalkyl (e.g., substituted and unsubstituted benzyl)). Further examples of substituents include: (1) Ci-e alkoxy; (2) Ci-e alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e. , -NH2) or a substituted amino (i.e. , -N(R N1 )2, where R N1 is as defined for amino); (4) Ce-m aryl-Ci-6 alkoxy; (5) azido; (6) halo; (7) (C2-9 heterocyclyl)oxy; (8) hydroxyl, optionally substituted with an O-protecting group; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) C1-7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) -CC>2R A , optionally substituted with an O- protecting group and where R A’ is selected from the group consisting of (a) C1-20 alkyl (e.g., Ci-e alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce-io aryl, (d) hydrogen, (e) Ci-e alk-Ce-io aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of -(CH 2 )s 2 (0CH 2 CH 2 )si(CH 2 )s30R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci-20 alkyl, and (h) amino-polyethylene glycol 0f -NR N1 (CH 2 )s2(CH2CH 2 O)si(CH2)s3NR N1 , wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted Ci-e alkyl; (15) -C(0)NR B’ R c’ , where each of R B’ and R c’ is, independently, selected from the group consisting of (a) hydrogen, (b) Ci-e alkyl, (c) Ce-m aryl, and (d) Ci-e alk-Ce-m aryl; (16) -SCteR 0’ , where R D’ is selected from the group consisting of (a) Ci-e alkyl, (b) Ce-m aryl, (c) Ci-e alk-Ce-m aryl, and (d) hydroxyl; (17) - S02NR E’ R f , where each of R E’ and R F is, independently, selected from the group consisting of (a) hydrogen, (b) Ci-e alkyl, (c) Ce-m aryl and (d) Ci-e alk-Ce-m aryl; (18) -C(0)R G’ , where R G’ is selected from the group consisting of (a) C1-20 alkyl (e.g., Ci-e alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce- aryl, (d) hydrogen, (e) Ci-e alk-Ce-m aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of - (CH 2 )s 2 (0CH 2 CH 2 )si(CH 2 )s30R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci-20 alkyl, and (h) amino-polyethylene glycol of - NR N1 (CH2) S 2(CH 2 CH 2 0) S I (CH 2 ) S3 NR n1 , wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted Ci-e alkyl;

(19) -NR H’ C(0)R r , wherein R H’ is selected from the group consisting of (a1) hydrogen and (b1) Ci-e alkyl, and R r is selected from the group consisting of (a2) C1-20 alkyl (e.g., Ci-e alkyl), (b2) C2-20 alkenyl (e.g., C2- 6 alkenyl), (c2) Ce-m aryl, (d2) hydrogen, (e2) Ci-e alk-Ce-m aryl, (f2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH 2 )s 2 (0CH 2 CH 2 )si(CH 2 )s30R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci-20 alkyl, and (h2) amino-polyethylene glycol of -

NR N1 (CH2) S 2(CH 2 CH 2 0) S I (CH 2 ) S3 NR n1 , wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted Ci-e alkyl;

(20) -NR J’ C(0)0R K’ , wherein R J’ is selected from the group consisting of (a1) hydrogen and (b1) Ci-e alkyl, and R K’ is selected from the group consisting of (a2) C1-20 alkyl (e.g., Ci-e alkyl), (b2) C2-20 alkenyl (e.g., C2- 6 alkenyl), (c2) Ce-m aryl, (d2) hydrogen, (e2) Ci-e alk-Ce-m aryl, (f2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH 2 )s 2 (0CH 2 CH 2 )si(CH 2 )s30R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci-20 alkyl, and (h2) amino-polyethylene glycol of - NR N1 (CH 2 ) S2 (CH 2 CH 2 0) S I (CH 2 ) S3 NR n1 , wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted Ci-e alkyl; (21) amidine; and (22) silyl groups such as trimethylsilyl, t-butyldimethylsilyl, and tri-isopropylsilyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl can be further substituted with an oxo group to afford the respective aryloyl substituent.

Those skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomers, geometric isomers, tautomers) and/or isotopic (e.g., in which one or more atoms has been substituted with a different isotope of the atom, such as hydrogen substituted for deuterium) forms. Unless otherwise indicated or clear from context, a depicted structure can be understood to represent any such isomeric or isotopic form, individually or in combination. In some embodiments, one or more compounds depicted herein may exist in different tautomeric forms. As will be clear from context, unless explicitly excluded, references to such compounds encompass all such tautomeric forms. In some embodiments, tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. In certain embodiments, a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form. Examples of moieties with prototropic tautomeric forms are ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1 H- and 3H-imidazole, 1 H- and 3H- 1 ,2,3-triazole, 1 H-, 2H- and 4H- 1 ,2,4-triazole, 1 H- and 2H- isoindole, and 1 H- and 2H-pyrazole. In some embodiments, tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. The invention embraces all of these forms.

Compounds described herein can have one or more asymmetric carbon atoms and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates, or mixtures of diastereoisomeric racemates. The optically active forms can be obtained for example by resolution of the racemates, by asymmetric synthesis or asymmetric chromatography (chromatography with a chiral adsorbent or eluant). That is, certain of the disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. "Enantiomer" means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that are not related as mirror images, most commonly because they contain two or more asymmetrically substituted carbon atoms and represent the configuration of substituents around one or more chiral carbon atoms. Enantiomers of a compound can be prepared, for example, by separating an enantiomer from a racemate using one or more well-known techniques and methods, such as, for example, chiral chromatography and separation methods based thereon. The appropriate technique and/or method for separating an enantiomer of a compound described herein from a racemic mixture can be readily determined by those of skill in the art. "Racemate" or "racemic mixture" means a compound containing two enantiomers, wherein such mixtures exhibit no optical activity; i.e., they do not rotate the plane of polarized light. “Geometric isomer" means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon- carbon double bond may be in an E (substituents are on 25 opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R," "S," "S*," "R*," "E," "Z," "cis," and "trans," indicate configurations relative to the core molecule. Certain of the disclosed compounds may exist in atropisomeric forms. Atropisomers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. The compounds described herein may be prepared as individual isomers by either isomer-specific synthesis or resolved from an isomeric mixture. Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide 35 of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by weight relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by weight optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%,

70%, 80%, 90%, 99%, or 99.9% by weight pure. Percent optical purity is the ratio of the weight of the enantiomer or over the weight of the enantiomer plus the weight of its optical isomer. Diastereomeric purity by weight is the ratio of the weight of one diastereomer or over the weight of all the diastereomers. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by mole fraction pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by mole fraction pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%,

70%, 80%, 90%, 99%, or 99.9% by mole fraction pure. Percent purity by mole fraction is the ratio of the moles of the enantiomer or over the moles of the enantiomer plus the moles of its optical isomer.

Similarly, percent purity by moles fraction is the ratio of the moles of the diastereomer or over the moles of the diastereomer plus the moles of its isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has at least one chiral center, it is to be understood that the name or structure encompasses either enantiomer of the compound free from the corresponding optical isomer, a racemic mixture of the compound, or mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomer(s), or mixtures of diastereomers in which one or more diastereomer is enriched relative to the other diastereomers. The invention embraces all of these forms.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

Definitions

In this application, unless otherwise clear from context, (i) the term “a” may be understood to mean “at least one”; (ii) the term “or” may be understood to mean “and/or”; and (iii) the terms “including” and “including” may be understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps.

As used herein, the terms “about” and “approximately” refer to a value that is within 10% above or below the value being described. For example, the term “about 5 nM” indicates a range of from 4.5 to 5.5 nM.

As used herein, the term “accumulation of oxalate” refers to an increased level of oxalate (e.g., urinary oxalate level) in a subject (e.g., human), as compared to a reference (e.g., the level of oxalate in a healthy subject or the level of oxalate prior to administration of a compound of the invention). In some embodiments, the level of oxalate is measured using an assay known in the art, e.g., an oxalate assay kit in which oxalate reacts with oxalate converter and oxalate enzyme mix to form an Intermediate that reacts in turn with a highiy specific probe to generate color that can be detected at 00=450 nrrt.

As used herein, the term “inhibiting accumulation of oxalate” refers to reducing the rate of accumulation of oxalate in a subject compared to the rate of accumulation of oxalate in the subject prior to administration of a compound of the invention (e.g., a decrease in the rate of accumulation by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease in the rate of accumulation of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease in the rate of accumulation by more than about 1.2-fold, about 1 .4-fold, about 1 .5-fold, about 1 .8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, 25 about 10-fold, about 15-fold, about 20- fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more).

By “level” is meant a level of, e.g., oxalate, glyoxylate, or glycine, as compared to a reference.

The reference can be any useful reference, as defined herein. By a “decreased level” or an “increased level” of oxalate is meant a decrease or increase in, e.g., oxalate level (e.g., urinary oxalate level or blood oxalate level), glyoxylate level, or glycine level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about 1 .2-fold , about 1 .4-fold, about 1 .5-fold, about 1 .8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, 25 about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more). A level of, e.g., oxalate (e.g., urinary oxalate level or blood oxalate level), glyoxylate, or glycine, may be expressed in mass/vol (e.g., g/dL, mg/ml_, pg/mL, or ng/ml_), in mass/vol/time (e.g., g/dL/hours, mg/mL/hours, pg/mL/hours, or ng/mL/hours), or percentage relative to total oxalate in a sample (e.g., urine sample or blood sample).

As used herein, the term “administration” refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreal.

As used herein, a “combination therapy” or “administered in combination” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In some embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or more than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.

As used herein, the terms “effective amount,” “therapeutically effective amount,” and “a “sufficient amount” of an agent refer to a quantity of an agent described herein sufficient to, when administered to the subject, including a human, effect beneficial or desired results, including clinical results, and, as such, an “effective amount” or synonym thereto depends on the context in which it is being applied. For example, in the context of treating HA01 -associated disorders and/or disorders that lead to oxalate accumulation, it is an amount of the agent that reduces the level and/or activity of HA01 or reduces the level of oxalate sufficient to achieve a treatment response as compared to the response obtained without administration of the agent that reduces the level and/or activity of HA01 or reduces the level of oxalate. The amount of a given agent that reduces the level and/or activity of HA01 or reduces the level of oxalate described herein that will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, and/or weight) or host being treated, and the like, but can nevertheless be routinely determined by one of skill in the art. Also, as used herein, a “therapeutically effective amount” of an agent is an amount which results in a beneficial or desired result in a subject as compared to a control. As defined herein, a therapeutically effective amount of an agent that reduces the level and/or activity of HA01 , reduces the level of oxalate, or treats an HA01 -associated disorder may be readily determined by one of ordinary skill by routine methods known in the art. Dosage regimen may be adjusted to provide the optimum therapeutic response.

As used herein, the term “HA01 -associated disorder” refers to a disorder in which an activity related to HA01 , or a downstream effect (e.g., increased level of oxalate or increased level of glyoxylate) is amplified compared to a reference. The agents described herein (e.g., a compound of Formula I) are useful for the treatment of an HA01 -associated disorder and/or alleviation of a symptom associated therewith.

The term “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

As used herein, the term “pharmaceutically acceptable salt” means any pharmaceutically acceptable salt of a compound described herein. For example, pharmaceutically acceptable salts of any of the compounds described herein include those that are within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al.,

J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting a free base group with a suitable organic acid.

The compounds described herein may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds described herein, be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparation of the appropriate salts are well-known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, 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, toluenesulfonate, undecanoate, and valerate salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.

The term “pharmaceutical composition,” as used herein, represents a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.

As used herein, “progression-free survival” (PFS) refers to the length of time during and after treatment during which a disease being treated (e.g., a HA01 -associated disorder) does not get worse. Progression-free survival may include the amount of time patients have experienced a complete response or a partial response, as well as the amount of time patients have experienced stable disease. In some embodiments, PFS may be defined as the time from randomization or the beginning of treatment to the first documented disease progression.

By “reducing the level of glycine,” is meant decreasing the level of glycine in a subject (e.g., human). The level of glycine may be measured using any method known in the art.

By “reducing the level of glyoxylate,” is meant decreasing the level of glyoxylate in a subject (e.g., human). The level of glyoxylate may be measured using any method known in the art. By “reducing the level of oxalate,” is meant decreasing the level of oxalate in a subject (e.g., human). The level of oxalate may be measured using any method known in the art.

By “reducing the level of HAOG is meant decreasing the level of HA01 in a cell or a subject (e.g., a human cell or a human subject). The level of HA01 is determined by methods known in the art either directly or indirectly. “Directly determining” means performing a process (e.g., performing an assay or test on a sample) to obtain the physical entity or value. “Indirectly determining” refers to receiving the physical entity or value from another party or source (e.g., a third-party laboratory that directly acquired the physical entity or value). Methods to measure HA01 level generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, liquid chromatography (LC)-mass spectrometry, microcytometry, microscopy, fluorescence activated cell sorting (FACS), and flow cytometry, as well as assays based on a property of a protein including, but not limited to, enzymatic activity or interaction with other protein partners. In some embodiments, the level of HA01 is decreased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; decreased by more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; or decreased by more than about 1 .2-fold, about 1 .4- fold, about 1 .5-fold, about 1 .8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, 25 about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more).

By “inhibiting the activity of HA01 ” is meant decreasing the level of an activity related to HA01 , or a related downstream effect (e.g., increased level of oxalate or increased level of glyoxylate). The activity level of HA01 may be measured using any method known in the art, e.g., a horseradish peroxidase- based assay as described herein. In some embodiments, an agent that inhibits the activity of HA01 is a compound as described herein. In some embodiments, the activity of HA01 is decreased by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; decreased by more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; or decreased by more than about 1 .2-fold, about 1 .4-fold, about 1 .5-fold, about 1 .8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0- fold, 25 about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100- fold, about 1000-fold, or more).

As used herein, the term “subject” refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.

As used herein, the terms "treat," "treated," or "treating" mean both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.

The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.

Detailed Description

The present disclosure features compounds and methods useful for treating HA01 -associated disorders and/or disorders (e.g., primary hyperoxalurias (e.g., primary hyperoxaluria 1)) that lead to oxalate accumulation and its symptoms (e.g., end stage renal disease). The disclosure further features compositions and methods useful for reduction of the level and/or activity of HA01 , e.g., for the treatment of disorders that lead to oxalate accumulation and its symptoms in a subject in need thereof (e.g., human).

Glyoxylate Enzymes

Alanine-glyoxylate aminotransferase (AGT), glyoxylate reductase-hydroxypyruvate reductase (GRHPR), and 4-hydroxy-2-oxoglutarate aldolase (HOGA1) are involved in the breakdown and processing of protein building blocks (e.g., amino acids) and other compounds. AGT is involved in the breakdown of L-alanine and glyoxylate. GRHPR is involved in the breakdown of glyoxylate and hydroxypyruvate. HOGA1 is involved in the breakdown of amino acids, which results in the formation of glyoxylate. Mutations in the genes encoding AGT, GRHPR, or HOGA1 lead to a decrease in production or activity of the respective proteins, which prevents the normal breakdown of glyoxylate. AGT and GRHPR gene mutations result in an accumulation of glyoxylate, which is then converted to oxalate for removal from the body as a waste product. Oxalate has a tendency to form insoluble calcium oxalate crystals, which are a major component of kidney stones.

Hydroxyacid oxidase 1 (HA01)

Hydroxyacid oxidase 1 (HA01) orglycolate oxidase (GO) is a protein encoded by the HA01 gene that is primarily expressed in the liver and pancreas. HA01 oxidizes glycolic acid (glycolate) to glyoxylate. Typically, the liver-specific peroxisomal enzymes alanine-glyoxylate aminotransferase (AGT) and glyoxylate reductase-hydroxypyruvate reductase (GRHPR) catalyze the detoxification (transamination) of glyoxylate to glycine. Loss of AGT and/or GRHPR function results in accumulation of glyoxylate, which is then oxidized to oxalate by lactate dehydrogenase (LDH). Oxalate is then filtered by the kidney and excreted in urine. Oxalate tends to precipitate as insoluble calcium oxalate crystals in tissues, leading to harmful kidney stones and bladder stones. Renal damage is caused by a combination of toxicity from oxalate (e.g., death of tubular epithelial cells of the renal tubules of the kidneys), nephrocalcinosis, and renal obstruction by the stones.

Thus, reduction of HA01 activity should reduce endogenous oxalate production, e.g., oxidation of glyoxylate by LDH, by reducing the production of glyoxylate from glycolic acid. Ultimately, this should lead to a reduction of oxalate and therefore calcium oxalate.

Primary Hyperoxaluria

Metabolic disorders can be caused by mutations in genes that encode enzymes of a metabolic pathway. Mutation can result in a harmful accumulation of a compound that is normally metabolized by the enzyme. Primary hyperoxalurias (PHs) are rare autosomal recessive disorders affecting the glyoxylate or hydroxyproline pathways that result in an overproduction or accumulation of oxalate.

Primary hyperoxaluria is estimated to affect 1 in 58,000 individuals worldwide.

There are three types of primary hyperoxaluria (PH): (i) primary hyperoxaluria type 1 (PH1), in which kidney stones typically begin to appear anytime from childhood to early adulthood and end stage renal disease (ESRD) can develop at any age; (ii) primary hyperoxaluria type 2 (PH2), a disorder similar to PH1 , but in which ESRD develops later in life; and (iii) primary hyperoxaluria type 3 (PH3), in which kidney stones typically develop in early childhood. PH1 accounts for approximately 80% of primary hyperoxaluria cases and eventually leads to renal failure after several years. PH2 and PH3 each account for about 10% of primary hyperoxaluria cases and have a less severe course. Mutation of alanine- glyoxylate aminotransferase (AGT), glyoxylate reductase-hydroxypyruvate reductase (GRHPR), and 4- hydroxy-2-oxoglutarate aldolase (HOGA1) cause primary hyperoxaluria types 1 , 2, and 3, respectively.

At present, permanent treatment of declining kidney function in PH1 patients involves combined liver and kidney transplant, a costly and often unfeasible treatment. Kidney stones can be treated, e.g., by surgical removal of the stones, dietary changes to increase fluid intake, dietary changes to restrict oxalate intake, urine alkalization, diuretics, and crystallization inhibitors (e.g., citrate, bicarbonate, and magnesium). Nonetheless, these therapeutic approaches do not address the origin of disorders that lead to oxalate accumulation.

Compounds

Compounds described herein reduce the level of an activity related to HA01 and/or reduce the level of oxalate for the treatment of disorders (e.g., primary hyperoxalurias (e.g., primary hyperoxaluria 1)) that lead to oxalate accumulation and its symptoms. Exemplary compounds described herein have the structure according to Formula I. Formula I is shown below: N or CR 7 ; X 8 is N or CR 8 ; X 9 is N or CR 9 ; Y is O or NR Y ; each of R 1 , R 2 , R 3 , R 4 , and R 5 is, independently, H, halogen, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R Y is H or optionally substituted C1-C6 alkyl; R 10 is H, optionally substituted C1-C6 alkyl, or optionally substituted C3- C7 cycloalkyl; each of R 11 and R 12 is, independently, H or optionally substituted C1-C6 alkyl; each of R 6 ,

R 7 , R 13 , and R 14 is, independently, H, halogen, hydroxyl, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R 6 and R 13 , R 13 and R 14 , or R 7 and R 14 , together with the carbon atoms to which each is attached, combine to form an optionally substituted 5- or 6-membered aryl or heteroaryl; and each of R 8 , R 9 , R 15 , and R 16 is, independently, H, halogen, hydroxyl, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; or R 8 and R 15 or R 9 and R 16 , together with the carbon atoms to which each is attached, combine to form an optionally substituted 5- or 6-membered aryl or heteroaryl, or a pharmaceutically acceptable salt thereof.

Pharmaceutical Uses

The compounds described herein are useful in the methods of the invention and, while not bound by theory, are believed to exert their desirable effects through their ability to modulate the level of oxalate, glyoxylate, and/or glycine, e.g., by modulating the level, status, and/or activity of HA01 in a cell in a subject (e.g., mammal (e.g., human)).

An aspect of the present invention relates to methods of treating disorders such as primary hyperoxalurias (e.g., primary hyperoxaluria 1 or primary hyperoxaluria 2) related to oxalate level and/or HA01 levels and/or activity in a subject in need thereof. In some embodiments, the compound is administered in an amount and for a time effective to result in one of (or more, e.g., two or more, three or more, four or more of): (a) reduced level of oxalate production (e.g., reduced urinary oxalate level); (b) reduced oxalate accumulation; (c) decreased frequency and/or size of kidney stones; (d) delayed onset of end stage renal disease; (e) increased survival of a subject (e.g., mammal (e.g., human)); and (f) increased progression free survival of a subject (e.g., human). For example, in some embodiments, the methods of the invention result in an increase in the length of time during and after treatment that the subject lives with an HA01 -associated disorder, but the disorder does not get worse (e.g., the amount of renal and/or bladder stones are stable or decreased and/or the amount of kidney and/or bladder infections are stable or decreased). Administration of compounds of the invention may result in reduced levels of oxalate. For example, after treatment, the level of oxalate is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment. Alternatively, administration of compounds of the invention may reduce further accumulation of oxalate in a subject or cell. For example, after treatment, further accumulation of oxalate is reduced by 5% or more (e.g., 10%, 20%,

30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to the level prior to treatment.

Administration of compounds of the invention may result in reduced levels of glyoxylate. For example, after treatment, the level of glyoxylate is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment.

Administration of compounds of the invention may result in reduced levels of glycine. For example, after treatment, the level of glycine is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment.

Administration of compounds of the invention may result in a decrease in number of kidney stones. For example, after treatment, kidney stone number is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment.

Administration of compounds of the invention may result in a reduced frequency of new kidney stones. For example, after treatment, the frequency of new kidney stones is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment.

Administration of compounds of the invention may result in decreased overall mass of kidney stones. For example, after treatment, the overall mass of kidney stones is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more) relative to number prior to treatment.

Administration of compounds of the invention may result in delayed onset of end stage renal disease in a subject. For example, after treatment, the onset of end stage renal disease is delayed by more than 30 days (more than 60 days, 90 days, or 120 days) or by more than 1 year (e.g., more than 2 years, more than 5 years, more than 10 years, more than 15 years, more than 20 years, more than 30 years, more than 40 years, or more than 50 years) in a subject. An increase in average onset of end stage renal disease of a population may be measured by any reproducible means. For example, an increase in average onset of end stage renal disease of a population may be measured by calculating for a population the average length of time following initiation of treatment with a compound described herein before onset of end stage renal disease. An increase in average length of time before onset of end stage renal stage disease may also be measured, for example, by calculating for a population the average length of time before onset of end stage renal stage disease following completion of a first round of treatment with a pharmaceutically acceptable salt of a compound described herein. Administration of compounds of the invention can result in an increase in average survival time of subjects treated according to the present invention in comparison to a population of untreated subjects. For example, the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days) or by more than 1 year (more than 2 years, more than 5 years, more than 10 years, or more than 20 years). An increase in average survival time of a population may be measured by any reproducible means. For example, an increase in average survival time of a population may be measured by calculating for a population the average length of survival following initiation of treatment with the compound described herein. An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of a compound described herein.

Administration of compounds of the invention can also result in a decrease in the mortality rate of subjects treated in comparison to an untreated population. For example, the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%). For example, a decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of a compound described herein. A decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a pharmaceutically acceptable salt of a compound described herein.

Additional Therapies

A method of the invention can be used alone or in combination with additional therapy (e.g., a compound (e.g., vitamin B6)) that treats HA01 -associated disorders and/or disorders that lead to oxalate accumulation (e.g., primary hyperoxalurias (e.g., primary hyperoxaluria 1 or primary hyperoxaluria 2)) or symptoms associated therewith. The dosages of one or more of the additional therapies (e.g., a compound (e.g., vitamin B6)) may be reduced from standard dosages when administered alone. For example, doses may be determined empirically from drug combinations and permutations or may be deduced by isobolographic analysis (e.g., Black et al., Neurology 65:S3-S6 (2005)). In this case, dosages of the compounds when combined should provide a therapeutic effect.

In any of the combination embodiments described herein, the first and second therapy are administered simultaneously or sequentially, in either order. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours up to 24 hours or up to 1-7, 1-14, 1-21 or 1-30 days or up to 1-5 years, 5-10 years, or 10-20 years before or after the second therapeutic agent.

Additional therapy that treats HA01 -associated disorders and/or disorders that lead to oxalate accumulation (e.g., primary hyperoxalurias (e.g., primary hyperoxaluria 1 or primary hyperoxaluria 2)) or symptoms associated therewith, can also be a non-drug treatment. For example, the additional therapy may be shockwave lithotripsy, cystoscopy and ureteroscopy, or percutaneous nephrolithotomy to remove kidney stones, or an oxalate-limited diet.

In some embodiments, two or more additional therapies are administered. In some embodiments, an additional therapy (e.g., a compound (e.g., vitamin B6)) is administered with an oxalate- limited diet. In other embodiments, the additional therapy (e.g., a compound (e.g., vitamin B6)) is administered without an oxalate-limited diet. Pharmaceutical Compositions

The pharmaceutical compositions described herein are preferably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.

The compounds described herein may be used in the form of the free base, in the form of salts, solvates, and as prodrugs. All forms are within the methods described herein. In accordance with the methods of the invention, the described compounds or salts, solvates, or prodrugs thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compounds described herein may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, intratumoral, ortransdermal administration and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.

A compound described herein may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard- or soft-shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, a compound described herein may be incorporated with an excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, and wafers. A compound described herein may also be administered parenterally. Solutions of a compound described herein can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO, and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington’s Pharmaceutical Sciences (2012, 22nd ed.) and in The United States Pharmacopeia: The National Formulary (USP 41 NF36), published in 2018. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that may be easily administered via syringe. Compositions for nasal administration may conveniently be formulated as aerosols, drops, gels, and powders. Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device. Alternatively, the sealed container may be a unitary dispensing device, such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use. Where the dosage form includes an aerosol dispenser, it will contain a propellant, which can be a compressed gas, such as compressed air or an organic propellant, such as fluorochlorohydrocarbon.

The aerosol dosage forms can also take the form of a pump-atomizer. Compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, where the active ingredient is formulated with a carrier, such as sugar, acacia, tragacanth, gelatin, and glycerine. Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base, such as cocoa butter. A compound described herein may be administered intratumorally, for example, as an intratumoral injection. Intratumoral injection is injection directly into the tumor vasculature and is specifically contemplated for discrete, solid, accessible tumors. Local, regional, or systemic administration also may be appropriate. A compound described herein may advantageously be contacted by administering an injection or multiple injections to the tumor, spaced for example, at approximately, 1 cm intervals. In the case of surgical intervention, the present invention may be used preoperatively, such as to render an inoperable tumor subject to resection. Continuous administration also may be applied where appropriate, for example, by implanting a catheter into a tumor or into tumor vasculature.

The compounds described herein may be administered to an animal, e.g., a human, alone or in combination with pharmaceutically acceptable carriers, as noted herein, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration, and standard pharmaceutical practice.

Dosages

The dosage of the compounds described herein, and/or compositions including a compound described herein, can vary depending on many factors, such as the pharmacodynamic properties of the compound; the mode of administration; the age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of the treatment, and the type of concurrent treatment, if any; and the clearance rate of the compound in the animal to be treated. One of skill in the art can determine the appropriate dosage based on the above factors. The compounds described herein may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. In general, satisfactory results may be obtained when the compounds described herein are administered to a human at a daily dosage of, for example, between 0.05 mg and 3000 mg (measured as the solid form). Dose ranges include, for example, between 10-1000 mg (e.g., 50-800 mg). In some embodiments, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 mg of the compound is administered.

Alternatively, the dosage amount can be calculated using the body weight of the patient. For example, the dose of a compound, or pharmaceutical composition thereof, administered to a patient may range from 0.1-50 mg/kg (e.g., 0.25-25 mg/kg). In exemplary, non-limiting embodiments, the dose may range from 0.5-5.0 mg/kg (e.g., 0.5, 1 .0, 1 .5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 mg/kg) or from 5.0-20 mg/kg (e.g., 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 mg/kg).

Kits

The invention also features kits including (a) a pharmaceutical composition including an agent (e.g., a compound of Formula I) that reduces the level of oxalate, glyoxylate, and/or glycine, and/or reduces the level and/or activity of HA01 in a cell or subject described herein, and (b) a package insert with instructions to perform any of the methods described herein. In some embodiments, the kit includes (a) a pharmaceutical composition including an agent (e.g., a compound of Formula I) that reduces the level of oxalate, glyoxylate, and/or glycine, and/or reduces the level and/or activity of HA01 in a cell or subject described herein, (b) an additional therapy (e.g., vitamin B6 or an oxalate-limited diet), and (c) a package insert with instructions to perform any of the methods described herein.

Examples The following examples are intended to illustrate but not to limit the invention. Additional compounds not specifically exemplified may be synthesized using conventional methods in combination with the methods described herein.

General Procedure A

Example 1 : 3-chloro-2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-4'-fluoro-[1 ,1 biphenyl]-4-carboxylic acid (Compound 6) Synthesis oftert-butyl (2-bromo-4-fluorobenzyl)carbamate (Step 1)

A mixture of (2-bromo-4-fluorophenyl)methanamine hydrochloride (1.0 g, 4.9 mmol), di-te/ -butyl- dicarbonate (1.6 g, 7.35 mmol), 2N sodium hydroxide solution (7.2 ml_, 15 mmol) and dioxane (32 ml_) was stirred at room temperature overnight. The mixture was diluted with diethyl ether (100 ml_) and washed successively with water (100 ml_), dilute hydrochloric acid (100 ml_) and water (100 ml_). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography using silica gel with a gradient of hexanes-ethyl acetate to afford te/ -butyl (2-bromo-4-fluorobenzyl)carbamate (1 .1 g, 74%). LC-MS (M+H) + = 304.5.

A solution of te/ -butyl (2-bromo-4-fluorobenzyl)carbamate (1.1 g, 3.63 mmol) in DMF (35 mL) at 0 °C was treated with 60% sodium hydride (316 mg, 7.92 mmol) and stirred for 10 minutes. The mixture was treated with methyl iodide (1.12 g, 7.92 mmol) and stirred overnight. The mixture was diluted with diethyl ether (100 mL) and washed successively with water (3 x 50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was dissolved in acetonitrile (50 mL) and washed with hexanes (3 x 30 mL) and concentrated under reduced pressure to afford te/ -butyl (2-bromo-4-fluorobenzyl)(methyl)carbamate (800 mg, 69%); LC-MS (M+H) + = 317.6

Synthesis of methyl 2'-(((tert-butoxycarbonyl)(methyl)amino)methyl)-3-chloro-4'- fluoro-[ 1, 1 '-biphenyl]-4- carboxylate (Step 3)

A mixture of the (3-chloro-4-(methoxycarbonyl)phenyl)boronic acid (269 mg, 1 .26 mmol), [1 ,1'- Bis(diphenylphosphino)ferrocene]-dichloropalladium(ll) dichloromethane complex (60 mg, mmol), tert- butyl (2-bromo-5-fluorobenzyl)(methyl)carbamate (250 mg, 0.79 mmol), toluene (4 mL), ethanol (4 mL), and saturated aqueous sodium bicarbonate solution (4 mL) was heated to 85°C and shaken overnight. The mixture was diluted with water, extracted with dichloromethane and the combined organic extracts were concentrated under reduced pressure. Material was purified 0-100% ethyl acetate in hexanes on a silica gel normal phase column to afford methyl 2'-(((fe/?-butoxycarbonyl)(methyl)amino)methyl)-3-chloro- 4'-fluoro-[1 ,T-biphenyl]-4-carboxylate (235 mg, 73% yield).

Methyl 2'-(((fe/if-butoxycarbonyl)(methyl)amino)methyl)-3-chloro-4' -fluoro-[1 ,1'-biphenyl]-4- carboxylate was treated 4N HCI-dioxane (5 ml_) and stirred for 2 h. The mixture was concentrated under reduced pressure to afford crude methyl 3-chloro-4'-fluoro-2'-((methylamino)methyl)-[1 ,1'-biphenyl]-4- carboxylate (200 mg, 100%). LC-MS (M+H) + =308.

Synthesis of methyl 3-chloro-2'-((6-chloro-N-methyl-1H-indole-2-carboxamido)meth yl)-4'-fluoro-[1, 1'- biphenyl]-4-carboxylate (Step 5)

Methyl 3-chloro-4'-fluoro-2'-((methylamino)methyl)-[1 ,T-biphenyl]-4-carboxylate (200 mg, 0.58 mmol) in DMF (5 mL) was treated with 6-chloro-1 H-indole-2-carboxylic acid (112 mg, 0.58 mmol), DIEA (303 uL, 1 .74 mmol), and HATU (285 mg, 0.75 mmol). The mixture was shaken overnight at room temperature. The mixture was diluted with ethyl acetate, washed with water, and concentrated under reduced pressure. Material was purified 0-100% ethyl acetate in hexanes on a silica gel normal phase column to provide methyl 3-chloro-2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-4'-fluoro-[1 ,T- biphenyl]-4-carboxylate (169 mg, 60%). LC-MS (M+H) + =485.3.

Synthesis of (3-chloro-2'-((6-chloro-N-methyl-1H-indole-2-carboxamido)met hyl)-4'-fluoro-[1, 1'-biphenyl]-4- carboxylic acid (Step 6)

Methyl 3-chloro-2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-4'-fluoro-[1 , 1 ’-biphenyl]- 4-carboxylate (169 mg, 0.35 mmol) was added dioxane (5 mL) and 2N NaOH solution (3 mL) and the resulting mixture was heated to 50°C for 1 h. This reaction mixture was then treated with 2N HCI solution (3 mL). The mixture was concentrated under reduced pressure. The crude product was dissolved in DMSO and purified on reverse phase using a gradient of water-acetonitrile (0.1% formic acid) to provide (3-chloro-2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-4'-fluoro-[1 ,1'-biphenyl]-4-carboxylic acid (86 mg, 52%), (Compound 6) 1 HNMR (300 MHz, CDCb) d 13.31 (s, 1 H), 11.72 (s, 1 H), 7.85 (d, 1 H), 7.61 (m, 2H), 7.46 (m, 3H), 7.28 (m, 1 H), 7.15 (d, 1 H), 7.04 (d, 1 H), 6.90 (s, 1 H) 4.71 (s, 2H), 3.18 (s, 3H). LC-MS (M+H) + = 471 .3.

Example 2: 3-chloro-2’-((6-chloro-Af-methylbenzofuran-2-carboxamido)m ethyl)-[1 ,1 ’-biphenyl]-4- carboxylic acid (Compound 1) The title compound was synthesized using General Procedure A using (2- bromophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- chlorobenzofuran-2-carboxylic acid in Step 5. LC-MS (M+H) + = 454.

Example 3: 3-chloro-2'-((6-chloro-At-cyclopropyl-1 H-indole-2-carboxamido)methyl)-5'-fluoro-[1 ,1 biphenyl]-4-carboxylic acid (Compound 3)

Synthesis oftert-butyl (2-bromo-4-fluorobenzyl)(cyclopropyl)carbamate (Steps A and B) Step A Step B

A mixture of 2-bromo-1-(bromomethyl)-4-fluorobenzene (1.02 g, 3.82 mmol) and cyclopropylamine (0.58 ml, 8.44 mmol) in absolute ethanol (15 mL) was stirred overnight then concentrated under reduced pressure. The mixture was treated with di-fe/if-butyl-dicarbonate (1 .24 g,

5.74 mmol), 2N sodium hydroxide solution (6 mL, 12 mmol) and dioxane (12 mL) and stirred at room temperature for 2 h. The mixture was diluted with diethyl ether (100mL) and washed successively with water (100 mL), dilute hydrochloric acid (100 mL), and water (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash column chromatography using silica gel with a gradient of hexanes-ethyl acetate to afford te/ -butyl (2-bromo-4-fluorobenzyl)(cyclopropyl)carbamate (853 mg, 65% over 2 steps). LCMS (M+H) + = 344. The title compound was synthesized using General Procedure A using fe/ -butyl (2-bromo-4- fluorobenzyl)(cyclopropyl)carbamate and 4-borono-2-chlorobenzoic acid in Step 3 and 6-chloro-1 H- indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 497. Example 4: 3-chloro-2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-4'-methyl-[1 ,1 biphenyl]-4-carboxylic acid (Compound 4)

The title compound was synthesized using General Procedure A using (2-bromo-5- methylphenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 467.

Example 5: S^'-dichloro^'- e-chloro-W-methyM H-indole-2-carboxamido)methyl)-[1 ,1 '-biphenyl]-4- carboxylic acid (Compound 5) The title compound was synthesized using General Procedure A using (2-bromo-5- chlorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 487.

Example 6: 2-chloro-4-(2-((6-chloro-/V-methyl-1H-indole-2-carboxamido)m ethyl)-5-fluoropyridin-3- yl)benzoic acid (Compound 7)

The title compound was synthesized using General Procedure A using (3-bromo-5-fluoropyridin- 2-yl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6-chloro-1 H- indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 472. Example 7: Synthesis of 3-chloro-2'-((6-chloro-N-methyl-1H-indole-2-carboxamido)meth yl)-[1,1'- biphenyl]-4-carboxylic acid (Compound 8)

The title compound was synthesized using General Procedure A using (2- bromophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 453.

Example 8: 2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-5'-fluoro-[1 ,1 '-biphenyl]-4- carboxylic acid (Compound 9)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-boronobenzoic acid in Step 3, and 6-chloro-1 H- indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 437. Example 9: 2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-5'-fluoro-3-(trifluoromethyl) - [1,1'-biphenyl]-4-carboxylic acid (Compound 10)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-(trifluoromethyl)benzoic acid in Step 3, and 6-chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 505. Example 10: 2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-5'-fluoro-3-methyl-[1 ,1 biphenyl]-4-carboxylic acid (Compound 11)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-methylbenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 451 .

Example 11 : 2'-((6-chloro-/V-methyl-1 H-indole-2-carboxamido)methyl)-3,5'-difluoro-[1 ,1 '-biphenyl]- 4-carboxylic acid (Compound 12)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-fluorobenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 455. Example 12: 3-chloro-5'-fluoro-2'-((/V-methyl-6-(trifluoromethyl)-1 H-indole-2-carboxamido)methyl)- [1 ,1'-biphenyl]-4-carboxylic acid (Compound 13)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- (trifluoromethyl)-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 505. Example 13: 3-chloro-5'-fluoro-2'-((6-methoxy-/V-methyl-1 H-indole-2-carboxamido)methyl)-[1 ,1 biphenyl]-4-carboxylic acid (Compound 14)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- methoxy-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 467.

Example 14: S-chloro^'- /V.e-dimethyMH-indole^-carboxamidoJmethylHj'-fluoro-II.I'-bi phenyl]- 4-carboxylic acid (Compound 15)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- methyl-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 451 . Example 15: S-chloro-S'-fluoro^'^e-fluoro-N-methyMH-indole^-carboxamidoJ methylHI .I'- biphenyl]-4-carboxylic acid (Compound 16)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- fluoro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 455. Example 16: 3-chloro-2'-((6-chloro-N-methyl-1 H-indole-2-carboxamido)methyl)-5'-fluoro-[1 ,1 biphenyl]-4-carboxylic acid (Compound 17)

The title compound was synthesized using General Procedure A using (2-bromo-4- fluorophenyl)methanamine hydrochloride in Step 1 , 4-borono-2-chlorobenzoic acid in Step 3, and 6- chloro-1 H-indole-2-carboxylic acid in Step 5. LC-MS (M+H) + = 471 .

Example 17: S^'-difluoro^'- e-fluoro-Af-methyM H-indole-2-carboxamido)methyl)-[1 ,1 '-biphenyl]- 4-carboxylic acid (Compound 2)

To a solution of 2-bromo-5-fluorobenzaldehyde (10 g, 49.5 mmol), MeNhb (4 ml_, methanol solution) in MeOH (100 ml_) was stirred at 25 °C for 8 h. Then the NaBhU (1 .9 g, 49.5 mmol) was added to the solution. After stirred for 0.5 h, the HCI (1 N) was added to the solution slowly to adjust the pH to 7. The solvent was evaporated, and the residue was diluted with H2O (20 ml_) and extracted with EA (10 ml_ x 3). The combined organic layer was washed with brine (10 ml_ x 3), then dried over with anhydrous Na 2 SC> 4 and filtered. The filtrate was concentrated under vacuum and the residue was purified by combi- flash to afford 1-(2-bromo-5-fluorophenyl)-/V-methylmethanamine (10 g, 93%) as a white solid. LC-MS (M+H) + = 218.

To a solution of 1-(2-bromo-5-fluorophenyl)-A/-methylmethanamine (10 g, 45.8 mmol), B0C2O (10 g, 45.8 mmol) in THF (100 mL) and H2O (100 mL) was added NaHCC>3 (3.8 g, 45.8 mmol) and the mixture was stirred at 25 °C for 5 h. The solution was diluted with H2O (100 mL), extracted with EA (200 mL). The extract was dried, concentrated and the residue was purified by combi-flash to provide tert- butyl (2-bromo-5-fluorobenzyl)(methyl)carbamate (15 g, 100%) as a white solid. LC-MS (M+H) + = 318.

Synthesis of tert-butyl(5-fluoro-2-(4, 4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)(methyl)car bamate (Step 3)

To a solution of fe/ -butyl (2-bromo-5-fluorobenzyl)(methyl)carbamate (15 g, 47.3 mmol), 4,4,4',4',5,5,5'-heptamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (11 .3 g, 47.3 mmol) in dioxane (200 mL) was added K2CO3 (6.9 g, 47.3 mmol), Pd(dppf)Cl2 (6.9 g, 9.5 mmol) under N2 and the mixture was stirred at 80 °C for 2 h. The solution was concentrated under vacuum and the residue was purified by corn-flash to afford fe/?-butyl(5-fluoro-2-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)benzyl)(methyl)carbamate (5 g, impure) as a brown solid. LC-MS (M+H) + = 366.

Synthesis of methyl-2'-(((tert-butoxycarbonyl)(methyl)amino)methyl)-3,4'- difluoro-[ 1, 1 '-biphenyl]-4- carboxylate (Step 4)

To a solution of fe/?-butyl(5-fluoro-2-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)benzyl)(methyl)carbamate (300 mg, 0.82 mmol), methyl 4-bromo-2-fluorobenzoate (190 mg, 0.82 mmol) in dioxane (20 mL) was added K2CO3 (106 mg, 0.82 mmol), Pd(dppf)Cl2 (60 mmol) under N2 and the mixture was stirred at 80 °C for 2 h. The solution was concentrated under vacuum and the residue was purified by combi-flash to provide methyl-2'-(((fe/if-butoxycarbonyl)(methyl)amino)methyl)-3,4' -difluoro- [1 ,T-biphenyl]-4-carboxylate (250 mg, 70%) as a brown solid. LC-MS (M+H) + = 392.

To a solution of methyl2'-(((fe/?-butoxycarbonyl)(methyl)amino)methyl)-3,4'-d ifluoro-[1 ,T- biphenyl]-4-carboxylate (250 mg, 0.64 mmol) in 10 mL DCM was added TFA (2 mL), the solution stirred for 2 h at rt. The solution was concentrated under vacuum to give 3,4'-difluoro-2'-((methylamino)methyl)- [1 ,T-biphenyl]-4-carboxylate (186 mg, 100%) as yellow oil. LC-MS (M+1) + = 292.

Synthesis of methyl 3, 4'-difluoro-2'-((6-fluoro-N-methyl-1H-indole-2-carboxamido)m ethyl)-[1,1 '-biphenyl]- 4-carboxylate (Step 6)

To a solution of 6-fluoro-1 H-indole-2-carboxylic acid in dichloromethane (10 mL) was added SOCI2 (1 mL) and DMF (5 drops) and the solution was stirred at rt for 5 h. The solvent was evaporated and the residue was dissolved in dichloromethane (5 mL) and added to a solution of 3,4'-difluoro-2'- ((methylamino)methyl)-[1 ,T-biphenyl]-4-carboxylate (176 mg, 0.64 mmol) dichloromethane (10 mL) and pyridine (1 mL) the mixture was stirred at 25 °C for 2 h. The solution was concentrated under vacuum and the residue was purified by combi-flash to afford methyl 3,4'-difluoro-2'-((6-fluoro-N-methyl-1 H-indole- 2-carboxamido)methyl)-[1 ,T-biphenyl]-4-carboxylate (50 mg, 17%) as a white solid. LC-MS (M+H) + =439.

Synthesis of3,4'-difluoro-2'-((6-fluoro-N-methyl-1H-indole-2-carboxami do)methyl)-[1,1'-biphenyl]-4- carboxylic acid (Step 7)

To a solution of methyl 3,4'-difluoro-2'-((6-fluoro-N-methyl-1 H-indole-2-carboxamido)methyl)-[1 ,T- biphenyl]-4-carboxylate (50 mg, 0.11 mmol), THF (5 mL), and H2O (5 mL) was added LiOH (4.4 mg, 0.11 mmol). The mixture stirred at 25 °C for 2 h. The solution was concentrated under vacuum and the residue was purified by prep-HPLC (chromatographic: column:-Gemini-C18 150 x 21 .2 mm 5 urn, flow term : ACN— H2O (0.1%TFA) gradient : 30-40) to give 3,4'-difluoro-2'-((6-fluoro-A/-methyl-1 H-indole-2- carboxamido)methyl)-[1 ,T-biphenyl]-4-carboxylic acid (12 mg, 21%) as a white solid. 1 H NMR (400 MHz, DMSO) 0 = 11.71 (s, 1 H), 7.62 (s, 2H), 7.36 (s, 1 H), 7.24 (m, 1 H), 7.13 (m, 4H), 6.91 (s, 1 H), 4.73 (m,

2H), 3.43 (m, 3H); LC-MS (M+H) + = 439. Example 18: Synthesis of Exemplary Compounds

The following compounds were prepared using the aforementioned methods or variations thereof and appropriate starting materials.

Example 19: HA01 Assay

Compound inhibition of HA01 enzymatic activity was determined using a kinetic fluorescence- based assay to quantitate the amount of hydrogen peroxide generated during the conversion of glycolic acid to glyoxylate. Recombinant HA01 at a concentration of 10 nM was added to test compounds dissolved in DMSO or DMSO alone as a control that had been dispensed into 384 well assay plates. After a 30-minute preincubation of HA01 with compound, hydrogen peroxide detection reagents were added such that the final concentrations were 50 pM Amplex Red and 0.5 U/ml horseradish peroxidase. The reaction was initiated by the addition of glycolic acid to a final concentration of 50 pM. The rate of fluorescence increase was read kinetically over 15 minutes using a Molecular Devices SpectraMax Paradigm Microplate Reader (ex. 563 nm, em. 587 nm). These rates were then plotted versus the compound concentrations to calculate IC50 values for the test compounds using a 4-parameter curve fit.

Table 2. HA01 Assay Data

‘+++’ = < 50 nM; ‘++’ = < 500 nM; ‘+’ = 500 -1000 nM

As shown in Table 2, compounds of the invention inhibit the conversion of glycolic acid to glyoxylate byHAOI with many of the compounds having IC50 values below 500 nM.

Example 20: CHO cell rescue assay

To illustrate the ability of compounds of the invention to prevent cell death from excess glyoxylate build up, the following cell-based assay is performed.

Protocol. CHO cells stably transformed with vector expressing glycolate oxidase (CHO-GO) are cultured in Ham’s F-12 /Glutamax media supplemented with 10% fetal bovine serum (FBS) and 100 units/mL penicillin/100 pg/mL streptomycin with additional supplementation of 400 pg/ml zeocin. CHO- WT cells are maintained in the same media conditions without the addition of Zeocin.

Cells are plated in 96-well plate (2000 cells/well) in standard medium (Ham’s F12 Glutamax F-12 with 10% FBS and antibiotics) and incubated with compound at different concentrations for 1 hour followed by addition of glycolic acid (final concentration 125 or 250 pM; Sigma). The final DMSO concentration in the assay is 0.5 %. Viability is monitored using live cells imager (IncuCyte Zoom) for up to 96 hours. CHO-WT cells are used as a control. Data Analysis. Data is analyzed using customized software developed internally. EC50 values (half maximum effective concentration) are calculated from dose-response curves using Prism software (Graph pad Software). At each compound concentration the % rescue is calculated as the % confluence of CHO-GO cells treated with the compound in presence of glycolic acid divided by the % confluence of CHO-GO cells treated with the compound in the absence of glycolic acid.

Other Embodiments

All publications, patents, and patent applications mentioned in this specification are incorporated herein by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Where a term in the present application is found to be defined differently in a document incorporated herein by reference, the definition provided herein is to serve as the definition for the term.

While the invention has been described in connection with specific embodiments thereof, it will be understood that invention is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims.

Other embodiments are in the claims.