Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PICOLINIC ACID DERIVATIVES AND USE THEREOF FOR TREATING DISEASES ASSOCIATED WITH ELEVATED CHOLESTEROL
Document Type and Number:
WIPO Patent Application WO/2020/237374
Kind Code:
A1
Abstract:
Formula I The present invention relates to compounds of Formula (I), wherein X is as defined above; Y is OR3; Z is H, optionally substituted alkyl or - CH2OR4; L is optionally substituted alkyl or heteroalkyl; R1 is H, -OH, optionally substituted alkyl, heterocycle, aryl or heteroaryl; R2 is NR5R6, or OR5; each of R3 and R4 is H or optionally substituted alkyl, benzyl, phenyl or heterocycle; or R3 and R4 combine to form an optionally substituted heterocycle; R5 is H or optionally substituted alkyl, cycloalkyl, heteroalkyl, alkylaryl or heterocycle; and R6 is H or alkyl, or pharmaceutically acceptable salts, solvates or compositions thereof. Compounds of formula (I) significantly increase low density lipoprotein receptor expression and activity, lower total circulating cholesterol and are useful for treating and delaying the onset of diseases associated with elevated cholesterol, such as atherosclerosis and hypercholesterolemia.

Inventors:
POIRIER STEVE (CA)
STRANIX BRENT RICHARD (CA)
MAYER GAÉTAN (CA)
Application Number:
PCT/CA2020/050721
Publication Date:
December 03, 2020
Filing Date:
May 27, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MONTREAL HEART INST (CA)
International Classes:
C07D491/056; A61K31/436; A61K31/44; A61K31/4439; A61K31/55; A61P3/06; A61P9/00; A61P9/10; C07D213/81; C07D401/06; C07D401/12
Domestic Patent References:
WO2019084681A12019-05-09
WO2019060850A12019-03-28
WO2009146555A12009-12-10
WO2011088549A12011-07-28
WO2019186164A12019-10-03
WO2017106547A12017-06-22
WO2015075082A12015-05-28
WO2014054425A12014-04-10
WO2011016234A12011-02-10
WO2019022637A12019-01-31
Foreign References:
US20060094761A12006-05-04
US20070299086A12007-12-27
Other References:
STRANIX BRENT R; WU JINZI J; MILOT GUY; BEAULIEU FRANÇIS; BOUCHARD JEAN-EMANUEL; GOUVEIA KRISTINE; FORTE ANDRÉ; GARDE SEEMA; WANG : "Pyridoxine hydroxamic acids as novel HIV-integrase inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 26, no. 4, 20 January 2016 (2016-01-20), pages 1233 - 1236, XP029409766, DOI: 10.1016/j.bmcl.2016.01.028
TRISTAN VERDELET; GUILLAUME MERCEY; NOBI CORREA; LUDOVIC JEAN; PIERRE-YVES RENARD: "Straightforward and efficient synthesis of 3-benzyloxy-4- bromopicolinate ester and 3-benzyloxy-5-bromopicolinate ester, common building blocks for pharmaceuticals and agrochemicals", TETRAHEDRON, vol. 67, no. 45, 8 September 2011 (2011-09-08), pages 8757 - 8762, XP028308713, DOI: 10.1016/j.tet.2011.09.024
W KORYTNYK, POTTI P G G: "Antagonists of Vitamin B6. Simultaneous and Stepwise Modification of the 2 and 4 Positions", JOURNAL OF MEDICINAL CHEMISTRY, vol. 20, no. 1, January 1977 (1977-01-01), pages 1 - 5, XP055762631
MORISAWA ET AL.: "Modification at the 2-Position of 4-Deoxypyridoxol and alpha4- Norpyridoxol", AGRICULTURAL AND BIOLOGICAL CHEMISTRY, vol. 40, no. 1, 1976, pages 101 - 105
MORISAWA YASUHIRO, KATAOKA MITSURU, WATANABE TAIICHIRO, KITANO NORITOSHI, MATSUZAWA TOSHIAKI: "Studies on Anticoccidial Agents. 1. Synthesis and Anticoccidial Activity of 4-Deoxypyridoxol and Its Esters", JOURNAL OF MEDICINAL CHEMISTRY, vol. 17, no. 10, 1 October 1974 (1974-10-01), pages 1083 - 1086, XP055762633
POCKER: "Synthesis of 2-Nor-2-formyIpyridoxal 5'-Phosphate, a Bifunctional Reagent Specific for the Cofactor Site in Proteins", J. ORG. CHEM., vol. 38, no. 25, 1973, pages 4295 - 4299
NIELS CLAUSON-KAAS, JOHN BRAMMER PETERSEN , GEORG OLE SORENSEN , GERT OLSEN , GERT JANSEN: "Preparation of derivatives of 3-hydroxypicolinic acid from furfural", ACTA CHEMICA SCANDINAVIAN, vol. 19, no. 5, 1965, pages 1147 - 1152, XP055381380, DOI: 10.3891/acta.chem.scand.19-1147
HEYL DOROTHEA, LUZ EILEEN, HARRIS STANTON A: "Chemistry of Vitamin B6. XII. Synthesis of 2,5-Bis-(hydroxymethyl)-3- hydroxy-4-methylpyridine, an Isomer of Pyridoxine", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 78, no. 17, 1 September 1956 (1956-09-01), pages 4474 - 4475, XP055762635
HONNORATY ET AL.: "Deracemization process of a-amino acids via pyridoxal. I. Synthesis and activity of polymerizable forms of pyridoxa l", BULLETIN DE LA SOCIETE CHIMIQUE DE FRANCE, vol. 132, no. 7, 1995, pages 709 - 720
KUZUHARA ET AL.: "Synthesis of a Chiral Pyridoxal Analogue as a Potential Catalyst for Stereospecific Nonenzymatic Reactions", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 99, no. 12, 8 June 1977 (1977-06-08), pages 4173 - 4175, XP055762636
Attorney, Agent or Firm:
VICKERS, Mark F. et al. (CA)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1 . A compound having the structure of formula (I):

Y is 0R3;

Z is H, optionally substituted C1-6 alkyl, or -CH20R4;

L is optionally substituted C1 -6 alkyl or optionally substituted C1-6 heteroalkyl;

Ri is H, -OH, optionally substituted C1-6 alkyl, optionally substituted C5-10 heterocycle, optionally substituted Ob-io aryl, or optionally substituted C5-10 heteroaryl;

R2 is NR5Re or OR5;

each of R3 and R4 is, independently, H, optionally substituted C1-6 alkyl, optionally substituted C1-6 fluoroalkyl, benzyl, phenyl, or optionally substituted C5-10 heterocycle; or R3 and R4 combine to form an optionally substituted heterocycle,

R5 IS H, optionally substituted C1 -6 alkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C1-6 fluoroalkyl, optionally substituted C1-6 alkyl Ce-io aryl, or optionally substituted C5-10 heterocycle;

R6 is H or optionally substituted C1 -6 alkyl;

or a pharmaceutically acceptable salt or solvate thereof.

O

2. The compound of claim 1 , wherein X is ¾*- 0Rs , H , or R, 6

3. The compound of claim 2, wherein R5 is methyl, propyl, cyclopropyl,

4. The compound of claim 2 or 3, wherein R6 is methyl.

OH

5. The compound of claim 1 , wherein

6. The compound of claim 5, wherein Rs is -CF3.

7. The compound of claim 1 , wherein X is cyano.

8. The compound of any one of claims 1-7, wherein Y is OH.

9. The compound of claim 8, wherein Z is methyl.

10. The compound of claim 8, wherein Z is -CH2OH.

1 1 . The compound of any one of claims 1-7, wherein Z is -CH2OR4.

12. The compound of claim 1 1 , wherein R3 and R4 combine to form an optionally substituted heterocycle.

13. The compound of claim 12, wherein the optionally substituted heterocycle is

16. The compound of claim 1 , wherein the compound has the structure of any one of compounds 1 -44:

or a pharmaceutically acceptable salt or solvate thereof.

17. A compound having the structure of any one of compounds 1 -44:

or a pharmaceutically acceptable salt or solvate thereof.

18. A pharmaceutical composition comprising the compound of any one of claims 1 -17, or a pharmaceutically acceptable salt or solvate thereof.

19. The pharmaceutical composition of claim 18, further comprising a

pharmaceutically acceptable carrier, excipient, or adjuvant.

20. A method of lowering cholesterol level in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

21 . A method of lowering elevated cholesterol level in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

22. A method of inducing expression of low density lipoprotein receptor (LDLR) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

23. The method of claim 22, wherein the expression is mRNA expression.

24. The method of claim 22, wherein the expression is protein expression.

25. A method of inducing activity of LDLR in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

26. The method of any one of claims 20-25, wherein:

(i) the subject has been diagnosed with an elevated cholesterol associated disease; or

(ii) the subject is predisposed to an elevated cholesterol associated disease.

27. A method of treating an elevated cholesterol associated disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

28. The method of claim 26 or 27, wherein the method reduces one or more symptoms of the elevated cholesterol associated disease in the subject.

29. A method of delaying the onset of an elevated cholesterol associated disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

30. A method of reducing the likelihood of occurrence of an elevated cholesterol associated disease in a subject in need thereof, the method comprising

administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

31. The method of claim 29 or 30, wherein the subject is predisposed to an elevated cholesterol associated disease.

32. The method of any one of claims 26-31 , wherein the elevated cholesterol associated disease is atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure.

33. The method of claim 32, wherein the elevated cholesterol associated disease is atherosclerosis.

34. The method of claim 32, wherein the elevated cholesterol associated disease is hypercholesterolemia.

35. A method of treating atherosclerosis, hypercholesterolemia,

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

36. The method of claim 33, wherein the method reduces one or more symptoms of atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic

complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure in the subject.

37. A method of delaying the onset of atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

38. A method of reducing the likelihood of occurrence of atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

39. The method of any one of claims 34-38, wherein the hypercholesterolemia is heterozygous familial hypercholesterolemia.

40. The method of any one of claims 34-38, wherein the hypercholesterolemia is homozygous familial hypercholesterolemia.

41. The method of any one of claims 20-40, further comprising administering to the subject a cholesterol lowering agent.

42. The method of any one of claims 20-40, wherein the subject has been treated with a cholesterol lowering agent prior to administering the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

43. The method of claim 42, wherein treating with the cholesterol lowering agent does not effectively lower cholesterol level in the subject.

44. The method of any one of claims 41-43, wherein the cholesterol lowering agent is a lipase inhibitor, an 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG CoA) reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a cholesterylester transfer protein (CETP) inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a microsomal triglyceride transfer protein (MTP) inhibitor, an Apolipoprotein B (ApoB) secretion inhibitor, a proprotein convertase subtilisin kexin type 9 (PCSK9) gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an acyl-coenzyme A:cholesterol acyltransferase (ACAT) inhibitor and a bile acid sequestrant, an HMG-CoA reductase gene expression inhibitor, or an HMG-CoA synthase gene expression inhibitor.

45. The method of any one of claims 41-44, wherein the cholesterol lowering agent is Atorvastatin, Fluvastatin, Lovastatin, Pravastatin, Rosuvastatin, Simvastatin, Pitavastatin, Ezetimibe, a combination of Ezetimibe and Simvastatin, Gemfibrozil, Fenofibrate, Clofibrate, Cholestyramine, Colestipol, Colesevelam, Alirocumab, Evolocumab, Inclisiran, AT04A, Lomitapide, or Mipomersen.

46. The method of any one of claims 41-44, wherein the cholesterol lowering agent is a statin.

47. The method of claim 46, wherein the statin is Atorvastatin, Fluvastatin, Lovastatin, Pravastatin, Rosuvastatin, Simvastatin, or Pitavastatin.

48. The method of any one of claims 20-47, further comprising administering to the subject an agent that increases bioavailability and/or slows metabolism of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof.

49. The method of claim 48, wherein the agent is an inhibitor of at least one isoform of cytochrome P450 (CYP450).

50. The method of claim 49, wherein the agent is an inhibitor of CYP1 A2, CYP2d6, CYP2C9, CYP2C19 or CYP3A4.

51. The method of claim 50, wherein the agent is an inhibitor of CYP3A4.

52. The method of claim 51 , wherein the agent is nefidipine or ritonavir.

53. The method of any one of claims 20-52, wherein the subject is a mammal.

54. A method of inducing expression of LDLR in a cell, the method comprising contacting the cell with an effective amount of the compound of any one of claims 1- 17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

55. The method of claim 54, wherein the expression is mRNA expression.

56. The method of claim 54, wherein the expression is protein expression.

57. A method of inducing activity of LDLR in a cell, the method comprising contacting the cell with an effective amount of the compound of any one of claims 1- 17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19.

58. The method of any one of claims 54-57, further comprising contacting the cell with a cholesterol lowering agent.

59. The method of claim 58, wherein the cholesterol lowering agent is a lipase inhibitor, an HMG CoA reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a MTP inhibitor, an ApoB secretion inhibitor, a PCSK9 gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an ACAT inhibitor and a bile acid sequestrant, an HMG-CoA reductase gene expression inhibitor, or an HMG-CoA synthase gene expression inhibitor.

60. The method of claim 58 or 59, wherein the cholesterol lowering agent is Atorvastatin, Fluvastatin, Lovastatin, Pravastatin, Rosuvastatin, Simvastatin, Pitavastatin, Ezetimibe, a combination of Ezetimibe and Simvastatin, Gemfibrozil, Fenofibrate, Clofibrate, Cholestyramine, Colestipol, Colesevelam, Alirocumab, Evolocumab, Inclisiran, AT04A, Lomitapide, or Mipomersen.

61. The method of claim 58 or 59, wherein the cholesterol lowering agent is a statin.

62. The method of claim 61 , wherein the statin is Atorvastatin, Fluvastatin, Lovastatin, Pravastatin, Rosuvastatin, Simvastatin, or Pitavastatin.

63. The method of any one of claims 54-62, further comprising contacting the cell with an agent that increases bioavailability and/or slows metabolism of the compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof.

64. The method of claim 63, wherein the agent is an inhibitor of at least one isoform of CYP450.

65. The method of claim 64, wherein the agent is an inhibitor of CYP1 A2,

CYP2d6, CYP2C9, CYP2C19 or CYP3A4.

66. The method of claim 65, wherein the agent is an inhibitor of CYP3A4.

67. The method of claim 66, wherein the agent is nefidipine or ritonavir.

68. The method of any one of claims 54-67, wherein the cell is a hepatic cell.

69. The method of any one of claims 54-68, wherein the cell is in a subject.

70. The method of claim 69, wherein the subject has been diagnosed with an elevated cholesterol associated disease.

71. The method of claim 70, wherein the elevated cholesterol associated disease is atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic

complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure.

72. The method of claim 71 , wherein the elevated cholesterol associated disease is atherosclerosis.

73. The method of claim 71 , wherein the elevated cholesterol associated disease is hypercholesterolemia.

74. The method of claim 73, wherein the hypercholesterolemia is heterozygous familial hypercholesterolemia.

75. The method of claim 73, wherein the hypercholesterolemia is homozygous familial hypercholesterolemia.

76. The method of any one of claims 69-75, wherein the subject is a mammal.

77. Use of the compound of any one of claims 1-17 or a pharmaceutically acceptable salt or solvate thereof in manufacture of a medicament for:

(i) lowering cholesterol in a subject or a cell;

(ii) inducing expression and/or activity of LDLR in a subject or a cell; and/or

(iii) treating, reducing one or more symptoms of, delaying the onset of and/or reducing the likelihood of occurrence of an elevated cholesterol associated disease in a subject

78. The use of claim 77, wherein the elevated cholesterol associated disease is atherosclerosis, hypercholesterolemia, hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure.

79. The use of claim 78, wherein the elevated cholesterol associated disease is atherosclerosis.

80. The method of claim 78, wherein the elevated cholesterol associated disease is hypercholesterolemia.

81 . The use of claim 80, wherein the hypercholesterolemia is heterozygous familial hypercholesterolemia.

82. The use of claim 80, wherein the hypercholesterolemia is homozygous familial hypercholesterolemia.

83. The compound of any one of claims 1 -17 or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 18 or 19 for use in:

(i) lowering cholesterol in a subject or a cell;

(ii) inducing expression and/or activity of LDLR in a subject or a cell; and/or

(iii) treating, alleviating one or more symptoms of, delaying the onset of and/or reducing the likelihood of occurrence of an elevated cholesterol associated disease in a subject.

84. The compound for use according to claim 83, wherein the elevated

cholesterol associated disease is atherosclerosis, hypercholesterolemia,

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure.

85. The compound for use according to claim 84, wherein the elevated cholesterol associated disease is atherosclerosis.

86. The compound for use according to claim 84, wherein the elevated cholesterol associated disease is hypercholesterolemia.

87. The compound for use according to claim 86, wherein the hypercholesterolemia is heterozygous familial hypercholesterolemia.

88. The compound for use according to claim 86, wherein the hypercholesterolemia is homozygous familial hypercholesterolemia.

Description:
PICOLINIC ACID DERIVATIVES AND USE THEREOF FOR TREATING DISEASES

ASSOCIATED WITH ELEVATED CHOLESTEROL

FIELD OF THE INVENTION

The present invention is directed to picolinic acid derived compounds of Formula I, pharmaceutically acceptable salts or solvates thereof, pharmaceutical compositions including one or more compounds of Formula I, methods of

synthesizing or manufacturing one or more compounds of Formula I, and use of one or more compounds of Formula I as modulators of low density lipoprotein receptor expression and activity. Compounds featured herein are useful for lowering total circulating cholesterol, and treating, reducing the symptoms of, delaying the onset of, reducing the likelihood of occurrence of, or delaying the progression of diseases that are associated with elevated cholesterol.

BACKGROUND OF THE INVENTION

Plasma apolipoprotein B (ApoB) lipoproteins such as low density lipoprotein (LDL) particles, which carry the majority of total circulating cholesterol, are cleared by binding to hepatic LDL receptor (LDLR), endocytosed and catabolized. An induction of LDLR leading to higher absorption of LDL, very low density lipoprotein (VLDL) and chylomicron remnants has been shown to be a method for significant reduction of circulating cholesterol.

Leaders of the medical profession have recently placed renewed emphasis on lowering plasma cholesterol and LDL cholesterol levels as an essential step in prevention of cardiovascular diseases (CVDs). Western populations are

predisposed to or at high risk of CVD. A disease of the arteries, atherosclerosis, is recognized to be the leading cause of death in Europe, Canada and the United States. Epidemiological investigation has firmly established hypercholesterolemia (high blood cholesterol) as a primary risk factor of CVD. Other risk factors include, being male, being a postmenauposal woman, age, hypertension, glucose

intolerance, obesity, familial history of CVD, metabolic syndrome, smoking, stress, unhealthy lifestyle, and left ventricular hypertrophy. The most important factor leading to atherogenesis (arterial plaque formation) is longstanding hypercholesterolemia and high circulating levels of LDL that result in cholesterol deposition in arterial vessels.

Successful treatment of hyperlipidemia in the general population, in diabetic subjects and in high CVD risk patients is of exceptional medical importance.

SUMMARY OF THE INVENTION

The present invention relates to a series of low density lipoprotein receptor (LDLR) inducers derived from picolinic acid esters and picolinamides, and pharmaceutically acceptable derivatives thereof (e.g., salts and solvates). Synthesis of novel compound libraries yielded potent inducers of LDLR expression in hepatic cells and led to significant lowering of circulating cholesterol in animal model. The compounds featured herein are useful for modulating LDLR activity and, in particular for inducing LDLR production and for treating elevated cholesterol and related conditions.

A first aspect features a compound having the structure of formula (I):

Y is OR 3 ;

Z is H, optionally substituted Ci- 6 alkyl, or -ChhOR^

L is optionally substituted Ci- 6 alkyl or optionally substituted Ci- 6 heteroalkyl;

Ri is H, -OH, optionally substituted Ci- 6 alkyl, optionally substituted C 5-10 heterocycle, optionally substituted Ce-io aryl, or optionally substituted C 5-10 heteroaryl;

R 2 is NR 5 Re or OR5;

each of R 3 and R 4 is, independently, H, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 fluoroalkyl, benzyl, phenyl, or optionally substituted C 5-10 heterocycle; or R 3 and R 4 combine to form an optionally substituted heterocycle, R 5 IS H, optionally substituted C 1 -6 alkyl, optionally substituted C 3-10 cycloalkyl, optionally substituted C 1-6 heteroalkyl, optionally substituted C 1-6 fluoroalkyl, optionally substituted C 1-6 alkyl Ce-io aryl, or optionally substituted C 5-10 heterocycle;

R 6 is H or optionally substituted C 1 -6 alkyl;

or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments,

In further embodiments, R 5 is methyl, propyl, cyclopropyl,

ln some embodiments, R6 is methyl.

OH

In some embodiments, X is A ¾ ^ 5 . In certain embodiments, R 5 is -CF 3 .

In some embodiments, X is cyano.

In some embodiments, Y is OH. In particular embodiments, Z is methyl or - CH 2 OH.

Alternatively, Y is OR 3 . In particular embodiments, Z is -CH 2 OR 4 . In some embodiments, R 3 and R 4 combine to form an optionally substituted heterocycle. In

particular embodiments, the optionally substituted heterocycle is

Some embodiments of the first aspect features a compound having the structure of any one of compounds 1 -44 of Table 1 , or a pharmaceutically acceptable salt or solvate thereof.

In another aspect is featured a compound having the structure of any one of compounds 1-44 of Table 1 , or a pharmaceutically acceptable salt or solvate thereof.

TABLE 1. Compounds of the Invention

Further provided herein are pharmaceutical compositions and pharmaceutically acceptable formulations containing a therapeutically effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the pharmaceutical composition may further include a pharmaceutically acceptable carrier, excipient, or adjuvant.

Also featured herein are methods of lowering cholesterol level (e.g., elevated cholesterol level) in a subject in need thereof (e.g., in bloodstream of a subject in need thereof) by administering to the subject a therapeutically effective amount (e.g., a cholesterol lowering amount) of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. Further provided are methods of inducing LDLR expression (e.g., mRNA expression and/or protein expression) and/or LDLR activity in a subject in need thereof (e.g., in a cell, such as a hepatic cell in a subject in need thereof) by administering to the subject a therapeutically effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, the subject has been diagnosed with or is predisposed to (e.g., at a risk of developing) a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease, such as atherosclerosis, hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia),

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure).

Further provided are methods of treating, reducing one or more symptoms of, delaying the onset of, and/or reducing the likelihood of occurrence of a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease) in a subject in need thereof by administering to the subject a therapeutically effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, the subject has been diagnosed with a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease). In other embodiments, the subject is predisposed to (e.g., at a risk of developing) a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease). Examples of diseases or conditions that are caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated diseases) include atherosclerosis, hypercholesterolemia (e.g., heterozygous familial

hypercholesterolemia or homozygous familial hypercholesterolemia),

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease, and congestive heart failure. In particular, diseases or conditions that are caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated diseases) include

atherosclerosis and hypercholesterolemia.

In some embodiments, any one of the methods featured herein further includes administering to the subject one or more cholesterol lowering agents. In some embodiments, the subject has been treated with one or more cholesterol lowering agents prior to administering a compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, treatment (e.g., prior treatment) with one or more cholesterol lowering agents does not effectively lower cholesterol level in the subject. Examples of cholesterol lowering agents (e.g., cholesterol lowering agents that can be

administered prior to, concurrent with, or after administering one or more compounds of the present invention) include a lipase inhibitor, an 3-hydroxy-3-methyl-glutaryl- coenzyme A (HMG CoA) reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a cholesterylester transfer protein (CETP) inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a microsomal triglyceride transfer protein (MTP) inhibitor, an Apolipoprotein B (ApoB) secretion inhibitor, a proprotein convertase subtilisin kexin type 9 (PCSK9) gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an acyl-coenzyme A:cholesterol acyltransferase (ACAT) inhibitor and a bile acid sequestrant, an HMG- CoA reductase gene expression inhibitor, and an HMG-CoA synthase gene expression inhibitor. In some embodiments, the cholesterol lowering agent (e.g., cholesterol lowering agent that can be administered prior to, concurrent with, or after administering one or more compounds of the present invention) is one or more of atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin,

pitavastatin, ezetimibe, a combination of ezetimibe and simvastatin, gemfibrozil, fenfibrate, clofibrate, cholestyramine, colestipol, colesevelam, alirocumab, evolocumab, inclisiran, AT04A, lomitapide, or mipomersen. In particular

embodiments, the cholesterol lowering agent (e.g., cholesterol lowering agent that can be administered prior to, concurrent with, or after administering one or more compounds of the present invention) is a cholesterol lowering agent listed in Table 3. In specific embodiments, the cholesterol lowering agent (e.g., cholesterol lowering agent that can be administered prior to, concurrent with, or after administering one or more compounds of the present invention) is a statin (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin

(Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)).

In some embodiments, any one of the methods featured herein further includes administering to the subject an agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention (e.g., one or more compounds of Formula I, such as one or more of compounds 1-44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, such agent (e.g., agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention) is an inhibitor of at least one isoform of cytochrome P450 (CYP450), such as an inhibitor of CYP1 A2, CYP2d6, CYP2C9, CYP2C19 or CYP3A4. In particular embodiments, such agent (e.g., agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention) is an inhibitor of CYP3A4 (e.g., nefidipine or ritonavir).

In some embodiments, any one of the methods featured herein further includes administering to the subject one or more cholesterol lowering agents, such as one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin

(Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the one or more compounds of the present invention (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

Further provided are methods of inducing LDLR expression (e.g., mRNA expression and/or protein expression) and/or LDLR activity in a cell (e.g., a hepatic cell) by contacting the cell with an effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, the method further includes contacting the cell with one or more additional cholesterol lowering agents (e.g., one or more of a lipase inhibitor, an HMG CoA reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a MTP inhibitor, an ApoB secretion inhibitor, a PCSK9 gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an ACAT inhibitor and a bile acid sequestrant, an HMG-CoA reductase gene expression inhibitor, or an HMG-CoA synthase gene expression inhibitor). In some

embodiments, the cholesterol lowering agent is one or more of atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, pitavastatin, ezetimibe, a combination of ezetimibe and simvastatin, gemfibrozil, fenfibrate, clofibrate, cholestyramine, colestipol, colesevelam, alirocumab, evolocumab, inclisiran, AT04A, lomitapide, or mipomersen. In particular embodiments, the cholesterol lowering agent is a cholesterol lowering agent listed in Table 3. In specific embodiments, the cholesterol lowering agent is a statin (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin

(Zocor®), or Pitavastatin (Livalo®)).

In some embodiments, the method further includes contacting the cell with an agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention (e.g., one or more compounds of Formula I, such as one or more of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, such agent (e.g., agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention) is an inhibitor of at least one isoform of

CYP450, such as an inhibitor of CYP1A2, CYP2d6, CYP2C9, CYP2C19 or CYP3A4. In particular embodiments, such agent (e.g., agent that increases bioavailability and/or slows metabolism of the one or more compounds of the present invention) is an inhibitor of CYP3A4 (e.g., nefidipine or ritonavir). In some embodiments, the method further includes contacting the cell with one or more cholesterol lowering agents, such as one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin

(Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the one or more compounds of the present invention (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

In some embodiments, the cell is in a subject. In some embodiments, the subject has been diagnosed with or is predisposed to (e.g., at a risk of developing) a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease, such as atherosclerosis,

hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure).

Further provided is the use of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of T able 1 ) or a pharmaceutically acceptable salt or solvate thereof for the

manufacture of a medicament for: (i) lowering cholesterol in a subject or a cell; (ii) inducing expression and/or activity of LDLR in a subject or a cell; and/or (iii) treating, reducing one or more symptoms of, delaying the onset of and/or reducing the likelihood of occurrence of a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease, such as atherosclerosis, hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia),

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject.

Also provided is at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1 -44 of Table 1 ) or a pharmaceutically acceptable salt or solvate thereof for use in: (i) lowering cholesterol in a subject or a cell; (ii) inducing expression and/or activity of LDLR in a subject or a cell; and/or (iii) treating, reducing one or more symptoms of, delaying the onset of and/or reducing the likelihood of occurrence of a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease, such as atherosclerosis, hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia),

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject.

In some embodiments of any one of the methods, use, and compounds for use featured herein, the subject is a mammal (e.g., a human).

In some embodiments of any one of the methods, use, and compounds for use featured herein, the disease or condition that is caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated disease) is atherosclerosis, hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure. In particular embodiment of any one of the methods, use, and compounds for use featured herein, the disease or condition that is caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated disease) is atherosclerosis. Alternatively or additionally, the disease or condition that is caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated disease) is hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia).

Chemical Terms

It is to be understood that the terminology employed herein is for the purpose of describing particular embodiments and is not intended to be limiting.

The term“acyl,” as used herein, represents a hydrogen or an alkyl group, as defined herein 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 1 1 , 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 "Ci- 6 alkyl" as used herein means saturated monovalent hydrocarbon radicals having straight or branched moieties and containing from 1 to 6 carbon atoms. Examples of such groups include, but are not limited to, methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, and tert-butyl.

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).

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).

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 invention can be an unsubstituted amino (i.e. , -NH 2 ) or a substituted amino (i.e., -N(R N1 ) 2 ).

The term“aryl,” as used herein, refers to an aromatic mono- or

polycarbocyclic 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 1H- 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 Ci- 6 alkyl Ce-io aryl, C M O alkyl Ce-io aryl, or Ci- 2 o alkyl Ce-io aryl), such as, benzyl and phenethyl. In some embodiments, the akyl and the aryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

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

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

The terms“carbocyclyl,” as used herein, refer to a non-aromatic C3-12 monocyclic, bicyclic, or tricyclic structure in which the rings are formed by carbon atoms. Carbocyclyl structures include cycloalkyl groups and unsaturated carbocyclyl radicals.

The term“cycloalkyl,” as used herein, refers to a saturated, non-aromatic, monovalent mono- or polycarbocyclic radical of three to ten, preferably three to six carbon atoms. This term is further exemplified by radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and adamantyl.

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

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. In some embodiments, the heteroalkyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkyl groups. 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. In some embodiments, the heteroalkenyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkenyl groups. 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. In some embodiments, the heteroalkynyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkynyl groups. 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 an aromatic mono- or polycyclic radical of 5 to 12 atoms having at least one aromatic ring containing one, two, or three ring heteroatoms selected from N, O, and S, with the remaining ring atoms being C. 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 Ci- 6 alkyl C2-9 heteroaryl, C1 -10 alkyl C2-9 heteroaryl, or C1-20 alkyl C2-9 heteroaryl). In some embodiments, the akyl and the heteroaryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

The term“heterocyclyl,” as used herein, denotes a mono- or polycyclic radical having 3 to 12 atoms having at least one ring containing one, two, three, or four ring heteroatoms selected from N, O or S, wherein no ring is aromatic. Examples of heterocyclyl groups include, but are not limited to, morpholinyl, thiomorpholinyl, furyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl, and 1 ,3-dioxanyl.

The term“heterocyclylalkyl,” as used herein, represents an alkyl group substituted with a heterocyclyl group. Exemplary unsubstituted heterocyclylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1 -6 alkyl C2-9 heterocyclyl, C1-10 alkyl C2-9 heterocyclyl, or C1 -20 alkyl C2-9 heterocyclyl). In some embodiments, the akyl and the heterocyclyl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

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,” 3 rd Edition (John Wiley & Sons, New York, 1999). /V-protecting groups include 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- 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, m ethoxy carbonyl, 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.

The alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, there will generally be 1 to 4 substituents present, unless otherwise specified. Substituents include, for example: 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., Nhh 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)).

Compounds of the invention 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 adsorbents 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 opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R," "S," "S * ," „ CjS M anc| "t rans " 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 of the invention 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 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.

Definitions

The term "solvate," as used herein, means a pharmaceutically acceptable solvate form of a compound of the present invention that retains the biological effectiveness of such compound. Examples of solvates include, but are not limited to, compounds of the invention in combination with water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine, or mixtures thereof. It is specifically contemplated that in the present invention one solvent molecule can be associated with one molecule of the compounds of the present invention, such as a hydrate. Furthermore, it is specifically contemplated that in the present invention, more than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a dihydrate. Additionally, it is specifically contemplated that in the present invention, less than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a hemihydrate. Furthermore, solvates of the present invention are contemplated as solvates of compounds of the present invention that retain the biological effectiveness of the non-hydrate form of the compounds. A "pharmaceutically acceptable salt" as used herein means a salt that retains the biological effectiveness of the free acids and bases of the specified derivative, containing pharmacologically acceptable anions or cations, and is not biologically or otherwise undesirable. Examples of pharmaceutically acceptable salts include, but are not limited to, acetate, acrylate, benzenesulfonate, benzoate (such as chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, and

methoxybenzoate), bicarbonate, bisulfate, bisulfite, bitartrate, borate, bromide, butyne-1 , 4-dioate, calcium edetate, camsylate, carbonate, chloride, caproate, caprylate, clavulanate, citrate, decanoate, dihydrochloride, dihydrogenphosphate, edetate, edislyate, estolate, esylate, ethylsuccinate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, heptanoate, hexyne-1 ,6-dioate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, y-hydroxybutyrate, iodide, isobutyrate, isothionate, lactate, lactobionate, laurate, malate, maleate, malonate, mandelate, mesylate, metaphosphate, methane-sulfonate, methylsulfate, monohydrogenphosphate, mucate, napsylate, naphthalene-1 -sulfonate,

naphthalene-2-sulfonate, nitrate, oleate, oxalate, pamoate (embonate), palmitate, pantothenate, phenylacetates, phenylbutyrate, phenylpropionate, phthalate, phospate/diphosphate, polygalacturonate, propanesulfonate, propionate, propiolate, pyrophosphate, pyrosulfate, salicylate, stearate, subacetate, suberate, succinate, sulfate, sulfonate, sulfite, tannate, tartrate, teoclate, tosylate, triethiodode, valerate salts, and cations, such as sodium, potassium, calcium, magnesium, ammonium, and tetraalkylammonium, among others.

The term "therapeutically effective amount," as used herein, means an amount of a compound of the present invention, or a pharmaceutically acceptable salt, solvate or composition thereof, that, when administered to a subject (e.g., a mammal) in need of such treatment, is sufficient to effect treatment, as defined herein.

The term "pharmaceutically acceptable formulation," or "pharmaceutical composition," as used herein, means a combination of a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, and a carrier, diluent, excipients, and/or adjuvants that are compatible with a compound of the present invention, and is not deleterious to the recipient thereof. Pharmaceutical formulations can be prepared by procedures known to those of ordinary skill in the art. For Example, the compounds of the present invention can be formulated with common excipients, diluents, carriers, or adjuvants, and formed into tablets, capsules, and the like. Examples of excipients, diluents, carriers, and adjuvants that are suitable for such formulations include the following: fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as

carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl pyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as povidone, sodium starch glycolate, sodium carboxymethylcellulose, agar agar, calcium carbonate, and sodium bicarbonate; agents for retarding dissolution such as paraffin; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adsorptive carriers such as keolin and bentonite; lubricants such as talc, calcium and

magnesium stearate and solid polyethylene glycols; and also, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethyleneglycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol, liposomes and wool fat. Final pharmaceutical forms may be pills, tablets, powders, lozenges, sachets, cachets, or sterile packaged powders, and the like, depending on the type of excipient used. Additionally, it is specifically contemplated that pharmaceutically acceptable formulations of the present invention can contain more than one active ingredient.

For example, such formulations may contain more than one compound according to the present invention. Alternatively, such formulations may contain one or more compounds of the present invention and one or more additional hypocholesterolemic agents. A pharmaceutically acceptable formulation may also include but is not limited to compounds, other than the compounds of formula I, having a structure such that, upon administration to a recipient or patient, a compound of this invention, active metabolite or residue thereof is directly or indirectly provided.

The term“elevated cholesterol associated disease,” as used herein, refers to a disease or condition that is caused or mediated by elevated cholesterol level (e.g., cholesterol level higher than that in a reference subject, such as a healthy subject). An elevated cholesterol associated disease can be caused by cellular and/or organ dysfunction stemming from elevated cholesterol level. Examples of elevated cholesterol associated disease include, but are not limited to atherosclerosis, hypercholesterolemia (e.g., heterozygous familial hypercholesterolemia or homozygous familial hypercholesterolemia), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease, and congestive heart failure. One or more elevated cholesterol associated diseases can be treated by one or more compounds, compositions, and methods described herein.

As used herein, the term“elevated cholesterol level” refers to cholesterol level (e.g., cholesterol level in blood) that is higher in a subject (e.g., a human with an elevated cholesterol associated disease) by 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) compared to a healthy control (e.g., a healthy human).

Elevated cholesterol level in a subject (e.g., in bloodstream of a subject) can be reduced by one or more compounds, compositions, and methods described herein.

As used herein, the term“hypercholesterolemia” refers to a condition that is caused or mediated by the presence of high levels of cholesterol in the blood. It is an elevated cholesterol associated disease that is characterized by high blood lipids and hyperlipoproteinemia or elevated levels of lipoproteins in the blood. Familial hypercholesterolemia is the familial or inherited form of hypercholesterolemia, and is characterized by high cholesterol levels, specifically, very high levels of low-density lipoprotein (LDL) in the blood and early cardiovascular disease. Familial

hypercholesterolemia is caused by mutation in the gene for the LDL receptor (LDLR), which is involved in passing LDL from the body. However, mutations in other genes (e.g., PCSK9 and the gene for Apolipoprotein B (ApoB)) can also cause inherited high cholesterol. Heterozygous familial hypercholesterolemia is familial hypercholesterolemia that is inherited from one parent, while homozygous familial hypercholesterolemia is familial hypercholesterolemia that is inherited from both parents. Hypercholesterolemia (e.g., familial hypercholesterolemia, such as heterozygous familial hypercholesterolemia and homozygous familial hypercholesterolemia) can be treated by one or more compounds, compositions, and methods described herein.

The term“coronary artery disease,” as used herein, includes atherosclerotic plaque prevention, regression, or stabilization, vulnerable plaque prevention, regression, or stabilization, vulnerable plaque area reduction, arterial calcification (e.g., calcific aortic stenosis), increased coronary artery calcium score, dysfunctional vascular reactivity, vasodilation disorders, coronary artery spasm, first myocardial infarction, myocardia re-infarction, ischemic cardiomyopathy, stent restenosis, PTCA restenosis, arterial restenosis, coronary bypass graft restenosis, vascular bypass restenosis, decreased exercise treadmill time, angina pectoris/chest pain, unstable angina pectoris, exertional dyspnea, decreased exercise capacity, ischemia, silent ischemia, increased severity and frequency of ischemic symptoms, and reperfusion after thrombolytic therapy for acute myocardial infarction.

The term“hypertension,” as used herein, is selected, but not limited to the group consisting of lipid disorders with hypertension, systolic hypertension, and diastolic hypertension.

The term“peripheral vascular disease,” as used herein, is selected, but not limited to the group consisting of peripheral vascular disease and claudication.

The term“diabetes”, as used herein, refers to any of a number of

diabetogenic states including type I diabetes, type II diabetes, Syndrome X,

Metabolic syndrome, lipid disorders associated with insulin resistance, impaired glucose tolerance, non-insulin dependent diabetes, microvascular diabetic complications, reduced nerve conduction velocity, reduced or loss of vision, diabetic retinopathy, increased risk of amputation, decreased kidney function, kidney failure, insulin resistance syndrome, pluri-metabolic syndrome, central adiposity

(visceral)(upper body), diabetic dyslipidemia, decreased insulin sensitization, diabetic retinopathy/neuropathy, diabetic nephropathy/micro and macro angiopathy and micro/macro albuminuria, diabetic cardiomyopathy, diabetic gastroparesis, obesity, increased glycosylated hemoglobin (including HbA1 C), impaired glucose control, impaired renal function (e.g., requiring dialysis, or end stage) and impaired hepatic function (e.g., mild, moderate, or severe).

“Metabolic syndrome,” also known as“SyndromeX,” refers to a common clinical disorder that is characterized by the presence of increased insulin

concentration in association with other disorders including visceral obesity, hyperlipidemia, dyslipidemia, hyperglycemia, hypertension, and potentially hyperuricemis and renal dysfunction.

The terms "treat", "treating", and "treatment" refer to any treatment of a disease or condition that is caused or mediated by elevated cholesterol level (e.g., an elevated cholesterol associated disease) in a subject, such as a mammal, particularly a human, and include: (i) preventing the disease or condition from occurring in a subject that may be predisposed to (e.g., at a risk of developing) the condition or reducing the likelihood of occurrence of the disease or condition in a subject that may be predisposed to (e.g., at a risk of developing) the condition, such that the treatment constitutes prophylactic treatment for the pathologic condition; (ii) modulating or inhibiting the disease or condition, i.e., arresting its development; (iii) delaying the onset of the disease or condition; (iv) relieving or reducing the disease or condition, i.e., causing regression of the disease or condition; or (v) relieving, reducing and/or alleviating the disease or condition or the symptoms resulting from the disease or condition, e.g., relieving or reducing an inflammatory response without addressing the underlying disease or condition.

As used herein, the terms“increase,”“increasing,”“induce” or“inducing” and “decrease,”“decreasing,”“reduce” or’’reducing” refer to modulating resulting in, respectively, greater or lesser amounts, of function, expression level, occurrence, or activity of a metric relative to a reference. For example, subsequent to

administration of one or more compounds described herein, cholesterol level may reduce or decrease in a subject by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) relative to cholesterol level prior to administration of the compounds. Also, subsequent to administration of one or more compounds described herein, one or more symptoms of an elevated cholesterol associated disease may reduce or decrease in a subject by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) relative to the symptoms of the disease prior to administration of the compounds. Furthermore, subsequent to administration of one or more compounds described herein, the likelihood or chance of occurrence of an elevated cholesterol associated disease may reduce or decrease in a subject by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) relative to the likelihood or chance of occurrence of the disease in a reference subject (e.g., a subject to whom the compounds has not been administered). Alternatively, subsequent to administration of one or more compounds described herein, LDLR expression (e.g., mRNA and/or protein expression) and/or LDLR activity may increase or induce in a subject by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) relative to LDLR expression and/or LDLR activity prior to administration of the compounds. Generally, the metric is measured subsequent to administration at a time that the administration has had the recited effect, e.g., at least one week, one month, 3 months, or 6 months, after a treatment regimen has begun. The term“reducing” is used interchangeably with the term“decreasing” herein. The term“increasing” is used interchangeably with the term“inducing” herein.

“Delaying the onset,” as used herein, refers to delaying or postponing the onset, start or occurrence of a condition relative to a reference. For example, subsequent to administration of one or more compounds described herein, the onset, start, or occurrence of an elevated cholesterol associated disease in a subject may be delayed or postponed by at least 1 month or more (e.g., 1 month, 2 months,

3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, 1 1 years, 12 years, 13 years, 14 years, 15 years, 16 years, 17 years, 18 years, 19 years, 20 years, or more) compared to a reference subject (e.g., a control subject to whom the compound has not been administered).

The term“predisposed to,” as used herein refers to being at a higher risk, having a higher chance, or having a higher likelihood of developing a condition or disease relative to a reference. For example, a subject predisposed to an elevated cholesterol associated disease may have a higher (e.g., at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20- 80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) risk, chance, or likelihood of developing the elevated cholesterol associated disease compared to a reference (e.g., a subject who is not predisposed to the disease).

The terms "co-administration", "co-administering",“co-administer”,“co administered”, or“combination therapy” as used herein, refer to the administration of a combination of at least a first agent and a second agent or two or more agents according to the present invention. Such co-administration can be performed such that two or multiple agents are part of the same composition or part of the same unitary dosage form. Co-administration also includes administering a first agent and a second agent, or more than two agents separately and as part of the same therapeutic regimen. The agents, if administered separately, need not necessarily be administered at essentially the same time, although they can be if so desired. Thus co-administration includes, for example, administering a first agent and a second agent as separate dosages or dosage forms, but at the same time. Co administration also includes separate administration at different times and in any order. Co-administration, as used herein, can include administration of more than one compound featured herein. Co-administration, as used herein, can also include administration of one or more of the featured compounds and an additional cholesterol lowering agent.

The terms "compound of the present invention," "compound of the invention," "compound featured herein," or“featured compound” refer to any of the above- mentioned compounds, as well as those in the Examples that follow, and include those generically described or those described as species. These terms also refers to pharmaceutically acceptable salts or solvates of these compounds. Such compounds include compounds of Formula I (e.g., compounds 1 -44 of Table 1 ).

The terms "comprising" and "including" as used herein, are used in their open, non-limiting sense.

Select abbreviations used throughout the application are provided in Table 2 below.

TABLE 2. Abbreviations Used

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1A and FIG. 1 B are graphs depicting half maximal effective

concentration (EC50) of Compound III (FIG. 1A) and Compound XXXVI (FIG. 1 B) on LDLR levels in HepG2 cells.

FIG. 2 is a graph depicting pharmacokinetic profile of Compound III in

Sprague Dawley rats.

DETAILED DESCRIPTION

The present disclosure describes picolinic acid derived compounds of Formula I (e.g., any one of compounds 1-44 of Table 1 ), pharmaceutically acceptable salts or solvates thereof, and pharmaceutical compositions including one or more of the compounds. Also featured herein are methods of synthesis or manufacture of the compounds and use of the compounds in inducing low density lipoprotein receptor (LDLR) expression and activity. Compounds featured herein are useful for lowering total circulating cholesterol, and treating, reducing the symptoms of, reducing the likelihood of occurrence of, delaying the onset of, or delaying the progression of diseases that are associated with elevated cholesterol.

Compounds

Featured in the disclosure are picolinic acid derived compounds of Formula I

(e.g., any one of compounds 1-44 of Table 1 ), or pharmaceutically acceptable salts or solvates thereof.

A compound featured herein may have the structure of formula (I):

Y is OR 3 ;

Z is H, optionally substituted Ci- 6 alkyl, or -CFhOR^ L is optionally substituted C1 -6 alkyl or optionally substituted C1-6 heteroalkyl; Ri is H, -OH, optionally substituted C1-6 alkyl, optionally substituted C5-10 heterocycle, optionally substituted Ob-io aryl, or optionally substituted C5-10 heteroaryl;

R 2 is N R5R6 or OR5;

each of R3 and R4 is, independently, H, optionally substituted C1-6 alkyl, optionally substituted C1-6 fluoroalkyl, benzyl, phenyl, or optionally substituted C5-10 heterocycle; or R 3 and R 4 combine to form an optionally substituted heterocycle,

R 5 IS H, optionally substituted C1 -6 alkyl, optionally substituted C3-10 cycloalkyl, optionally substituted C1-6 heteroalkyl, optionally substituted C1-6 fluoroalkyl, optionally substituted C1-6 alkyl Ce-io aryl, or optionally substituted C5-10 heterocycle;

R6 is H or optionally substituted C1 -6 alkyl;

or a pharmaceutically acceptable salt or solvate thereof.

In further embodiments, R 5 is methyl, propyl, cyclopropyl,

In some embodiments, R6 is methyl.

OH

In some embodiments, X is * ¾· Rs . In certain embodiments, R5 is -CF3.

In some embodiments, X is cyano.

In some embodiments, Y is OH. In particular embodiments, Z is methyl or -

CH 2 OH. Alternatively, Y is OR 3 . In particular embodiments, Z is -CH 2 OR 4 . In some embodiments, R 3 and R 4 combine to form an optionally substituted heterocycle. In

particular embodiments, the optionally substituted heterocycle is

In some embodiments, a compound of Formula I may have the structure of any one of compounds 1-44 of Table 1 , or a pharmaceutically acceptable salt or solvate thereof.

Also provided herein are compounds having the structure of any one of compounds 1-44 of Table 1 , or a pharmaceutically acceptable salt or solvate thereof.

Synthesis of compounds

The present invention also describes methods of synthesizing the compounds that are featured herein (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ).

The first approach (Scheme 1 ) starts from pyridoxine that is modified to produce intermediate I using methodologies described in Paul et al. (J Med Chem 1977, 20:745). Compound I is etherified by Williamson synthesis approach using methane sulphonyl chloride to generate intermediate II. The resulting mesylate/alkyl halide is then converted to intermediate Ilia, lllb or lllc with the appropriate nucleophiles; alcoholate, phenolate, thiolate, amine or heterocyclic anions.

Scheme 1

In another approach (Scheme 2), intermediate I is converted in the presence of a non-nucleophilic base to an alcoholate and reacted in situ to an electrophile such as an alkyl halide. In one particular case, the alkyl halide is a para-methoxy benzyl halide which generates the useful intermediate compound. This compound can be further modified to IV by heating with an excess alkylamine. The resulting amide may be deprotected from its acetonide form with formic acid to the form V.

In Scheme 3, the useful compounds are formed from the starting material pyridoxine VI by first protecting the compound as acetonide VII, followed by etherification to VIII through a procedure similar to Scheme 1 . The resulting compound is sequentially oxidized (IX, X, XI) to compounds of form Ilia. These compounds are then deprotected from the acetonide XII and the 4 position reduced to a methyl group by catalytic hydrogenation of the corresponding acetate. Reacting the ester with an amine either neat or in an appropriate solvent gives compound XIII. Compound I can be oxidized selectively to compound XIV using manganese dioxide and further reacted with carbanions such as -CF 3 to trifluoroalcohols XV. Compounds I and XV may be reacted with amines either neat or in the presence of solvent to the corresponding amides XVI or XVI I, which themselves may be deprotected of the acetonide with formic acid to the corresponding useful compounds XVIII and XIX.

Scheme 3

Scheme 4

Pharmaceutical composition

Pharmaceutical compositions contemplated herein include at least one compound of the present invention, and pharmaceutically acceptable salts, solvate or composition thereof, with a pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethyleneglycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, liposomes and wool fat. Compounds of the present invention that are basic may be prepared as a salt using suitable methods known in the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid; hydrobromic acid; sulfuric acid; nitric acid; phosphoric acid; and the like, or with an organic acid, such as acetic acid; maleic acid; succinic acid; mandelic acid; fumaric acid; malonic acid; pyruvic acid; oxalic acid; glycolic acid; salicylic acid; pyranosidyl acid, such as glucuronic acid or galacturonic acid; alpha-hydroxy acid, such as citric acid or tartaric acid; amino acid, such as aspartic acid or glutamic acid; aromatic acid, such as benzoic acid or cinnamic acid; sulfonic acid, such as p-toluenesulfonic acid or methanesulfonic acid; and the like.

It is understood by those skilled in the art that the compounds of the present invention, salts, or solvates thereof may exist in different crystal or polymorphic forms that are within the scope of the present invention and specified formulas.

Basic compounds of the present invention can form a variety of salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, it is common practice to first isolate the compound of the present invention as a pharmaceutically unacceptable salt and then convert to a free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition salt. The acid addition salts of the base compounds of this invention can be prepared by treating the base compound with a substantially equivalent amount of the selected mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or ethanol.

Compounds of the present invention that are acidic may be prepared as a salt using suitable methods known in the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary, or tertiary); an alkali metal or alkaline earth metal hydroxide; or the like. Examples of suitable salts include organic salts derived from amino acids such as glycine and arginine;

ammonia; primary, secondary, and tertiary amines; and cyclic amines, such as piperidine, morpholine, and piperazine; as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.

Acidic compounds of the present invention can form base salts with various pharmacologically acceptable cations. Examples of such salts include the alkali metal or alkaline-earth metal salts and particularly, the sodium and potassium salts, which can be prepared using conventional techniques. The chemical bases suitable as reagents in preparing the pharmaceutically acceptable base salts of this invention are those that form non-toxic base salts with the acidic compounds of the present invention. Such non-toxic base salts include those derived from such

pharmacologically acceptable cations as sodium, potassium calcium and

magnesium, etc. These salts can be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, preferably under reduced pressure. Alternatively, they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together, and then evaporating the resulting solution to dryness in the same manner as before. In either case, stoichiometric quantities of reagents are preferably employed in order to ensure completeness of reaction and maximum yields of the desired final product.

To treat or reduce the likelihood of occurrence of diseases or conditions caused or mediated by elevated cholesterol (e.g., elevated cholesterol associated diseases), a pharmaceutical composition including at least one of the compounds of the present invention is administered in a pharmaceutically acceptable formulation prepared by combining a therapeutically effective amount of the compound with one or more pharmaceutically suitable carriers including diluents, excipients and auxiliaries that facilitate processing of the active compounds into a pharmaceutically acceptable formulation. Carriers employed may be either solid or liquid. Exemplary solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid, and the like. Exemplary liquid carriers are syrup, peanut oil, olive oil, water, and the like. Similarly, the inventive compositions may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate, or the like. Further additives or excipients may be added to achieve the desired formulation properties. For example, a bioavailability enhancer, such: as Labrasol®, Gelucire®, or the like, or formulators, such as CHIC (carboxy- methylcellulose), PG (propyleneglycol), or PEG (polyethyleneglycol), may be added. Gelucire®, a semi-solid vehicle that protects active ingredients from light, moisture and oxidation, may be added, e.g., when preparing a capsule formulation. If a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form, or formed into a troche or lozenge. The amount of solid carrier may vary, but generally will be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation may be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or non- aqueous liquid suspension. The inventive compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g., parenteral or oral

administration.

To obtain a stable water-soluble dose form, a pharmaceutically acceptable salt of a compound of the present invention may be dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3 M solution of succinic acid or citric acid. If a soluble salt form is not available, the agent may be dissolved in a suitable co solvent or combinations of co-solvents. Examples of suitable co-solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin, and the like in concentrations ranging from 0-60% of the total volume. In an exemplary embodiment, a compound of the present invention is dissolved in DMSO and diluted with water. The composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution.

Pharmaceutical preparations for oral use can be obtained using a solid excipient in an admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include: fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl-cellulose, sodium

carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

The pharmaceutical compositions, comprising the compounds of the present invention may also contain suitable solid- or gel-phase carriers or excipients. These carriers and excipients may provide marked improvement in the bioavailability of poorly soluble drugs. Examples of such carriers or excipients include calcium carbonate, calcium, phosphate, sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Furthermore, additives or excipients such as Gelucire®, Capryol®, Labrafil®, Labrasol®, Lauroglycol®, Plurol®, Peceol®, Transcutol®, and the like may be used. Further, the pharmaceutical composition may be incorporated into a skin patch for delivery of the drug directly onto the skin.

The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those that increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral bioavailability, increase solubility to allow administration by injection, alter metabolism, or alter rate of excretion ( Pharmacokinetic Optimization in Drug

Research, Testa, B. et ai, 2001 , Wiley-VCH, VCHA).

The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally, or via an implanted reservoir, and are preferably administered orally or parenterally. The pharmaceutical compositions of this invention may contain any conventional non toxic pharmaceutically acceptable carriers, adjuvants or vehicles. The term “parenteral” or“parenterally” as used herein includes sub-cutaneous, intra- cutaneous, intra-venous, intra-muscular, intra-articular, intra-synovial, intra-sternal, intra-thecal, intra-lesional and intracranial injection or infusion techniques.

Pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer’s solution and isotonic sodium chloride solutions. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. Pharmaceutical compositions of the invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspension and solutions. In the case of tablets for oral administration, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral

administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring, and/or coloring agents may be added.

Pharmaceutical compositions of the invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient that is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax, and polyethylene glycols.

The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.

Dosage

Methods of treatment, their dosage levels and requirements featured herein may be selected by those of ordinary skill in the art from available methods and techniques.

It will be appreciated that the actual dosages of the compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) will vary according to the particular compound being used, the particular composition formulated, the mode of administration, and the particular site, host, and disease being treated. Those skilled in the art using conventional dosage-determination tests in view of the experimental data for a given compound may ascertain optimal dosages for a given set of conditions. For oral administration, an exemplary daily dose generally employed will be from about 0.001 to about 1000 mg/kg of body weight, with courses of treatment repeated at appropriate intervals, preferably between 0.01 and about 25 mg/kg body weight per day, and more preferably between about 0.5 and about 25 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of elevated cholesterol, including high circulating LDL.

Furthermore, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 750 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 mg to about 500 mg, or from about 100 mg to about 500 mg. Additionally, the pharmaceutically acceptable formulations of the present invention may contain a compound of the present invention, or a

pharmaceutically acceptable salt or solvate thereof, in an amount from about 0.5 w/w % to about 95 w/w %, or from about 1 w/w % to about 95 w/w %, or from about 1 w/w % to about 75 w/w %, or from about 5 w/w % to about 75 w/w %, or from about 10 w/w % to about 75 w/w %, or from about 10 w/w % to about 50 w/w %.

Typically, the pharmaceutical compositions of this invention will be

administered from about 1 to about 5 times per day, or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the patient treated and the particular mode of administration. A typical preparation will contain from about 5% to about 75% active compound (w/w). Preferably, such preparations contain from about 20% to about 50% active compound.

Upon improvement of a patient’s condition, a maintenance dose of a

compound, composition or combination of this invention may be administered if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained. When the symptoms have been reduced or alleviated to the desired level, treatment should cease, at least in principle. Patients may, however, require intermittent treatment on a long-term basis, upon any recurrence of disease symptoms, especially for high levels of cholesterol. As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease (e.g., elevated cholesterol associated disease), the patient’s disposition to the disease and the judgment of the treating physician.

With respect to the compounds of the present invention, the particular pharmaceutical formulation, the dosage, and the number of doses given per day to a mammal requiring such treatment are all choices within the knowledge of one of ordinary skill in the art and can be determined without undue experimentation.

Treatment

The compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) are also useful as commercial reagents that effectively lower circulating cholesterol. As commercial reagent, the compounds of this invention, and their derivatives, may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications. These and other uses that characterize commercial cholesterol lowering agents will be evident to those of ordinary skill in the art.

The compounds of the present invention can be used alone (monotherapy) or administered in combination with one or more additional cholesterol lowering agents for the treatment of high blood lipids associated diseases, such as diseases or conditions that are caused or mediated by elevated cholesterol level (e.g., elevated cholesterol associated diseases), including, but not limited to, atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure in a subject (e.g., a mammal, such as a human).

The compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) may be administered in combination with other cholesterol lowering agents (e.g., additional cholesterol lowering agents) that target other steps in the cholesterol metabolism. These agents include, but are not limited to: a lipase inhibitor, an 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG CoA) reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a cholesterylester transfer protein (CETP) inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a microsomal triglyceride transfer protein (MTP) inhibitor, an Apolipoprotein B (ApoB) secretion inhibitor, a proprotein convertase subtilisin kexin type 9 (PCSK9) gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an acyl-coenzyme A:cholesterol acyltransferase (ACAT) inhibitor and a bile acid sequestrant, an HMG-CoA reductase gene expression inhibitor, and an HMG-CoA synthase gene expression inhibitor. For example, one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of T able 1 ) may be administered in combination with one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)).

Combination therapies according to this invention may exert an additive or combined effect on reduction of elevated cholesterol because each therapeutic agent of the combination acts on a different site of cholesterol metabolism. The use of such combination therapies may also advantageously enable a reduction in the dosage of each cholesterol lowering agent, compared to administration of either agent alone as a monotherapy, while providing an equivalent or better therapeutic or prophylactic effect. Administration of lower doses of each therapeutic agent often reduces or even eliminates side effects or toxicity relative to monotherapy.

Furthermore, combination therapies may reduce the potential for the development of undesired side effects to the agents administered compared to monotherapy.

Administration of the compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination therapies with other agents to patients may be sequential or concurrent. One or more cholesterol lowering agents (e.g., one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®),

Simvastatin (Zocor®), or Pitavastatin (Livalo®))) may be administered to a subject (e.g., a mammal, such as a human (e.g., a human patient)) prior to, concurrent with, or following the administration of one or more compounds of Formula I (e.g., any one of compounds 1-44 of Table 1 ) featured herein. Furthermore, pharmaceutical or prophylactic compositions of this invention may include a combination of cholesterol lowering agent of this invention (e.g., one or more compounds of Formula I, such as any one of compounds 1-44 of Table 1 , featured herein) and another therapeutic or prophylactic agent, such as one or more additionalcholesterol lowering agents (e.g., one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin

(Zocor®), or Pitavastatin (Livalo®))).

In some embodiments, a subject (e.g., a mammal, such as a human (e.g., a human patient)) may have been treated with one or more cholesterol lowering agents (e.g., one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®),

Simvastatin (Zocor®), or Pitavastatin (Livalo®))) prior to administering one or more compounds of the present invention (e.g., at least one compound of Formula I, such as any one of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. In some embodiments, treatment (e.g., prior treatment) with one or more cholesterol lowering agents (e.g., one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®))) may not effectively lower cholesterol level in the subject.

Some embodiments of the current disclosure describe methods of inducing LDLR expression (e.g., mRNA expression and/or protein expression) and/or LDLR activity in a cell (e.g., a hepatic cell), such as a cell in a subject (e.g., a hepatic cell in a subject) by contacting the cell with an effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. Such methods may further include contacting the cell with one or more additional cholesterol lowering agents (e.g., one or more of a lipase inhibitor, an HMG CoA reductase inhibitor, an HMG CoA synthase inhibitor, an ATP citrate lyase inhibitor, a LDLR degradation inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase or cyclase inhibitor or a combination of both, a MTP inhibitor, an ApoB secretion inhibitor, a PCSK9 gene expression inhibitor, an anti-PCSK9 antibody, a PCSK9 mRNA silencer, a fibrate, a niacin or a combination of niacin with a statin, an ion-exchange resin, an ACAT inhibitor and a bile acid sequestrant, an HMG-CoA reductase gene expression inhibitor, or an HMG-CoA synthase gene expression inhibitor). For example, methods of inducing LDLR expression (e.g., mRNA expression and/or protein expression) and/or LDLR activity in a cell (e.g., a hepatic cell), such as a cell in a subject (e.g., a hepatic cell in a subject) may include contacting the cell with an effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1-44 of Table 1 , or a pharmaceutically acceptable salt, solvate or composition thereof) in combination with one or more cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin

(Zocor®), or Pitavastatin (Livalo®)).

Examples of cholesterol lowering agents useful for treating elevated

cholesterol, suitable for combination therapies with the compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ), and/or suitable for treating subjects (e.g., mammals, such as humans (e.g., human patients) prior to administration of compounds of this invention are listed in Table 3 below.

TABLE 3. Cholesterol Lowering Agents

Furthermore, compounds of the present invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) may be administered in combination with an additional agent or pharmaceutical composition that increases the bioavailability or slows the metabolism of the compounds. Agents or

pharmaceutical compositions that may increase the bioavailability or slow the metabolism of the compounds featured herein include inhibitors of at least one isoform of the cytochrome P450 (CYP450) enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4. Suitable agents that may be used to inhibit CYP 3A4 include, but are not limited to, nefidipine and ritonavir. For example, compounds of the present invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of Table 1 ) may be administered in combination with one or more inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir).

In some embodiments, compounds of the present invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) may be administered in combination with one or more cholesterol lowering agents (e.g., cholesterol lowering agents listed in Table 3, such as statins (e.g., Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®))) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

Such combinations may be administered such that a compound or compounds of the present invention are present in a single formulation or in the form of separate formulations that may be administered sequentially with an appropriate period of time in between or simultaneously. The choice of whether to include the compound or compounds of the present invention in the same formulation as the additional agent or agents is within the knowledge of one of ordinary skill in the art.

Some embodiments of the current disclosure describe methods of inducing LDLR expression (e.g., mRNA expression and/or protein expression) and/or LDLR activity in a cell (e.g., a hepatic cell), such as a cell in a subject (e.g., a hepatic cell in a subject) by contacting the cell with an effective amount of at least one compound of the present invention (e.g., at least one compound of Formula I, such as at least one of compounds 1 -44 of Table 1 ), or a pharmaceutically acceptable salt, solvate or composition thereof. Such methods may further include contacting the cell with an additional agent or pharmaceutical composition that increases the bioavailability or slows the metabolism of the compounds. Agents or pharmaceutical compositions that may increase the bioavailability or slow the metabolism of the compounds featured herein include inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4.

Suitable agents that may be used to inhibit CYP3A4 include, but are not limited to, nefidipine and ritonavir. Therapeutic effects

Featured herein are compositions and methods for reducing cholesterol level (e.g., cholesterol level in blood) in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) in need thereof by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents (e.g., cholesterol lowering agents listed in Table 3) and/or agents that increase the bioavailability or slow the metabolism of the compounds). In some embodiments, cholesterol level (e.g., cholesterol level in blood) can be reduced in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) in need thereof by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of T able 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)). In other embodiments, cholesterol level (e.g., cholesterol level in blood) can be reduced in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) in need thereof by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of T able 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1 A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, cholesterol level (e.g., cholesterol level in blood) can be reduced in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) in need thereof by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®),

Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

In certain embodiments, the subject may have elevated cholesterol level, such as cholesterol level (e.g., cholesterol level in blood) that is higher by 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100% (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more)) compared to a healthy control (e.g., a healthy human). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can reduce cholesterol level (e.g., cholesterol level in blood, such as elevated cholesterol level in blood) in a subject (e.g., a subject with an elevated cholesterol associated disease) by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to cholesterol level prior to administration of the compounds, or relative to cholesterol level in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compounds have not been administered). In some embodiments, one or more compounds of the invention, whether used in monotherapy or in combination therapy, can reduce cholesterol level in a subject (e.g., a subject with an elevated cholesterol associated disease) by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to cholesterol level prior to administration of the compounds, or relative to cholesterol level in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compound have not been administered).

Also featured herein are compositions and methods for inducing LDLR expression (e.g., mRNA and/or protein expression) in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds). In some embodiments, LDLR expression (e.g., mRNA and/or protein expression) can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL),

Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin

(Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)). In other embodiments, LDLR expression (e.g., mRNA and/or protein expression) can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

Alternatively, LDLR expression (e.g., mRNA and/or protein expression) can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®),

Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can induce LDLR expression (e.g., mRNA and/or protein expression) in a cell by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to LDLR expression prior to contacting with the compounds, or relative to LDLR expression in a reference cell, such as a control cell that has not been contacted with the compounds. In some embodiments, one or more compounds of the invention, whether used in monotherapy or in combination therapy, can induce LDLR expression in a cell by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to LDLR expression prior to contacting with the compounds, or relative to LDLR expression in a reference cell, such as a control cell that has not been contacted with the compounds.

Also featured herein are compositions and methods for inducing LDLR activity in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as compounds 1- 44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds). In some embodiments, LDLR activity can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)). In other embodiments, LDLR activity can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

Alternatively, LDLR activity can be induced in a cell, such as a cell in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) or a cell in a culture (e.g., a culture generated from a human (e.g., a human with an elevated cholesterol associated) sample, or a repository of human sample) by contacting the cell with one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1 -44 of T able 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin

(Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can induce LDLR activity in a cell by at least 5% or more (e.g., between 5- 20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to LDLR activity prior to contacting with the compounds, or relative to LDLR activity in a reference cell, such as a control cell that has not been contacted with the compounds. In some embodiments, one or more

compounds of the invention, whether used in monotherapy or in combination therapy, can induce LDLR activity in a cell by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to LDLR activity prior to contacting with the compounds, or relative to LDLR activity in a reference cell, such as a control cell that has not been contacted with the compounds.

Additionally, provided herein are compositions and methods for treating one or more elevated cholesterol associated disease (e.g., atherosclerosis,

hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds).

In some embodiments, one or more elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be treated in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin

(Livalo®)). In other embodiments, one or more elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be treated in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, one or more elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be treated in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®),

Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1 A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)).

Featured herein are compositions and methods for reducing one or more symptoms or indications of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject (e.g., a human, such as a human with an elevated cholesterol associated disease) in need thereof by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds). In some embodiments, one or more symptoms or indications of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®),

Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)). In other embodiments, one or more symptoms or indications of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, one or more symptoms or indications of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can reduce one or more symptoms or indications of an elevated cholesterol associated disease in a subject (e.g., a subject with the elevated cholesterol associated disease) by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to the symptoms or indications prior to administration of the compounds, or relative to the symptoms or indications in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compounds have not been administered). In some

embodiments, one or more compounds of the invention, whether used in

monotherapy or in combination therapy, can reduce one or more symptoms or indications of an elevated cholesterol associated disease in a subject (e.g., a subject with the elevated cholesterol associated disease) by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to the symptoms or indications prior to administration of the compounds, or relative to the symptoms or indications in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compounds have not been administered).

Also featured herein are compositions and methods for delaying the onset of an elevated cholesterol associated disease (e.g., atherosclerosis,

hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject (e.g., a human, such as a human predisposed to an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds). In some

embodiments, the onset of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin

(Livalo®)). In other embodiments, the onset of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, the onset of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®),

Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1 A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can delay the onset of an elevated cholesterol associated disease in a subject (e.g., a human, such as a human predisposed to an elevated cholesterol associated disease) by at least 1 month or more (e.g., 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months,

8 months, 9 months, 10 months, 1 1 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, 1 1 years, 12 years, 13 years, 14 years, 15 years, 16 years, 17 years, 18 years, 19 years, 20 years, or more) relative to a reference subject to whom the compounds have not been administered (e.g., a subject predisposed to the elevated cholesterol associated disease to whom the compounds have not been administered).

Also provided herein are compositions and methods for reducing the likelihood or chance of occurrence of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject (e.g., a human, such as a human predisposed to an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds).

In some embodiments, the likelihood or chance of occurrence of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia

(heterozygous and homozygous familial hypercholesterolemia included),

hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®),

Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)). In other embodiments, the likelihood or chance of occurrence of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, the likelihood or chance of occurrence of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be reduced in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin

(Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can reduce the likelihood or chance of occurrence of an elevated

cholesterol associated disease in a subject (e.g., a human, such as a human predisposed to an elevated cholesterol associated disease) by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to a reference subject to whom the

compounds have not been administered (e.g., a subject predisposed to the elevated cholesterol associated disease to whom the compounds have not been

administered). In some embodiments, one or more compounds of the invention, whether used in monotherapy or in combination therapy, can reduce the likelihood or chance of occurrence of an elevated cholesterol associated disease in a subject (e.g., a human, such as a human predisposed to an elevated cholesterol associated disease) by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to a reference subject to whom the compounds have not been administered (e.g., a subject predisposed to the elevated cholesterol associated disease to whom the compounds have not been administered). Also featured herein are compositions and methods for delaying the progression of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) alone (monotherapy) or in combination (combination therapy) with one or more additional agents (e.g., cholesterol lowering agents and/or agents that increase the bioavailability or slow the metabolism of the compounds).

In some embodiments, the progression of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, LescoKD XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®), Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin

(Livalo®)). In other embodiments, the progression of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more agents that increase the bioavailability or slow the metabolism of the compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). Alternatively, the progression of an elevated cholesterol associated disease (e.g., atherosclerosis, hypercholesterolemia (heterozygous and homozygous familial hypercholesterolemia included), hypertriglyceridemia, diabetic complications, dyslipidemia, hyperlipidemia, hypoalphalipoproteinemia, metabolic syndrome, stroke, vascular dementia, chronic kidney disease, coronary heart disease, coronary artery disease, retinopathy, inflammation, thrombosis, peripheral vascular disease or congestive heart failure) can be delayed in a subject in need thereof (e.g., a human, such as a human with an elevated cholesterol associated disease) by administering to the subject one or more compounds of this invention (e.g., compounds of Formula I, such as any one of compounds 1-44 of Table 1 ) in combination with one or more statins (e.g., one or more statins listed in Table 3, such as Atorvastatin (Lipitor®), Fluvastatin (Lescol®, Lescol® XL), Lovastatin (Mevacor®, Altoprev®), Pravastatin (Pravachol®),

Rosuvastatin (Crestor®), Simvastatin (Zocor®), or Pitavastatin (Livalo®)) and one or more agents that increase the bioavailability or slow the metabolism of the

compounds (e.g., inhibitors of at least one isoform of CYP450 enzymes, preferably inhibitors of CYP1A2, CYP2d6, CYP2C9, CYP2C19 and CYP3A4, such as inhibitors of CYP3A4 (e.g., nefidipine and ritonavir)). One or more compounds of the invention, whether used in monotherapy or in combination therapy, can delay the progression of an elevated cholesterol associated disease in a subject (e.g., a subject with the elevated cholesterol associated disease) by at least 5% or more (e.g., between 5-20%, between 5-50%, between 10-50%, between 10-80%, between 20-80%, or between 20-100%) relative to the progression prior to administration of the compounds, or relative to progression in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compounds have not been administered). In some embodiments, one or more compounds of the invention, whether used in

monotherapy or in combination therapy, can delay the progression of an elevated cholesterol associated disease in a subject (e.g., a subject with the elevated cholesterol associated disease) by at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more relative to the progression prior to administration of the compounds, or relative to progression in a reference subject to whom the compounds have not been administered (e.g., a subject with the elevated cholesterol associated disease to whom the compounds have not been

administered).

EXAMPLES

The following examples are put forth to provide those of ordinary skill in the art with a description of how the compounds described herein may be synthesized and used, and how the methods featured herein may be evaluated. The examples are intended to be purely exemplary of the invention and are not intended to limit the scope of the claims.

Method

Preparative chromatography was performed by flash chromatography, using silica gel 60 (EM Science) with the indicated solvent systems and positive air pressure, to allow for a proper rate of elution. Detection of the compounds was carried out by exposing eluted plates (analytical or preparative) to iodine, UV light and/or treating analytical plates with a 2% solution of p-anisaldehyde in ethanol containing 3% sulfuric acid and 1 % acetic acid, followed by heating. Alternatively, analytical plates can be treated with a 0.3% ninhydrin solution in ethanol containing 3% acetic acid and/or a CAM solution made of 20 g (NH4) q Mq7q24 and 8.3 g

Ce(SC>4)2 polyhydrate in water (750 mL) containing concentrated sulfuric acid (90 mL) and/or silica gel infused with l 2 .

Unless otherwise indicated, all starting materials were purchased from a commercial source such as Aldrich Co. or Sigma Co; mass spectra were recorded on a Hewlett Packard LC/MSD 1 100 system APCI either in negative mode or positive mode; nuclear magnetic resonance (NMR) spectra were recorded on a Bruker AMX 500 equipped with a reversed or QNP probe.

Samples were dissolved in deutero-chloroform (CDC ), deuterium oxide (D2O), deuterated Methanol (MeOD), or deutero-dimethylsulfoxide (DMSO-d 6 ) for data acquisition, and tetramethylsilane was used as internal standard. Chemical shifts (q) are expressed in parts per million (ppm), coupling constants (J) are expressed in hertz (Hz) and multiplicities are denoted as s for singlet, d for doublet, dd for doublet of doublets, t for triplet, q for quartet, quint for quintet, m for multiplet, and br s for broad singlet.

Example 1.

Methyl-5-((5-Fluoro-1 H-indol-1 -yl)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5- c]pyridine-8-carboxylate

In a procedure similar to Scheme 1 , methyl 5-(hydroxymethyl)-2,2-dimethyl- 4H-[1 ,3]dioxino[4,5-c]pyridine-8-carboxylate (Paul et al. , J Med Chem, 1977, 20:745) was dissolved in DCM and cooled to 0°C.

Triethyl amine (TEA) was added and methanesulphonyl chloride added dropwise over 20 minutes. TLC revealed a complete conversion of the starting material. Meanwhile, 4-fluoroindole was dissolved in tetrahydrofuran THF and potassium tert-butoxide (KOtB) was added. The dichloromethane solution was evaporated without heating and the solvent replaced with THF. The methyl 5- (methansulphoxymethyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8-carboxylate THF solution was dripped slowly over 20 minutes into the Indole solution and left to warm to room temperature (RT). A gel formed, and the solution was then heated with a heat gun to liquefy. After 2h, TLC revealed completion of the reaction. The THF was partially removed and EtAc was added. This solution was then extracted with 5% citric acid followed by 10% NaC03 and finally a 50% NaCI (brine) solution. The organic phase was then evaporated to yield the product.

1 H-NMR (500MHz, MeOD) : d = 7.52 (s, 1 H), 7.34 (m 2H), 6.88 (m 2H), 6.50 (s 2H), 5.49 (s 1 H), 4.81 (s, 2H), 4.75(s, 2H), 3.8(s 3H), 1 .50 (s6H); MS-ESI m/z 371.3

[MH] + Example 2.

N-Cyclopropyl-5-((5-Fluoro-1 H-indol-1-yl)methyl)-2,2-dimethyl-4H-

[1 ,3]dioxino[4,5-c]pyridine-8-carboxamide

The product from Example 1 was dissolved in neat cyclopropyl amine and heated to 55 °C for 12 h. TLC showed 1 product.

1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 7.12 (d 2H), 5.40 (d 2H), 5.01 1 (d 1 H), 4.56 (s, 2H), 4.00(s, 3H)1 6(s 6H); MS-ESI m/z 383.34 [MH] + Example 3.

N-Cyclopropyl-3-hydroxy-4-(methyl)-5-((4- methoxybenzyloxy)methyl)picolinamide Example 3a

5-((4-Methoxybenzyloxy)methyl)-2,2,8-trimethyl-4H-[1,3]di oxino[4,5-c]pyridine

In a procedure similar to that illustrated in Scheme 3, anhydrous THF (200 ml) was added to 2.2 g NaH (60%, mmol) at 0°C under a nitrogen atmosphere. To this suspended mixture was added a solution of 7g 4-methoxybenzyl alcohol in THF (400 ml). The resulting mixture was refluxed for 30 min and a precipitate accumulated during the reflux. After cooling to RT, 1 1 g (0.05 mol) 5-(methansulphoxymethyl)-2,2- dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8-methyl was introduced dropwise and the resulting mixture was refluxed for another 8 h. The reaction was quenched carefully by adding ice-cold water to the viscous mixture at 0 °C and diluted with a saturated ammonium chloride solution followed by extraction with methylene chloride. The combined organic extracts were washed with brine, dried (Na 2 S0 4 ), and

concentrated to give a brown oil. The crude product was purified by chromatography (10% ethyl acetate/petroleum ether) to afford the title compound (6.2g, 37%): LC-MS

(M+H) + m/z 331.

1 H-NMR (500MHz, MeOD) : d = 7.82 (s, 1 H), 7.24 (d 2H), 6.88 (d 2H), 4.81 (s, 2H), 4.55(s, 2H), 3.83 (d 2H), 3.26 (3H), 1.50 (s6H)

Example 3b

5-((4-Methoxybenzyloxy)methyl)-2,2,8-trimethyl-4H-[1,3]di oxino[4,5-c]pyridine

7-oxide

5-((4-Methoxybenzyloxy)methyl)-2,2,8-trimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine (6 g, 18 mmol, 1.0 equiv) was dissolved in dry CHC (200 mL) and the solution was cooled to 0 °C. To this solution was added m-chloroperbenzoic acid (85% purity of the reagent, 5.0 g, 1 .2 equiv). After being stirred at 23 °C for 12 h, the reaction mixture was extracted with Na 2 SO 3 (10%, 2x200 mL), NaHCC> 3 (5%, 2x200 mL), H 2 0, dried (Na 2 SC>4), filtered and the solvent was removed under reduced pressure. The crude product was purified by chromatography (10% methanol/ethyl acetate) to afford the title compound as pale-yellow solid (35 g, 68%); LC-MS (M+H) + m/z 346.

1 H-NMR (500MHz, MeOD) : d = 7.92 (s, 1 H), 7.24 (d 2H), 6.89 (d 2H), 4.81 (s, 2H), 4.55(s, 2H), 4.44 (d 2H), 3.76 (3H), 2.40 (s3H)1.50 (s6H)

Example 3c

(5-((4-Methoxybenzyloxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridin-8- yl)methanol

Trifluoroacetic anhydride (3.5 mL, 32 mmol) was added to a solution of 5-((4- methoxybenzyloxy)methyl)-2,2,8-trimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine 7-oxide (5.0 g, 61 mmol) in dry CH2CI2 (200 ml) at 0 °C and stirred for 5 min. An additional amount of trifluoroacetic anhydride (10.5 mL) was added and the reaction mixture was stirred overnight at 23 °C. Then, the reaction mixture was cooled to 0 °C and MeOH (150 mL) was added while stirring was continued. The solvents were evaporated and the resulting residue was dissolved in CH2CI2 and washed with Na2CC>3 (20% aqueous) and H2O until pH was neutral. The organic phase was dried (Na2SC>4), filtered and concentrated in vacuo. The residue was crystallized from EtOH-CH 2 CI 2 to afford the title compound (4.45 g, 93%); LC-MS (M+H) + m/z 346.

Example 3d

5-((4-Methoxybenzyloxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8- carbaldehyde

To a solution of (5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H- [1 ,3]dioxino[4,5-c]pyridin-8-yl)methanol (4.40 g, mmol) in DCM (500 mL) was added MnC>2 20.5 g, 128 mmol) and the suspension was heated to reflux for 4 h. The precipitate was removed by filtration and the filtrate was concentrated under reduced pressure to afford the title compound (4.0 g, 95%). The crude material was used for the next step without further purification: LC-MS (M+H) + m/z 344. Example 3e

Methyl 5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H-[1,3]dioxino[ 4,5- c]pyridine-8-carboxylate

To the solution of 5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H- [1 ,3]dioxino[4,5-c]pyridine-8-carbaldehyde (4.0 g) in anhydrous MeOH (120 mL),

KOH (85%, 5.3 g, 78 mmol) and iodine (9.9 g, 39 mmol) were added at 0 °C. The reaction mixture was kept at 23 °C and stirred for 12 h until no starting material was detected by TLC. Then, the solution was treated with Na 2 S0 3 (solid) and the pH was adjusted to 7. The solid was filtered and solvent was removed under reduced pressure. The residue was dissolved in EtOAc and washed with water. The combined organic extracts were dried (Na2S04), filtered and concentrated in vacuo. The crude residue was purified by chromatography (S1O2) with petroleum etherethyl acetate (5:1 ) as eluent to afford the title compound as pale-yellow solid (3.8 g, 78%): LC-MS (M+H) + m/z 374.

Example 3f

Methyl 3-hydroxy-4-(hydroxymethyl)-5-((4-methoxybenzyloxy)methyl)pi colinate

The solution of methyl 5-((4-methoxybenzyloxy)methyl)-2,2-dimethyl-4H- [1 ,3]dioxino[4,5-c]pyridine-8-carboxylate (3.8 g, 23.6 mmol) in 200 mL of HCI/MeOH was stirred at 23 °C for 24 h. MeOH (500 mL) was added to dissolve the suspension and NaHCCb (solid) was added to neutralize the reaction mixture. The excess solid was filtered and the solvent was removed under reduced pressure. The residue was dissolved in EtOAc and washed with water. The combined organic extracts were dried (Na2SC>4), filtered and concentrated in vacuo to afford the title compound as a light yellow solid (3.0 g, 100%): LC-MS (M+H) + m/z 334. Example 3g

Methyl 3-acetoxy-4-(acetoxymethyl)-5-((4-methoxybenzyloxy)methyl)pi colinate

To the solution of methyl 3-hydroxy-4-(hydroxymethyl)-5-((4- methoxybenzyloxy)methyl)picolinate (3.5g) in EtOAc was added acetic anhydride (2.2 eq) and triethylamine. The solution was left for 4 h (TLC completion) and then extracted with citric acid (10%), dried (Na2SC>4), filtered and concentrated in vacuo. The compound was then dissolved into a solution of Pd/C (500mg) in EtOAc

(degassed and N 2 flushed). Hydrogen gas was then slowly bubbled in over 4 h with TLC monitoring (1 : 1 Hexanes Ethyl Acetate). The reaction was degassed, flushed with argon and filtered through a pad of Celite. Evaporation of the solvent yielded the desired product.

N-Cyclopropyl 3-hydroxy-4-(methyl)-5-((4- methoxybenzyloxy)methyl)picolinamide

Methyl 3-hydroxy-4-(methyl)-5-((4-methoxybenzyloxy)methyl)picolinat e (500 mg) was dissolved in neat cyclopropylamine and heated to 55 °C for 12h. TLC monitoring showed complete conversion. Evaporation of the amine followed by trituration with 1 % citric acid yielded a white solid that was filtered off and dried.

1 H-NMR (500MHz, MeOD) : d = 8.02 (s1 H), 7.32 (d 2H), 6.88 (d 2H), 4.81 (s, 2H), 4.55(m, 2H), 3.73 (s3H), 2.7-2.9 (m 1 H) 0.50-0.77 (dd 3H); MS-ESI m/z 343.2 [MH] +

Example 4.

N-Cyclopropyl-5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]- pyridine-8-carboxamide

Compound 28b of US 8,742,123 Methyl 5-((4-fluorophenoxy)methyl)-3- hydroxy-4- (hydroxymethyl)picolinate was used, dissolved in neat cyclopropylamine and refluxed for 12 h at 59 °C.

Evaporation of the amine and flash chromatography on silica gel (EtoAc eluent) gave the desired product. 1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 6.88 (d 2H), 5.40 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H) 0.50-0.77 (dd 3H); MS- ESI m/z 333.1 [MH] +

Example 5.

methyl 5-[(1,3-dioxo-2,3-dihydro-1 H-isoindol-2-yl)methyl]-2,2-dimethyl-2H,4H- [1 ,3]dioxino[4,5-c]pyridine-8-carboxylate

In a procedure similar to that shown in Scheme 1 , phthalimide was dissolved in a solution of THF with KOtBu. To this was dropped a solution of methyl 5- (methansulphoxymethyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8-carboxylate THF solution (Examplel ). TLC showed complete conversion. The solvent was evacuated and EtoAc added. Extraction with 5% Citric acid followed by 10%

Na2CC>3 and brine gave a clear organic phase. The residue was then evacuated and spontaneously crystallized.

1 H-NMR (500MHz, MeOD) : d = 8.21 (s, 1 H), 7.64 (d 2H), 7.92 (d 2H), 5.65 (s 2H), 5.01 1 (d 1 H), 4.56 (s, 2H), 3.98(s, 3H)1 6(s 6H); MS-ESI m/z 414 [MH] +

Example 6.

methyl 5-{[(2H-1,3-benzodioxol-5-yl)amino]methyl}-2,2-dimethyl-2H,4 H- [1 ,3]dioxino[4,5-c]pyridine-8-carboxylate Methyl 5-formyl-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]picolinate, (J Org Chem, 1999, 64:4537) was added to acetic acid in methanol. 3,4 methylenedioxy aniline was added and the mixture stirred for 30 minutes before sodium cyanoborohydride was added. The mixture was stirred for 12 h and the solvent evacuated. Addition of EtoAc followed by extraction with 10% AcOH, followed by 10% Na 2 C0 3 and brine. The EtoAc was evacuated and the dark residue flashed chromatographed on silica gel (EtoAc eluent). The solvent was evacuated and the residue crystallized.

1 H-NMR (500MHz, MeOD) : d = 8.19 (s, 1 H), 6.6 (d 1 H), 6.3 (d 1 H), 6.1 1 (m

1 H)5.80 (d 2H), 5.53 (d 2H), 1 6(s 6H); MS-ESI m/z 371 .2 [MH] +

Example 7.

N-Cyclopropyl-5-(2-trifluoroethanol)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5- c]pyridine-8-carboxamide In a procedure similar to that illustrated in Scheme 4, methyl 5-formyl-2,2- dimethyl-4H-[1 ,3]dioxino[4,5-c]picolinate, (J Org Chem, 1999, 64:4537) was dissolved in DCM. Trimethylsilyltrifluoromethane was added and stirred at 0 °C. 2mg, (~1 %) tetramethylammonium fluoride (TMAF) was added and the reaction proceeded. After 1 h, the DCM was evacuated and the residue flashed

chromatographed on silica gel (EtoAc) to yield the desired product.

1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 5.40 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H), 1 6(s 6H), 0.50-0.77 (dd 3H); MS-ESI m/z 347.3[MH] +

Example 8.

Methyl (2S)-1 -{[8-(methoxycarbonyl)-2,2-dimethyl-2H,4H-[1 ,3]dioxino[4,5- c]pyridin-5-yl]methyl}pyrrolidine-2-carboxylate

The compound was made in a similar fashion as illustrated in Scheme 1 , using D-proline methyl ester.

1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 7.12 (d 2H), 5.40 (d 2H), 5.01 1 (d 1 H), 4.56 (s, 2H), 4.00(s, 3H)1 6(s 6H); MS-ESI m/z 414 [MH] + Example 9.

N-Cyclopropyl-5-(Hydroxymethyl)-2, 2-dimethyl -4H-[1 ,3]dioxino[4,5-c]pyridine- 8-carboxamide

As illustrated in Scheme 4, the procedure is as follows; methyl 5- (hydroxymethyl-2, 2-dimethyl-4H-[1 ,3]dioxino[4,5-c]picolinate, (J Org Chem, 1999,

64:4537) was dissolved in neat cyclopropylamine. The mixture was stirred at 60 °C for 12 h until TLC indicated completion. The amine was evaporated and the residue flashed chromatographed on silica gel (EtoAc eluent). The solvent was evacuated and the residue crystallized. 1 H-NMR (500MHz, MeOD) : d = 8.1 1 (s, 1 H), 4.8 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H), 1 6(s 6H), 0.50-0.77 (dd 3H); MS-ESI m/z 279.3 [MH] + Example 10.

Cyclopropyl- 5-(hydroxymethyl)-3-hydroxy-4-hydroxymethyl-picolinamide

The compound from the above example was dissolved in 90% formic acid and heated at 75 °C for 4 h. The solvent was evacuated under vacuum, the residue triturated with water, and then the residue flashed chromatographed on silica gel (EtoAc eluent). The solvent was evacuated and the residue crystallized.

1 H-NMR (500MHz, MeOD) : d = d = 8.1 1 (s, 1 H), 4.98 (d 2H), 4.66 (s, 2H), 2.7-2.9 (m 1 H), 0.50-0.78 (dd 3H); MS-ESI m/z 414 [MH] +

Example 11.

N-(n-Propyl)- 5-(4-Methoxy-benzyloxymethyl)-3-hydroxy-4-hydroxymethyl- picolinamide

It was synthesized in a procedure similar to that of Scheme 3, Example 2, using propyl amine.

1 H-NMR (500MHz, MeOD) : d = 8.16 (s, 1 H), 7.31 (d 2H), 6.94 (d 2H), 4.7 (m 4H), 4.5 (m 2H), 4.56 (s, 2H), 3.37 (s 3H), 3.3 (m 2H) 1 6(s 2H), 1 .00 ( m 3H); MS-ESI m/z 361.4 [MH] + Example 12.

N-(n-Propyl)- 5-(4-Methoxy-benzyloxymethyl)-3-hydroxy-4-methyl-picolinamid e

It was synthesized in a procedure similar to that of Scheme 3, Example 2, using n-propyl amine.

1 H-NMR (500MHz, MeOD) : d = 8.16 (s, 1 H), 7.31 (d 2H), 6.94 (d 2H), 4.7 (m 4H), 4.5 (m 2H), 3.93 (s 3H), 3.79(s, 3H), 3.3 (m 2H) 1 6(s 2H), 1.00 ( m 3H); MS-ESI m/z 345.4 [MH] + Example 13.

3-hydroxy-4,5-bis(hydroxymethyl)-N-propylpyridine-2-carbo xamide

The desired compound was made in a procedure similar to Example 10, using n-propyl amine. 1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 7.12 (d 2H), 5.40 (d 2H), 5.01 1 (d 1 H), 4.56 (s, 2H), 4.00(s, 3H)1 6(s 6H); MS-ESI m/z 241.1 [MH] +

Example 14.

Methyl-3-hydroxy-4-(hydroxymethyl)-5-{[(2S)-2-(methoxycarbon yl)pyrrolidin-1 - yl]methyl}pyridine-2-carboxylate

The product was made in a procedure similar to Example 6, using the D- Proline methyl ester. 1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 7.12 (d 2H), 5.40 (d 2H), 5.01 1 (d 1 H), 4.56 (s, 2H), 4.00(s, 3H)1 6(s 6H); MS-ESI m/z 414 [MH] +

N-[(3-chloro-4-fluorophenyl)methyl]-3-hydroxy-4-(hydroxymeth yl)-5-{[(4- methoxyphenyl)methoxy]methyl}pyridine-2-carboxamide

It was synthesized in a procedure similar to that of Scheme 3, Example 2, using 3-chloro-4 fluorobenzyl amine. MS-ESI m/z 461 .8 [MH] + Example 16.

N- Cyclopropyl-5-((4-fluorophenoxy)methyl)-3-hydroxy-4-(hydroxy methyl)- picolinamide

Compound 28b of US 8,742,123 Methyl 5-((4-fluorophenoxy)methyl)-3- hydroxy-4-(hydroxymethyl)picolinate was used, dissolved in neat cyclopropylamine, and refluxed for 12 h at 59 °C. Evaporation of the amine and flash chromatography on silica gel (EtoAc eluent) gave the desired product. 1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 6.88 (d 2H), 5.40 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H) 0.50-0.77 (dd 3H); MS-ESI m/z 333.1 [MH] +

Example 17.

tert-butyl 2-({5-[(4-fluorophenoxy)methyl]-3-hydroxy-4-(hydroxymethyl)p yridin- 2 -y l}f o rma m i d o )a cetate

Compound 28b of US 8,742,123 Methyl 5-((4-fluorophenoxy)methyl)-3- hydroxy-4-(hydroxymethyl)picolinate was used, dissolved in DMF, trimethylamine and O-tert-butyl Glycine hydrochloride was added, refluxed for 12 h at 80 °C.

Workup using 10% citric acid and 10% NaCCb extractions vs EtOAc and flash chromatography on silica gel (EtoAc eluent) gave the desired product. MS-ESI m/z 407.1 [MH] +

Example 18.

N-(n-Propyl)-5-((4-fluorobenzyloxy)methyl)-3-hydroxy-4-(h ydroxymethyl)- picolinamide

It was synthesized in a procedure similar to that of Scheme 3, Example 2, using 4-fluorobelzyl bromide in the second step and ending with propyl amine refluxed for 12 h at 59 °C. Evaporation of the amine and flash chromatography on silica gel (EtoAc eluent) gave the desired product. 1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 6.88 (d 2H), 5.40 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H) 0.50-0.77 (dd 3H); MS-ESI m/z 349.1 [MH] +

Example 19.

5-[(4-fluorophenoxy)methyl]-3-hydroxy-4-(hydroxymethyl)-N -(2-oxoazepan-3- yl)pyridine-2 -carboxamide

Compound 28b of US 8,742,123 Methyl 5-((4-fluorophenoxy)methyl)-3- hydroxy-4-(hydroxymethyl)picolinate was used, dissolved in DMF and added trimethylamine, L-aminocaprolactam, refluxed for 12 h at 80 °C. Workup using 10% citric acid and 10% NaHCOs extractions vs EtOAc and flash chromatography on silica gel (EtoAc eluent) gave the desired product. MS-ESI m/z 407.1 [MH] +

Example 20.

N-(2’-methoxyethyl)-5-((4-fluorophenoxy)methyl)-3-hydro xy-4-

(hydroxymethyl)picolinamide

It was synthesized in a procedure similar to that of Scheme 1 , Example 2, using methoxy ethyl amine, and refluxed for 12 h at 59 °C. Evaporation of the amine and flash chromatography on silica gel (EtoAc eluent) gave the desired product.

1 H-NMR (500MHz, MeOD) : d = 8.01 (s, 1 H), 7.34 (d 2H), 6.88 (d 2H), 5.40 (d 2H), 4.56 (s, 2H), 2.7-2.9 (m 1 H) 0.50-0.77 (dd 3H); MS-ESI m/z 350.1 [MH] + Example 21.

N-cyclopropyl 5-((4-fluoro-3-chlorophenoxy)methyl)-3-hydroxy-4- (hydroxymethyl)-Picolinamide The compound was synthesized using the general procedure outlined in

Scheme 1 , then reacted with cyclopropylamine and deprotected. MS-ESI m/z 367.4 [MH] +

Example 22.

5-[(3-chloro-4-fluorophenoxy)methyl]-3-hydroxy-4-(hydroxy methyl)-N-(2- methoxyethyl)pyridine-2-carboxamide

The compound was synthesized using the general procedure outlined in Scheme 1 . MS-ESI m/z 385.2 [MH] +

Example 23.

N-propyl-5-((4-fluoro-3-chlorophenoxy)methyl)-3-hydroxy-4 -(hydroxymethyl)

Picolinamide

The compound was synthesized using the general procedure outlined in

Scheme 1.

MS-ESI m/z 369.3 [MH]

Example 24.

N-(2’-methoxyethyl)-5-((4-fluorophenoxy)methyl)-3-hydro xy-4-(methyl)- picolinamide

The compound was synthesized using the general procedure outlined in Scheme 3.

MS-ESI m/z 318.1 [MH] +

Example 25.

Methyl-5-((5-methoxyindole)methyl)-2,2-dimethyl-4H-[1,3]d ioxino[4,5- c]pyridine-8-carboxylate The compound was synthesized using the general procedure outlined in

Scheme 1 , Example 1.

MS-ESI m/z 383.5 [MH] +

Example 26.

Methyl-5-((5-iodoindole)methyl)-2, 2-dimethyl -4H-[1,3]-dioxino[4,5-c]pyridine-8- carboxylate

The compound was synthesized using the general procedure outlined in Scheme 1 , Example 1.

MS-ESI m/z 479.5 [MH] +

Example 27.

Methyl 5-[(6-chloro-5-fluoroindol-1 -yl)methyl]-2, 2-dimethyl -4H-[1 ,3]dioxino[4,5- c]pyridine-8-carboxylate

The compound was synthesized using the general procedure outlined in Scheme 1 , Example 1 using 6-chloro-5fluoroindole. MS-ESI m/z 405.3 [MH] +

Example 28.

5-[(5-fluoro-1 H-indol-1 -yl)methyl]-N,2,2-trimethyl-2H,4H-[1 ,3]dioxino[4,5- c]pyridine-8-carboxamide

50 mg of compound 1 was dissolved into a 33% solution of methyl amine in ethanol. The solution was stirred for 18 h and evaporated off. A 1 % solution of citric acid (1 ml_) was added and the paste triturated to yield an off-white solid. MS-ESI m/z 370.3 [MH] +

Example 29.

5-[(5-methoxyindol-1-yl)methyl]-N,2,2-trimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-

8-carboxamide

50 mg of compound 25 was dissolved into a 33% solution of methyl amine in ethanol. The solution was stirred for 18 h and evaporated off. A 1 % solution of citric acid (1 ml_) was added and the paste triturated to yield an off-white solid. Yields 43mg (90%). LCMS MS-ESI m/z 382.3 [MH] +

Example 30.

5-[(5-iodoindol-1 -yl)methyl]-N,2,2-trimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8- carboxamide

50 mg of compound 26 was dissolved into a 33% solution of methyl amine in ethanol. The solution was stirred for 18 h and evaporated off. A 1 % solution of citric acid (1 ml_) was added and the paste triturated to yield an off-white solid. Yields 45 mg (90%). LCMS MS-ESI m/z 478.2 [MH] + Example 31.

5-((6-chloro-5-fluoroindol-1-yl)methyl)-N,2,2-trimethyl-4 H-[1 ,3]dioxino[4,5- c]pyridine-8-carboxamide

50 mg of compound 27 was dissolved into a 33% solution of methyl amine in ethanol. The solution was stirred for 18 h and evaporated off. A 1 % solution of citric acid (1 ml_) was added and the paste triturated to yield an off-white solid. Yields 45 mg (90%). LCMS MS-ESI m/z 404.3 [MH] + Example 32.

N-cyclopropyl-5-[(5-methoxyindol-1 -yl)methyl]-2, 2-dimethyl -4H-[1 ,3]dioxino[4, 5- c]pyridine-8-carboxamide

The compound from Example 25 (65 mg) was stirred in a sealed vial containing 500 uL of cyclopropylamine at 45 °C for 48 h until completion.

Evaporation and flash chromatography yielded 27 mg product. LCMS MS-ESI m/z 408.7[MH] + Example 33.

5-(3-chloro-4-fluorophenoxymethyl)-N,2,2-trimethyl-4H-[1 ,3]dioxino[4,5- c]py rid i ne-8-ca rboxa mi de

The compound was synthesized using the general procedure outlined in

Scheme 1 , Example 1 using 3-chloro-4-fluorophenol in the first step. The resulting compound was stirred in a 33% solution of methylamine in ethanol for 4 h. MS-ESI m/z 381.2 [MH] + Example 34.

N-Methyl-5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]- pyridine-8-carboxamide

The compound was synthesized using the general procedure outlined in Scheme 1 , Example 1 and reacted with ethanolic methylamine. MS-ESI m/z 347.5[MH] +

Example 35.

N-Methyl-5-((4-fluorophenoxy)methyl)-3-hydroxy-4-(hydroxy methyl)- picolinamide

The compound was synthesized using the general procedure outlined in

Scheme 1 , Example 1 and reacted with ethanolic methylamine. MS-ESI m/z 307.4 [MH] +

Example 36.

N-Methyl-5-((4-fluoro-3-chlorophenoxy)methyl)-3-hydroxy-4 - hydroxymethyl)picolinamide

The compound was synthesized using the general procedure outlined in Scheme 1 , Example 1 and reacted with ethanolic methylamine. MS-ESI m/z 341 3[MH] +

Example 37.

Methyl-5-((4-trifluoromethylphenylamino)methyl)-2,2-dimethyl -4H-

[1 ,3]dioxino[4,5-c]pyridine-8-carboxylate

Methyl 5-formyl-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]picolinate, (J Org Chem,

1999, 64:4537) was added to acetic acid in methanol. 4-trifluoroaniline was added and the mixture stirred for 30 minutes before sodium cyanoborohydride was added. The mixture was stirred for 12 h and the solvent evacuated. EtoAc was added, followed by extraction with 10% AcOH, followed by 10% Na2CC>3 and brine. The EtoAc was evacuated and the dark residue flashed chromatographed on silica gel (EtoAc eluent). The solvent was evacuated and the residue crystallized.

1 H-NMR (500MHz, MeOD) : d = 8.19 (s, 1 H), 6.6 (d 1 H), 6.3 (d 1 H), 6.1 1 (m 1 H)5.80 (d 2H), 5.53 (d 2H), 1 6(s 6H); MS-ESI m/z 397.2 [MH] + Example 38.

N-Cyclopropyl-5-((4-trifluoromethylphenylamino)methyl)-2, 2-dimethyl-4H-

[1 ,3]dioxino[4,5-c]pyridine-8-carboxamide

The compound from Example 37 (65 mg) was stirred in a sealed vial containing 500 uL of cyclopropylamine at 45 °C for 48 h until completion.

Evaporation and flash chromatography yielded 27 mg product. Example 39.

Methyl 5-((4-trifluoromethylphenoxy)methyl)-2,2-dimethyl-4H-[1,3]di oxino[4,5- c]pyridine-8-carboxylate

Methyl 5-(hydroxymethyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8- carboxylate (Paul et al., J Med Chem, 1977, 20:745) was dissolved in ethyl acetate and cooled to 0 °C. Triethyl amine (TEA) was added and methanesulphonyl chloride added dropwise over 20 minutes. TLC revealed a complete conversion of the starting material. Meanwhile, 4-trifluorphenol was dissolved in tetrahydrofuran THF and potassium tert-butoxide (KOtB) was added. The mesylate solution was evaporated without heating and the solvent replaced with DMF. The methyl 5- (methansulphoxymethyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8-carboxylate DMF solution was dripped slowly over 20 minutes into the phenoxide solution and left to warm to RT. A gel formed and the solution was then heated with a heat gun to liquefy. After 2 h, TLC revealed completion of the reaction. The DMF was partially removed and EtAc was added. This solution was then extracted with 5% citric acid followed by 10% Na2CC>3 (2X) and finally a 50% NaCI (brine) solution. The organic phase was then evaporated to yield the product. LCMS MS-ESI m/z 398.3 [MH] + Example 40.

5-[(6-chloro-5-fluoroindol-1 -yl)methyl]-N-cyclopropyl-2,2-dimethyl-4H-

[1,3]dioxino[4,5- c]pyridine-8-carboxamide The compound from Example 27 (65 mg) was stirred in a sealed vial containing 500 uL of cyclopropylamine at 45 °C for 48 h until completion.

Evaporation and flash chromatography yielded 27 mg product. LCMS MS-ESI m/z 430.3 [MH] +

Example 41.

N-Methyl-5-((4-trifluoromethylphenylamino)methyl)-2,2-dimeth yl-4H-

[1 ,3]dioxino[4,5-c]pyridine-8-carboxamide

The compound was synthesized from the compound from Example 37 using the general procedure outlined in Scheme 1 , Example 1 , and reacted with ethanolic methylamine. MS-ESI m/z 396.4 [MH] +

Example 42.

5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H-[1,3]dioxino[4,5 -c]-pyridine-8- carbonitrile

5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8- carbaldehyde was prepared in a fashion similar to Example 17d of US 8,742,123, using 5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H-[1 ,3]dioxino[4,5-c]pyridine-8- hydroxymethyl in lieu of Example 17c of US 8,742,123. The resulting aldehyde was then dissolved in ethanol and 50% NH2OH aq., was added with stirring. A brief heating with a heat gun gave a clear solution which upon cooling in an ice water bath precipitated white crystalline 5-((4-fluorophenoxy)methyl)-2,2-dimethyl-4H- [1 ,3]dioxino[4,5-c]pyridine-8-carboxime. The compound was dried and taken up in THF. Carbonyldiimidazole was then added and the mixture stirred at RT. After 1 h, the temperature was raised to reflux. The THF was evacuated and EtoAc added. Extraction with citric acid, Na 2 C0 3 , brine and dried over Mg 2 S0 4 . The clear organic phase was then evacuated to yield the desired product. 1 H-NMR (500MHz, MeOD) : d = 7.52 (s, 1 H), 7.04 (d 2H), 6.88 (d 2H), 4.81 (s, 2H), 4.75(s, 2H), 1.50 (s6H); MS-ESI m/z 315.3 [MH] +

Example 43.

5-((4-fluorophenoxy)methyl)-3-hydroxy-4-(hydroxymethyl)-P yridine-2-(2- trifluoroethanol)

To the product of Example 2 was added trimethylsilyltrifluoromethane in THF. To the vigorously stirred mixture was then added caesium fluoride. The mixture bubbled vigorously and subsided within 1 minute. TLC showed complete conversion of the starting material. The residue after evaporation was then chromatographed on silica gel to yield a clean product.

1 H-NMR (500MHz, MeOD) : d = 7.92 (s, 1 H), 7.04 (d 2H), 6.88 (d 2H), 4.98 (m 2H), 4.81 (s, 2H), 4.75(s, 2H), 1.50 (s6H); MS-ESI m/z 348.2 [MH] +

Example 44.

N,N-Dimethyl-5-((4-fluorophenoxy)methyl)-3-hydroxy-4-(hyd roxymethyl)- picolinamide

The compound was synthesized using the general procedure outlined in Scheme 1 , Example 1 and reacted with a solution of dimethylamine hydrochloride and triethylamine in DMF for 3 days until complete conversion. MS-ESI m/z 320.7 [MH] +

Example 45. Cell culture and treatment

Human hepatoma cell line HepG2 was routinely cultivated in Dulbecco’s modified Eagle’s medium (DMEM; catalog no. 319-005-CL, Wisent) supplemented with 10% fetal bovine serum (FBS; catalog no. 080-350, Wisent). For phenotypic screening of LDLR, cells were incubated overnight in conditioned media containing small molecule inducers compounds 1-32 resuspended in DMSO at concentration ranging from 10 nM to 10 mM.

Example 46. Western blot analysis

Treated human hepatoma cells HepG2 were washed three times in

phosphate-buffered saline (PBS) and lysed in radioimmune precipitation assay buffer (50 mM Tris/HCI, pH 8.0, 1 % (v/v) Nonidet P-40, 0.5% sodium deoxycholate, 150 mM NaCI, and 0.1 % (v/v) SDS) supplemented with a complete protease inhibitor mixture (catalog no. 1 1 697 498 001 , Roche Applied Science). Proteins were separated by 8% SDS polyacrylamide gel electrophoresis, blotted on nitrocellulose membranes (Bio-Rad), and blocked for 1 h in Tris-buffered saline-Tween 20 (TBS-T; 50 mM Tris-HCI, pH 7.5, 150 mM NaCI, 0.1 % Tween 20 (Polysorbate 20)) containing 5% nonfat dry milk. Membranes were then incubated overnight in TBS-T

supplemented with 1 % nonfat milk and the indicated antibodies; goat anti-human LDLR (1 :1000; catalog no. AF2148, R&D Systems), rabbit anti-actin (1 :5000; catalog no. A2066, Sigma-Aldrich). Appropriate HRP-conjugated secondary antibodies (1 :10,000; GE healthcare) were used for detection using the Western Lightning Ultra chemiluminescence kit (catalog no. NEI1 12001 EA, PerkinElmer Life Sciences) and BioFlex EC Films (catalog no. CLEC810, InterScience). Unsaturated films were numerized using the high resolution CanoScan 9000F scanner and corresponding bands were quantified using the ImageJ software (NIH). Relative quantification of LDLR protein levels were normalized to that of actin and compared to vehicle

(DMSO) herein determined as 1. Results are described in Table 4.

Example 47. Cell viability assay

Cells were seeded into 96-well plates and incubated at confluency overnight with 1 , 5, 10, 50, 100, or 500 mM of given compounds in phenol red-free conditioned media (catalog no. 319-051 -CL, Wisent). Following incubation, media was replaced and cells incubated for 4 h with 100 pL of phenol red-free DMEM containing 0.5 mg/mL of MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide; thiazolyl blue; catalog no. M5655, Sigma-Aldrich), according to manufacturer's instructions. MTT is converted to dark blue, water-insoluble MTT formazan by mitochondrial dehydrogenases present in living cells. The blue crystals of MTT formazan were solubilized with 100 pL acidic isopropanol (0.06 N) and absorbance measured at a wavelength of 570 nm with background subtraction at 650 nm. Arbitrary units of cell viability of a given example were determined relative to equal volume of DMSO (vehicle) for each concentration used. Results are shown in Table 4.

TABLE 4. Relative LDLR Protein Levels Following Treatment with Exemplified

Compounds

Example 48. Microsomal stability assay

Pooled human liver microsomes (HLM; Cat# X008067 lot IHG) and pooled male Sprague Dawley rat liver microsomes (RLM; Cat# M00001 Lot NNK) were purchased from BioreclamationIVT, Baltimore, Maryland. NADPH (Cat # N1630, 95% HPLC) and DMSO (GC grade) were purchased from Sigma-Aldrich, Canada. Potassium phosphate buffer (100 mM) with magnesium chloride pH 7.4 was prepared freshly. Potassium phosphate buffer (100 mM, pH 7.4 + 2 mM MgCh) containing human or rat liver microsomes (0.50 mg/ml) was pre-incubated separately with compound 2 (1 mM) or positive control (loperamide, 1 pM) in a water bath with the temperature set at 37 °C for 5 min (N=2). Reactions were initiated by adding NADPH (final concentration 1 mM) in all the wells. Reactions without NADPH were also incubated (final point only) to rule out non-NADPH metabolism or chemical instability in the incubation buffer. Reactions were terminated at each time point (0, 5, 10, 20, 30, 45, and 60 min) by transferring 50 pL in a new 96-well plate and adding 100 pi of acetonitrile containing an internal standard (labetalol at 2 pM). The plates were centrifuged at 4000 rpm for 5 min, and an aliquot of supernatant was diluted with one volume of water + 0.1 % formic acid before analysis by LC-MS/MS. Reference samples were also prepared at concentrations between 0.002 pM and 2 pM in buffer containing microsomes, by adding the diluted solutions of the compound to the buffer containing microsomes quenched with 2 volume of acetonitrile containing the internal standard (whereas the incubation TO is obtained by quenching the microsomes containing the compound, then adding NADPH).

Samples were monitored for parent compound disappearance in Multi Reaction Monitoring (MRM) mode using LC-MS/MS. The peak area ratios of analyte versus internal standard compared to the calibration curve were used to calculate concentration at each time point. The elimination constant (ke, min-1 ) was obtained from the log-linear regression of the concentration (y) versus time (t). The half-life was calculated as T1/2 (min) = 0.693/ke. The microsomal intrinsic clearance (Clint, pL/min/mg of protein) was calculated by the following equation:

Clint (pL/min/mg of protein) = V x T °^ 93 = V x Ke

Volume of incubation (mί)

Where V (m L/mg) =

Protein in the incubation (mg)

Results are shown in Table 5 and illustrated in Figures 1A and 1 B.

TABLE 5. Microsomal Stability

Example 49. Animal studies

All animal studies were approved by the Montreal Heart Institute (MHI) Animal Care and Ethical committee. Pharmacokinetic experiments were performed at the Montreal Heart Institute in collaboration with the Platform of Biopharmacy at

Universite de Montreal. Briefly, compound 3 (formulation PG:H 2 0; 50:50 stock solution 2.5 mg/ml) was administered intravenously (iv) at 5 mg/kg (n=4) or by oral gavage (PO) at 25 mg/kg (n=4) in adult Sprague-Dawley male rats (~200g). Blood samples were collected at 5, 15, 30, 60, 120, 240, 360, 480, 1440 min for which 125 pL of precipitation solution (80% acetonitrile and 20% methanol containing IS

Loperamide at 0.1 pM) were added. Samples were mixed and centrifuged at 15,000 rpm for 5 min. 50 pL of supernatant were transferred to an HPLC plate, and 100 pL of water + 0.1 % formic acid were added. A 16-point calibration curve with Example 2 was prepared in blank rat blood, ranging from 0.002 to 15 mM. Samples were analyzed in MRM mode using LC-MS/MS. The calibration curve was plotted using the ratio of the analyte peak area and the IS peak area, using a quadratic regression. Pharmacokinetic (PK) parameters were calculated using Kinetica Software for PK/PD Data Analysis, Simulation and Reporting (Thermo Fisher), based on the blood concentrations of each animal. Results are described in Table 6 and illustrated in Figure 2.

TABLE 6. Pharmacokinetic Parameters

For in vivo animal studies, wild-type C57BL/6 male mice (~25 g; obtained from Charles River Laboratories) or Golden Syrian (-120 g obtained from Envigo++) were housed in the 12 hour light/12 hour dark cycle, temperature and humidity controlled MHI animal facility. Following 5 days of acclimatization, animals were fed a chow or Western diet containing 48.5% w/w carbohydrate, 21 .2% w/w fat, 17.3% w/w protein and 0.2% w/w cholesterol (catalog no. TD.88137, Envigo) for 14 days. Example 3 was mixed in either ethanol/propylene glycol/water 1 :7:8 and administered by oral gavage at 40 mg/kg/day for 10 days in mice maintained on Western diet. Equivalent volume of vehicle containing corresponding formulations were orally administered to mice daily and designated as placebo (vehicle). Plasma lipoprotein cholesterol profiles were obtained from 100 pl_ of pooled plasma injected on a Superose 6 10/300 GL (catalog no. 17-5172-01 , GE Life Sciences) and eluted with PBS at a flow rate of 0.1 mL per min at 4 °C mounted on a AKTA explorer system (GE Healthcare). Total cholesterol (catalog no. 439-17501 , Wako) was quantified in total plasma and in each 0.3 mL collected fractions or in-line (Lipidomics Core of The Group on Molecular and Cell Biology of Lipids, University of Alberta) according to the manufacturer’s instructions. Relative quantitation of AUC of total cholesterol corresponding to non-HDL lipoprotein positive fractions obtained from FPLC profiles are included in Table 4. Serum aspartate aminotransferase (AST) and alanine transaminase (ALT) were measured by the Montreal Heart Institute biochemical clinical chemistry platform according to manufacturer’s recommendations. Results are described in Table 7.

TABLE 7. Efficacy and Safety of Compound 3 in Hypercholesterolemic Animal Models

Other Embodiments

While the invention has been described in connection with specific

embodiments thereof, it will be understood that it 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 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.

All publications, patents, and patent applications are herein incorporated 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.