Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
BENZODIOXINONE COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2019/099457
Kind Code:
A1
Abstract:
Provided herein are benzodioxinone compounds of Formula I that are useful as inhibitors of ACC1 and/or ACC2. The benzodioxinone compounds described herein can be used for treating a disease or disorder associated with aberrant ACC1 and/or ACC2 activities, for example, non-alcoholic steatohepatitis (NASH), acne, obesity, diabetes, and cancer. Also provided herein are pharmaceutical compositions comprising the benzodioxinone compound of Formula I, or pharmaceutically acceptable salt thereof.

Inventors:
XIONG YUSHENG (US)
GUAN HONG-PING (US)
HUANG WENJUN (US)
ZHANG ZENGHONG (US)
Application Number:
PCT/US2018/060937
Publication Date:
May 23, 2019
Filing Date:
November 14, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
QUIXGEN INC (US)
XIONG YUSHENG (US)
GUAN HONG PING (US)
HUANG WENJUN (US)
ZHANG ZENGHONG (US)
International Classes:
C07D221/20; A61K31/4747; C07D319/08
Foreign References:
US7410976B22008-08-12
US20110288058A12011-11-24
US8110570B22012-02-07
US20090005375A12009-01-01
Other References:
BOURBEAU ET AL.: "Recent Advances in the Development of Acetyl-CoA Carboxylase (ACC) Inhibitors for the Treatment of Metabolic Disease", JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, 2015, pages 525 - 536, XP055311723, DOI: doi:10.1021/jm500695e
Attorney, Agent or Firm:
LIU, Xiaoxiang (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of Formula I, or a pharmaceutically acceptable salt thereof,

Formula I,

wherein:

R1 and R2 are each independently hydrogen, halogen, cyano, an optionally substituted Ci-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted C6-10 aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl,— NR10Rn,— COOR12,— CONR13R14,— CN,— S(0)nR15, or—OR16; wherein

R10 and R11 are each independently hydrogen, an optionally substituted Ci-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted Ci-6 alkanoyl, an optionally substituted C3-6 cycloalkanoyl, an optionally substituted C6-10 aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl,— COOR12, or— CONR13R14;

R12, R13 and R14 are each independently hydrogen or an optionally substituted Ci-6 alkyl; n is 0, 1, or 2;

R15 is an optionally substituted Ci-6 alkyl, an optionally substituted C3-6 cycloalkyl, or — NR10Rn;

R16 is hydrogen, an optionally substituted Ci-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted Ci-6 alkanoyl, an optionally substituted C3-6 cycloalkanoyl, an optionally substituted C6-10 aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl or — CONR13R14; and

R3 is an optionally substituted C6-ioaryl or optionally substituted 5-10 membered heteroaryl.

2. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein R1 is halogen, an optionally substituted Ci-6 alkyl, an optionally substituted Ci-6 alkoxy, an optionally substituted C3-6 cycloalkyl, or an optionally substituted C3-6 cycloalkoxy.

3. The compound of claim 1 or 2, or pharmaceutically acceptable salt thereof, wherein R1 is C14 alkyl, C14 alkoxy, C3-6 cycloalkyl, or C3-6 cycloalkoxy, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and C1-4 alkyl.

4. The compound of any one of claims 1-3, or pharmaceutically acceptable salt thereof, wherein R1 is C14 alkyl or C1-4 alkoxy, each optionally substituted with 1-3 fluorines.

5. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein R1 is a 5- 10 membered heteroaryl optionally substituted with 1 or 2 substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C14 alkyl, optionally substituted C14 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy.

6. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein R1 is a 4- 6 membered heterocyclyl optionally substituted with 1 or 2 substituents independently chosen from halogen, oxo, optionally substituted C14 alkyl, optionally substituted C14 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy.

7. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein R1 is — NR10Rn.

8. The compound of claim 7, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is an optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl.

9. The compound of claim 7, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is a 5-membered heteroaryl optionally substituted with 1 or 2 substituents independently chosen from halogen, hydroxyl, cyano and C1-4 alkyl.

10. The compound of claim 7, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is a 5-membered heteroaryl having 2-4 ring nitrogen atoms, which is optionally substituted with 1 or 2 substituents independently chosen from halogen, hydroxyl, cyano and CM alkyl.

11. The compound of claim 7, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is a pyrazole, triazole or tetrazole, each optionally substituted with 1 or 2 independently chosen Ci-4 alkyl.

12. The compound of any one of claims 1-11, or pharmaceutically acceptable salt thereof, wherein R2 is H.

13. The compound of any one of claims 1-12, or pharmaceutically acceptable salt thereof, wherein R3 is a C6-ioaryl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, optionally substituted CM alkyl, optionally substituted C alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy.

14. The compound of any one of claims 1-12, or pharmaceutically acceptable salt thereof, wherein R3 is an optionally substituted 8-10 membered bicyclic heteroaryl.

15. The compound of any one of claims 1-12, or pharmaceutically acceptable salt thereof, wherein R3 is a naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1- 3) substituents independently chosen from halogen, cyano, hydroxyl, CM alkyl optionally substituted with 1-3 halogens, CM alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens.

16. The compound of any one of claims 1-12, or pharmaceutically acceptable salt thereof, wherein R3 is a naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1- 3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, ethyl, trifluoromethyl, trifluoromethoxy, methoxy, ethoxy, and cyclopropyl.

17. A compound of Formula la, or a pharmaceutically acceptable salt thereof,

Formula la,

wherein:

R1 is hydrogen, halogen, cyano, an optionally substituted C1-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted C6-10 aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl,— NR10Rn,— COOR12, — CONR13R14,— CN,— S(0)nR15, or—OR16;

wherein

R10 and R11 are each independently hydrogen, an optionally substituted C1-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C1-6 alkanoyl, an optionally substituted C3-6 cycloalkanoyl, an optionally substituted Ob-io aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl,— COOR12, or— CONR13R14;

R12, R13 and R14 are each independently hydrogen or an optionally substituted C1-6 alkyl; n is 0, 1, or 2;

R15 is an optionally substituted C1-6 alkyl, an optionally substituted C3-6 cycloalkyl, or — NR10Rn; R16 is hydrogen, an optionally substituted Ci-6 alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C 1-6 alkanoyl, an optionally substituted C3-6 cycloalkanoyl, an optionally substituted C6-10 aryl, an optionally substituted 5-10 membered heteroaryl, an optionally substituted 4-6 membered heterocyclyl or — CONR13R14; and

R3 is an optionally substituted C6-ioaryl or optionally substituted 5 to 10 membered heteroaryl.

18. The compound of claim 17, or pharmaceutically acceptable salt thereof, wherein R1 is an optionally substituted C 14 alkyl.

19. The compound of claim 17 or 18, or pharmaceutically acceptable salt thereof, wherein R1 is methyl, ethyl, isopropyl, or tert-butyl.

20 The compound of claim 17, or pharmaceutically acceptable salt thereof, wherein R1 is an optionally substituted C alkoxy.

21 The compound of claim 20, or pharmaceutically acceptable salt thereof, wherein R1 is methoxy, trifluoromethoxy, ethoxy or isopropoxy.

22 The compound of claim 17 having Formula lb, or pharmaceutically acceptable salt

thereof,

Formula lb,

wherein one of R10 and R11 is an optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl.

23. The compound of claim 22, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is an optionally substituted 5 or 6 membered heteroaryl.

24. The compound of claim 22, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is a 5-membered heteroaryl having 2-4 ring nitrogen atoms, which is optionally substituted with 1 or 2 substituent chosen from halogen, hydroxyl, cyano and CM alkyl.

25. The compound of claim 22, or pharmaceutically acceptable salt thereof, wherein one of R10 and R11 is a pyrazole, triazole or tetrazole, each optionally substituted with a CM alkyl.

26. The compound of claim 22, or pharmaceutically acceptable salt thereof, wherein one of , and the other of R10 and R11 is hydrogen or methyl.

27. The compound of any one of claims 17-26, or pharmaceutically acceptable salt thereof, wherein R3 is a C6-ioaryl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C alkyl, optionally substituted CM alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy.

28. The compound of any one of claims 17-26, or pharmaceutically acceptable salt thereof, wherein R3 is an optionally substituted 8-10 membered bicyclic heteroaryl.

29. The compound of any one of claims 17-26, or pharmaceutically acceptable salt thereof, wherein R3 is a naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1- 3) substituents independently chosen from halogen, cyano, hydroxyl, C alkyl optionally substituted with 1-3 halogens, CM alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens.

30. The compound of any one of claims 17-26, or pharmaceutically acceptable salt thereof, wherein R3 is

each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, trifluoromethyl, ethyl, methoxy, trifluoromethoxy, ethoxy, and cyclopropyl.

31. The compound of any one of claims 17-26, or pharmaceutically acceptable salt thereof, wherein R3 is

32. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein:

R1 is hydrogen.

33. The compound of claim 32, or pharmaceutically acceptable salt thereof, wherein R3 is a C6-ioaryl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C1-4 alkyl, optionally substituted CM alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy.

34. The compound of claim 32, or pharmaceutically acceptable salt thereof, wherein R3 is an optionally substituted 8-10 membered bicyclic heteroaryl.

35. The compound of claim 32, or pharmaceutically acceptable salt thereof, wherein R3 is a naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, cyano, hydroxyl, C alkyl optionally substituted with 1-3 halogens, CM alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens.

36. The compound of claim 32, or pharmaceutically acceptable salt thereof, wherein R3 is

each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, trifluoromethyl, ethyl, methoxy, trifluoromethoxy, ethoxy, and cyclopropyl.

37. The compound of claim 32, or pharmaceutically acceptable salt thereof, wherein R3 is an optionally substituted benzo[b]thienyl.

38. The compound of claim 37, or pharmaceutically acceptable salt thereof, wherein R3 is

optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, -NR30R31, cyano, hydroxyl, CM alkyl optionally substituted with 1-3 halogens, CM alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens, wherein R30 and R31 are each independently hydrogen, an optionally substituted alkyl (e.g., an optionally substituted C1-6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), an optionally substituted alkanoyl (e.g., an optionally substituted C1-6 alkanoyl), an optionally substituted cycloalkanoyl (e.g., an optionally substituted C3-6 cycloalkanoyl), an optionally substituted aryl (e.g., an optionally substituted C6-10 aryl), an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl), an optionally substituted heterocyclyl (e.g., an optionally substituted 4-6 membered heterocyclyl), -COOR32, or -CONR33R34;

wherein R32, R33 and R34 are each independently hydrogen or an optionally substituted alkyl (e.g., an optionally substituted Ci-6 alkyl).

39. The compound of claim 38, or pharmaceutically acceptable salt thereof, wherein - NR30R31 is selected from NH2, NHCOOR32 or NHCONR33R34, wherein R32 is a Ci-6 alkyl, R33 and R34 are each independently hydrogen or a Ci-6 alkyl optionally substituted with 1- 3 fluorines.

40. The compound of any one of claims 17-26 and 32-39, or pharmaceutically acceptable salt thereof, wherein R3 is

41 The compound of any one of claims 17-40, or pharmaceutically acceptable salt thereof, wherein the compound is chosen from: Ġ 80

42. A pharmaceutical composition comprising a compound of any one of claims 1-41, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

43. The pharmaceutical composition of claim 42, formulated for intravenous injection or infusion, oral administration, inhalation, or topical administration.

44. The pharmaceutical composition of claim 42 or 43, formulated in the form of a topical solution, lotion, shampoo, transdermal spray, topical film, foam, powder, paste, sponge, transdermal patch, tincture, tape, cream, gel, or ointment.

45. The pharmaceutical composition of any one of claims 42-44, comprising an amount of the compound of any one of claims 1-41, or pharmaceutically acceptable salt thereof, effective for inhibiting one or more activities in a human in need thereof, wherein the one or more activities are chosen from acetyl-CoA carboxylases ACC1 and/or ACC2 activities, lipogenesis, proliferation of cells (e.g., adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, or skin stem cells) in epidermis, dermis, and/or hypodermis, proliferation of human sebocytes, proliferation of human

keratinocytes, differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers, sebum production, inflammation, and combinations thereof.

46. A method of inhibiting one or more activities in a subject in need thereof, wherein the one or more activities are chosen from acetyl-CoA carboxylases (ACC1 and/or ACC2) activities, malonyl-CoA production, lipogenesis, proliferation of sebocytes, proliferation of keratinocytes, proliferation of cells (e.g., adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, and skin stem cells) in epidermis, dermis, and/or hypodermis, differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers, inflammation, and combinations thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-41, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 42-45.

47. The method of claim 46, wherein the compound of any one of claims 1-41, or pharmaceutically acceptable salt thereof, or pharmaceutical composition of any one of claims 42-45 is administered to the subject topically.

48. A method of treating a disease or disorder associated with aberrant sebocyte and/or

keratinocyte activity in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-41, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 42-45.

49. The method of claim 48, wherein the disease or disorder is acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, actinic keratosis, sebaceous adenoma, sebaceous cyst, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea, fibrofolliculomas in Birt-Hogg-Dube syndrome, or a combination thereof.

50. The method of claim 48 or 49, wherein the disease or disorder is acne.

51. The method of any one of claims 48-50, wherein the compound of any one of claims 1- 41, or pharmaceutically acceptable salt thereof, or pharmaceutical composition of any one of claims 42-45 is administered to the subject topically.

52. The method of any one of claims 48-51, further comprising treating the subject with one or more additional therapies effective for the treatment of the disease or disorder associated with aberrant sebocyte and/or keratinocyte activity.

53. The method of any one of claims 48-51, further comprising administering to the subject an antibiotic (e.g., clindamycin, erythromycin, metronidazole, sulfacetamide, or tetracyclines such as doxycycline and minocycline) or retinoid (e.g., adapalene, isotretinoin, retinol, tazarotene, or tretinoin) in a therapeutically effective amount for treating acne and/or inflammation.

54. A method for treating non-alcoholic fatty liver disease (e.g., non-alcoholic steatohepatitis (NASH)) in a subject in need thereof, the method comprises administering to the subject a therapeutically effective amount of a compound of any one of claims 1-41, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 42-45.

55. The method of claim 54, further comprising administering to the subject one or more additional agents in a therapeutically effective amount for treating the non-alcoholic fatty liver disease.

56. The method of claim 54 or 55, wherein the one or more additional agents are chosen from angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacyl glycerol O-acyltransferase 1 or 2 (DGAT1 or DGAT2) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, famesoid X receptor (FXR) agonists, galectin-3 inhibitors, glucagon- like peptide 1 (GLP1) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 1 ^-hydroxy steroid dehydrogenase (11 b- HSD1) inhibitors, IL-Ib antagonists, IL-6 antagonists, IL-10 agonists, IL-17 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PLC) inhibitors, PPAR-alpha agonists, PPAR-gamma agonists, PPAR-delta agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturase-l inhibitors, thyroid hormone receptor b agonists, tumor necrosis factor alpha (TNF-alpha) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, and combinations thereof.

57. The method of any one of claims 54-56, wherein the one or more additional agents are chosen from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX- 1002, cenicriviroc, cobiprostone, colesevelam, emricasan, enalapril, GFT-505, GR-MD- 02, hydrochlorothiazide, icosapent ethyl ester (ethyl eicosapentaenoic acid), IMM-124E, KD-025, linagliptin, liraglutide, mercaptamine, MGL-3196, obeticholic acid, olesoxime, peg-ilodecakin, pioglitazone, PX-102, remogliflozin etabonate, SHP-626, solithromycin, tipelukast, TRX-318, ursodeoxycholic acid, VBY-376, and combinations thereof.

58. A method for treating obesity and/or diabetes in a subject in need thereof, the method comprises administering to the subject a therapeutically effective amount of a compound of any one of claims 1-41, or a pharmaceutically acceptable salt thereof, or a

pharmaceutical composition of any one of claims 42-45.

59. The method of claim 58, further comprising administering the subject one or more

additional agents in a therapeutically effective amount for treating obesity and/or diabetes.

60. The method of claim 58 or 59, wherein the one or more additional agents are chosen from angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacyl glycerol O-acyltransferase 1 or 2 (DGAT1 or DGAT2) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, famesoid X receptor (FXR) agonists, galectin-3 inhibitors, glucagon- like peptide 1 (GLP1) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 1 ^-hydroxy steroid dehydrogenase (11 b- HSD1) inhibitors, IL-Ib antagonists, IL-6 antagonists, IL-10 agonists, IL-17 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PLC) inhibitors, PPAR-alpha agonists, PPAR-gamma agonists, PPAR-delta agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturase-l inhibitors, thyroid hormone receptor b agonists, tumor necrosis factor alpha (TNF-alpha) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, and combinations thereof.

61. The method of any one of claims 58-60, wherein the one or more additional agents are chosen from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX- 1002, cenicriviroc, cobiprostone, colesevelam, emricasan, enalapril, GFT-505, GR-MD- 02, hydrochlorothiazide, icosapent ethyl ester (ethyl eicosapentaenoic acid), IMM-124E, KD-025, linagliptin, liraglutide, mercaptamine, MGL-3196, obeticholic acid, olesoxime, peg-ilodecakin, pioglitazone, PX-102, remogliflozin etabonate, SHP-626, solithromycin, tipelukast, TRX-318, ursodeoxycholic acid, VBY-376, and combinations thereof.

62. A method of delivering salicyclic acid or a derivative thereof in a subject in need thereof, the method comprising administering to the subject the compound of any one of claims 1- 41, or a pharmaceutically acceptable salt thereof.

63. The method of claim 62, comprising administering to the subject the compound of any one of claims 17-41, wherein R1 is hydrogen, or a pharmaceutically acceptable salt thereof.

64. The method of claim 63, wherein the administering provides an anti-inflammatory

effective amount of salicyclic acid.

65. The method of claim 64, wherein the administering provides an extended release of

salicyclic acid.

66. The method of any one of claims 62-65, wherein the administering is topical

administration.

67. A method of treating inflammation in a subj ect in need thereof, the method comprising administering to the subject the compound of any one of claims 1-41, or a

pharmaceutically acceptable salt thereof.

68. The method of claim 67, comprising administering to the subject the compound of any one of claims 17-41, wherein R1 is hydrogen, or a pharmaceutically acceptable salt thereof.

69. The method of claim 68, wherein the administering provides an anti-inflammatory

effective amount of salicyclic acid.

70. The method of claim 69, wherein the administering provides an extended release of salicyclic acid.

71. The method of any one of claims 67-70, wherein the administering provides the

compound in an effective amount to inhibit ACC1 and/or ACC2 activities in the subject.

72. The method of any one of claims 67-71, wherein the administering is topical

administration.

Description:
BENZODIOXINONE COMPOUNDS

CROSS REFERENCE TO RELATED APPLICATIONS

[1] This application claims benefit of U.S. Provisional Application No. 62/585,880, filed on November 14, 2017, the content of which is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

Field of the Invention

[2] In various embodiments, the present invention is generally related to a novel class of benzodioxinone compounds, salts thereof, pharmaceutical compositions comprising the same, synthetic methods therefor, and uses thereof.

Background Art

[3] Fatty acids are essential components for normal function of living cells including

membrane biosynthesis, the production of signaling lipids, post-translational modifications, energy production and storage, etc. Acetyl-CoA carboxylase (ACC) is a cytoplasmic enzyme catalyzing the conversion of acetyl-CoA to malonyl-CoA, the rate-limiting step for fatty acid metabolism in cells. Cytoplasmic malonyl-CoA controls two downstream pathways of lipid metabolism, de novo fatty acid synthesis and fatty acid b-oxidation. Malonyl-CoA is the building block for long chain fatty acids synthesis and elongation. Malonyl-CoA can also allosterically inhibit the fatty acid transporter on mitochondrial membrane, carnitine palmitoyltransferase- 1, and block the entry of fatty acyl-CoA into mitochondria. By inhibiting ACC, cellular long chain fatty acid synthesis can be blocked and b-oxidation can be stimulated in the mitochondria. As a result, cellular lipid contents are reduced.

[4] ACC has two isoforms, ACC1 and ACC2, localized in cytosol and mitochondrial

membrane respectively. ACC protein is composed of three domains, in which biotin carboxylase domain catalyzes the carboxylation of biotin that is covalently linked to the biotin carboxyl carrier protein domain, and the carboxyltransferase domain catalyzes the transfer of the carboxyl group from carboxyl biotin to acetyl-CoA (Zhang et al, Science 299:2064-2067 (2003)). Molecular masses of ACC1 and ACC2 are about 265 and about 280 kDa respectively. Although both isoforms are found in different tissues, ACC1 is mainly expressed in lipogenic tissues such as liver, adipose, and lactating mammary gland, and ACC2 is predominantly expressed in muscle tissues and heart. In rodents, ACC1 is mainly responsible for de novo fatty acid synthesis and ACC2 for fatty acid beta-oxidation (Abu- Elheiga et al, The Journal of biological chemistry 272: 10669-10677 (1997); Abu-Elheiga et al, Proceedings of the National Academy of Sciences of the United States of America 92:4011-4015(1995)). In humans, ACC2 isoforms are also abundantly expressed in adipose tissue and liver and capable of de novo lipogenesis in these tissues (Castle et al, PloS one 4: e4369(2009)). Genetic studies show that ACC1 is essential for development and ACC1 deficiency causes embryonic lethality (Abu-Elheiga et al., Proceedings of the National Academy of Sciences of the United States of America 97: 1444-1449(2000)). In contrast, ACC2 mutant mice live and breed normally although they are leaner due to increased fatty acid oxidation, and resistant to high-fat high-carbohydrate diets in terms of body weight gain and insulin resistance (Abu-Elheiga et al, Science 297:2613-2616(2001); Abu-Elheiga et al, Proceedings of the National Academy of Sciences of the United States of America

102 12011-12016(2005); Abu-Elheiga et al, Proceedings of the National Academy of Sciences of the United States of America 799: 10207-10212(2003)).

[5] Various diseases or disorders are associated with aberrant ACC1 and/or ACC2 activities.

Accordingly, there is a need for novel compounds that can modulate ACC1 and/or ACC2 activities.

BRIEF SUMMARY OF THE INVENTION

[6] In various embodiments, the present disclosure provides a benzodioxinone compound that can be an inhibitor of ACC1 and/or ACC2, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure also provides pharmaceutical compositions comprising the benzodioxinone compound or pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure also provides methods of using the

benzodioxinone compound or pharmaceutically acceptable salt thereof for treating a disease or disorder associated with aberrant ACC1 and/or ACC2 activities, for example, non alcoholic steatohepatitis (NASH), acne, obesity, diabetes, and cancer (e.g., non-small cell lung cancer). [7] Certain embodiments of the present disclosure are directed to a compound of Formula I or a pharmaceutically acceptable salt thereof:

Formula I,

wherein R 1 , R 2 and R 3 are defined herein. In some embodiments, the compound of Formula I is a compound of Formula 1-1, Formula la, Formula lb, Formula II-1 to II-4, Formula III-1 to III- 10 or Formula IV- 1 to Formula IV- 10 as defined herein. In any of the embodiments described herein, the benzodioxinone compound can be r-(4,8-dimethoxy quinoline-2 - carbonyl)-6-isopropyl-4H-spiro[benzo[d][l,3]dioxine-2,4'-pip eridin]-4-one, l'-(4,8- dimethoxy-2-naphthoyl)-6-isopropyl-4H-spiro[benzo[d][l,3]dio xine-2,4'-piperidin]-4-one, 6- isopropyl-r-(2-methyl-lH-benzo[d]imidazole-6-carbonyl)-4H-sp iro[benzo[d][l,3]dioxine- 2,4'-piperidin]-4-one, l'-(lH-indazole-5-carbonyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one, F-(lH-indazole-6-carbonyl)-6-isopropyl- 4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one, 6-isopropyl-l'-(lH-pyrrolo[3,2- b]pyridine-2-carbonyl)-4H-spiro[benzo[d][l,3]dioxine-2,4'-pi peridin]-4-one, 6-isopropyl- G- (6-methoxyquinoline-3-carbonyl)-4H-spiro[benzo[d][l,3]dioxin e-2,4'-piperidin]-4-one, l'-(2- ethyl-lH-benzo[d]imidazole-6-carbonyl)-6-isopropyl-4H-spiro[ benzo[d][l,3]dioxine-2,4'- piperidin]-4-one, 6-isopropyl-l'-(3-methyl-2-oxo-2,3-dihydro-lH-benzo[d]imidaz ole-5- carbonyl)-4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-on e, l'-(4,8-dimethoxy-2- naphthoyl)-6-((l-methyl-lH-pyrazol-5-yl)amino)-4H-spiro[benz o[d][l,3]dioxine-2,4'- piperidin]-4-one, r-(4,8-dimethoxyquinoline-2-carbonyl)-4H-spiro[benzo[d][l,3] dioxine- 2,4'-piperidin]-4-one, tert-butyl (3-(4-oxo-4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r- ylcarbonyl)benzo[b]thiophen-2-yl)carbamate, r-(2-aminobenzo[b]thiophene-3-carbonyl)-4H- spiro [benzo[d] [1,3] dioxine-2,4'-piperidin] -4-one, or 1 -ethyl-3 -(3-(4-oxo-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r-ylcarbonyl)benz o[b]thiophen-2-yl)urea, or a pharmaceutically acceptable salt thereof.

[8] Certain embodiments of the present disclosure are directed to a pharmaceutical

composition comprising a benzodioxinone compound of Formula I (e.g., a compound of Formula 1-1, Formula la, Formula lb, Formula II-1 to II -4, Formula III-1 to III-10 or Formula IV-l to Formula IV-10, or Examples 2-15) as defined herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutical composition further comprises one or more additional active agents.

[9] The pharmaceutical composition described herein can be formulated for different routes of administration. In some embodiments, the pharmaceutical composition can be formulated for intravenous injection or infusion, oral administration, inhalation, or topical administration. In some embodiments, the pharmaceutical composition can be formulated in the form of a topical solution, lotion, shampoo, transdermal spray, topical film, foam, powder, paste, sponge, transdermal patch, tincture, tape, cream, gel, or ointment.

[10] Certain embodiments of the present disclosure are directed to a method of inhibiting

ACC1 and/or ACC2 activities in a cell. In some embodiments, the method comprises contacting the cell with a benzodioxinone compound described herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein.

[11] In some embodiments, the present disclosure also provides a method of inhibiting

malonyl-CoA production, lipogenesis in a cell (e.g., a sebocyte, an adipocyte, or a hepatocyte), proliferation of cells (e.g., sebocytes, keratinocytes, adipocytes, melanocytes, squamous cells, Merkel cells, Langerhans cells, or skin stem cells, e.g., in epidermis, dermis, and/or hypodermis), differentiation of fibroblast to adipocytes, sebum production, or a combination thereof, in a subject in need thereof. In some embodiments, the subject is characterized as having a disease or disorder chosen from acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea,

fibrofolliculomas in Birt-Hogg-Dube syndrome, and combinations thereof. In some embodiments, the method comprises administering to the subject an effective amount of a benzodioxinone compound described herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein.

[12] Certain embodiments of the present disclosure are directed to a method of treating a

disease or disorder associated with ACC1 and/or ACC2 in a subject in need thereof. In some embodiments, the method comprises administering to the subject in need thereof a therapeutically effective amount of a benzodioxinone compound described herein (e.g., a compound of Formula I, Formula 1-1, Formula la, Formula lb, Formula II-1 to II-4, Formula III-1 to III- 10 or IV-l to IV- 10, or Examples 2-15), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, the method further comprises treating the subject in need thereof with one or more additional therapies for the respective disease or disorder (e.g., as described herein). Non-limiting diseases or disorders suitable to be treated with the methods described herein include a skin disease, such as a disease or disorder associated with aberrant sebocyte and/or keratinocyte activities (e.g., acne), a non-alcoholic fatty liver disease (such as nonalcoholic steatohepatitis (NASH)), a metabolic disease or disorder (e.g., obesity or diabetes), and cancer (e.g., non small cell lung cancer).

[13] In some embodiments, the present disclosure also provides a method of delivering

salicyclic acid or a derivative thereof to a subject in need thereof. In some embodiments, the method comprises administering (e.g., topically) to the subject a benzodioxinone compound of the present disclosure (e.g., a compound of Formula I, Formula 1-1, Formula la, Formula lb, Formula II-1 to II-4, Formula III-1 to III-10 or IV-l to IV-10, or Examples 2-15), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, R 1 and R 2 in Formula I of the benzodioxinone compound herein are both hydrogen, and the administering can deliver salicyclic acid to the subject, for example, in an anti-inflammatory amount.

[14] In some embodiments, the present disclosure also provides a method of treating

inflammation in a subject in need thereof. In some embodiments, the method comprises administering to the subject a benzodioxinone compound of the present disclosure (e.g., a compound of Formula I, Formula 1-1, Formula la, Formula lb, Formula II-1 to II-4, Formula III-1 to III- 10 or IV-l to IV- 10, or Examples 2-15), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, R 1 and R 2 in Formula I of the benzodioxinone compound herein are both hydrogen, and the administering can deliver salicyclic acid to the subject, for example, in an anti-inflammatory amount. In some embodiments, the inflammation is associated with a skin disease, e.g., as described herein.

[15] Additional embodiments and advantages of the disclosure will be set forth, in part, in the description that follows, and will flow from the description, or can be learned by practice of the disclosure. The embodiments and advantages of the disclosure will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. [16] It is to be understood that both the foregoing summary and the following detailed description are exemplary and explanatory only, and are not restrictive of the invention as claimed.

BRIEF DESCRIPTION OF THE DRAWINGS

[17] Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention.

[18] FIG. 1 shows images of Immunohistochemical (IHC) staining of human normal skin structure including layers of epidermis, dermis, and hypodermis. The top rows show images with lOx magnification, and the bottom rows show images with 40 x magnification of the sebaceous gland appearing in the images on top. Images on the left column show IHC staining by using control rabbit IgG; whereas images on the right column show IHC staining by using rabbit anti-ACC antibody. As shown in the bottom right image, ACC proteins are abundantly and specifically expressed in sebaceous gland, see e.g., where Arrow 1 points to. The figure also shows nuclei stained by hematoxylin (see e.g., where Arrow 2 points to) and cytosol stained by eosin (see e.g., where Arrow 3 points to).

[19] FIG. 2 shows images of Immunohistochemical (IHC) staining of human normal skin.

The left column shows images with lOx magnification, and the right column shows images with 40x magnification of the epidermis part shown on the left images. Images on the top row show IHC staining by using control rabbit IgG; whereas images on the bottom row show IHC staining by using rabbit anti-ACC antibody. The figure also shows nuclei stained by hematoxylin (see e.g., where Arrow 2 points to) and cytosol stained by eosin (see e.g., where Arrow 3 points to). There is no apparent difference of dark color between top (IgG control) and bottom pictures (anti-ACC), indicating ACC proteins are not abundantly expressed in keratinocytes of human normal skin.

[20] FIG. 3 shows images of Immunohistochemical (IHC) staining of human in situ squamous cell carcinoma (indicated by arrows) with adjacent skin tissue including sebaceous gland.

The left column shows images with lOx magnification, and the right column shows images with 40x magnification of the epidermis part shown on the left images. Images on the top row show IHC staining by using control rabbit IgG; whereas images on the botom row show IHC staining by using rabbit anti-ACC antibody. The figure also shows nuclei stained by hematoxylin (see e.g., where Arrow 2 points to) and cytosol stained by eosin (see e.g., where Arrow 3 points to). Tumor cells in the squamous cell carcinoma, sebaceous gland and epidermal squamous cells are stained with comparable density of dark color (see e.g., where Arrow 1 points to), indicating abundant expression of ACC proteins.

[21] FIG. 4 shows images of Immunohistochemical (IHC) staining of human seborrheic

keratosis. The left column shows images with lOx magnification, and the right column shows images with 40x magnification of the epidermis part shown on the left images.

Images on the top row show IHC staining by using control rabbit IgG; whereas images on the bottom row show IHC staining by using rabbit anti-ACC antibody. The figure also shows nuclei stained by hematoxylin (see e.g., where Arrow 2 points to) and cytosol stained by eosin (see e.g., where Arrow 3 points to). Keratin layer is apparent in this disease and keratinocytes next to the keratin layer are stained with dark color (see e.g., where Arrow 1 points to), indicating abundant expression of ACC proteins in keratinocytes in skin disease of keratosis.

DETAILED DESCRIPTION OF THE INVENTION

[22] The present disclosure is generally related to a novel class of benzodioxinone compounds that can be used as an ACC inhibitor (ACC1 and/or ACC2 inhibitor). In various

embodiments, the present disclosure provides compounds, pharmaceutical compositions, methods, and uses related to the novel benzodioxinone compounds described herein.

Benzodioxinone Compounds

[23] Certain embodiments of the present disclosure are directed to novel benzodioxinone compounds. In some embodiment, the present disclosure provides a compound of Formula I, or a pharmaceutically acceptable salt thereof,

Fonnula I,

wherein:

R 1 and R 2 are each independently hydrogen, halogen, cyano, an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), an optionally substituted alkenyl (e.g., an optionally substituted C2-6 alkenyl), an optionally substituted alkynyl (e.g., an optionally substituted C2-6 alkynyl), an optionally substituted aryl (e.g., an optionally substituted C 6- 10 aryl), an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl), an optionally substituted heterocyclyl (e.g., an optionally substituted 4-6 membered heterocyclyl), -NR 10 R n , -COOR 12 , -CONR 13 R 14 , -CN, -S(0) n R 15 , or -OR 16 ;

wherein

R 10 and R 11 are each independently hydrogen, an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), an optionally substituted alkanoyl (e.g., an optionally substituted Ci-6 alkanoyl), an optionally substituted cycloalkanoyl (e.g., an optionally substituted C3-6 cycloalkanoyl), an optionally substituted aryl (e.g., an optionally substituted C6-10 aryl), an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl), an optionally substituted heterocyclyl (e.g., an optionally substituted 4-6 membered heterocyclyl), -COOR 12 , or -CONR 13 R 14 ;

R 12 , R 13 and R 14 are each independently hydrogen or an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl);

n is 0, 1, or 2;

R 15 is an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), or -NR !O R 11 ;

R 16 is hydrogen, an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), an optionally substituted alkanoyl (e.g., an optionally substituted Ci- 6 alkanoyl), an optionally substituted cycloalkanoyl (e.g., an optionally substituted C3-6 cycloalkanoyl), an optionally substituted aryl (e.g., an optionally substituted C6-10 aryl), an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl), an optionally substituted heterocyclyl (e.g., an optionally substituted 4-6 membered heterocyclyl) or -CONR 13 R 14 ; and

R 3 is an optionally substituted aryl (e.g., an optionally substituted C 6 -io aryl) or an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl).

[24] In some embodiments, both R 1 and R 2 are hydrogen. In some embodiments, only one of R 1 and R 2 is hydrogen. In some embodiments, neither of R 1 and R 2 is hydrogen.

[25] In some preferred embodiments, the compound of Formula I is a compound of Formula I- 1 :

Formula 1-1,

wherein R 1 , R 2 and R 3 are as defined herein. In some embodiments, R 1 is not hydrogen. In some embodiments, R 1 is hydrogen.

[26] In any of the embodiments described herein, R 2 can be hydrogen. For example, in some embodiments, the compound of Formula I is a compound of Formula la.

Formula la,

wherein R 1 and R 3 are as defined herein. In some embodiments, R 1 is not hydrogen. In some embodiments, R 1 is hydrogen. As would be appreciated by those skilled in the art, when R 1 is hydrogen, the compound of Formula la can be hydrolyzed into salicyclic acid, in vitro or in vivo. Thus, such compounds can serve at least a dual function, an ACC inhibitor and a precursor to salicyclic acid, and therefore can have anti-inflammatory effects independent of ACC inhibition.

[27] In some embodiments, R 1 is a halogen, such as F, Cl, Br, or I. In some embodiments, R 1 is -CN. When R 1 is a halogen, the compound of Formula I (e.g., a compound of Formula la) can also serve as a synthetic intermediate for the synthesis of other compounds, such as compounds of Formula I wherein R 1 is an optionally substituted alkyl, an optionally substituted cycloalkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl, -NR 10 R n , -COOR 12 , -CONR 13 R 14 , -CN,

-S(0)nR 15 , or -OR 16 , through one or more coupling reactions (e.g., a palladium mediated or copper mediated coupling reaction). For example, the aryl halide can be first converted into an aryl boronic acid or ester derivative, which can then couple with various groups such as an amine, to form a compound of Formula I. In some embodiments, the aryl halide can be first converted into a pinacol arylboronate, which can then be converted into the corresponding aryl boronic acid for use in the coupling reactions (e.g., a palladium mediated or copper mediated coupling reaction).

[28] In some embodiments, R 1 is an optionally substituted alkyl. In some embodiments, R 1 is an optionally substituted Ci- 6 alkyl. In some preferred embodiments, R 1 is an optionally substituted CM alkyl. For example, in some embodiments, R 1 can be a CM alkyl (e.g., methyl, ethyl, isopropyl, sec-butyl, tert-butyl). In some embodiments, R 1 can be a CM alkyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and CM alkyl. For example, R 1 can be a CM alkyl optionally substituted with 1-3 fluorines, such as CF 3 , CF 3 CH 2 , etc.

[29] In some embodiments, R 1 is an optionally substituted alkoxy. In some embodiments, R 1 is an optionally substituted C1-6 alkoxy. In some preferred embodiments, R 1 is an optionally substituted CM alkoxy. For example, R 1 can be a CM alkoxy (e.g., methoxy, ethoxy, isopropoxy, sec-butoxy, tert-butoxy). In some embodiments, R 1 can be a CM alkoxy optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and CM alkyl. For example, R 1 can be a CM alkoxy optionally substituted with 1-3 fluorines, such as CF 3 O, CF 3 CH 2 O, etc.

[30] In some embodiments, R 1 is an optionally substituted cycloalkyl. In some embodiments, R 1 is an optionally substituted C 3-6 cycloalkyl. In some embodiments, R 1 is a C 3-6 cycloalkyl. In some embodiments, R 1 is a C 3-6 cycloalkyl, optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C M alkyl. For example, in some embodiments, R 1 is cyclopropyl or cyclobutyl. In some embodiments, R 1 is cyclopropyl or cyclobutyl, optionally substituted with 1-3 fluorines. [31] In some embodiments, R 1 is an optionally substituted cycloalkoxy. In some embodiments, R 1 is an optionally substituted C3-6 cycloalkoxy. In some embodiments, R 1 is a C3-6 cycloalkoxy. In some embodiments, R 1 is a C3-6 cycloalkoxy, optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C 14 alkyl. For example, in some embodiments, R 1 is cyclopropoxy or cyclobutoxy. In some embodiments, R 1 is cyclopropoxy or cyclobutoxy, optionally substituted with 1-3 fluorines.

[32] In some preferred embodiments, R 1 is halogen, an optionally substituted Ci- 6 alkyl, an optionally substituted Ci- 6 alkoxy, an optionally substituted C3-6 cycloalkyl, or an optionally substituted C3-6 cycloalkoxy. In some preferred embodiments, R 1 is C1-4 alkyl, C14 alkoxy, C3-6 cycloalkyl, or C3-6 cycloalkoxy, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and C14 alkyl. In some preferred embodiments, R 1 is C14 alkyl or C14 alkoxy, each optionally substituted with 1-3 fluorines.

[33] In some embodiments, R 1 is an optionally substituted aryl, such as optionally substituted C6-12 aryl (e.g., phenyl). In some embodiments, R 1 is a C 6- i2 aryl (e.g., phenyl) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C 14 alkyl, optionally substituted C 14 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy. In some embodiments, R 1 is a C 6- i2 aryl (e.g., phenyl) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen (e.g., F, Cl), cyano, C 14 alkyl, C 14 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C 14 alkyl.

[34] In some embodiments, R 1 is an optionally substituted heteroaryl. For example, in some embodiments, R 1 is a 5-10 membered heteroaryl (e.g., a 5 or 6 membered heteroaryl as described herein) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C 14 alkyl, optionally substituted C 14 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy. In some embodiments, R 1 is a 5-10 membered heteroaryl (e.g., a 5 or 6 membered heteroaryl as described herein) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen (e.g., F, Cl), hydroxyl, cyano, C 1-4 alkyl, C 14 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and CM alkyl.

[35] In some embodiments, R 1 can also be an optionally substituted heterocyclyl. For

example, in some embodiments, R 1 is a 4-6 membered heterocyclyl (e.g., piperidinyl, piperazinyl, morpholinyl, etc.) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen, oxo, optionally substituted C1-4 alkyl, optionally substituted CM alkoxy, optionally substituted C 3-6 cycloalkyl, and optionally substituted C 3-6 cycloalkoxy. In some embodiments, R 1 is a 4-6 membered heterocyclyl (e.g., piperidinyl, piperazinyl, morpholinyl, etc.) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen (e.g., F, Cl), C alkyl, CM alkoxy, C 3-6 cycloalkyl, and C 3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C M alkyl.

[36] In some preferred embodiments, R 1 is -NR 10 R n . For example, in some embodiments, the compound of Formula la is a compound of Formula lb:

Formula lb,

wherein R 10 , R 11 and R 3 are as defined herein. In some embodiments, one of R 10 and R 11 is an optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl (e.g., as described herein). In some embodiments, one of R 10 and R 11 is a 5-membered heteroaryl (e.g., those having 2-4 nitrogen atoms as described herein) optionally substituted with one or more (e.g., 1 or 2) substituents independently chosen from halogen, hydroxyl, cyano and C M alkyl. For example, in some embodiments, one of R 10 and R 11 is a pyrazolyl, triazolyl or tetrazolyl. In some embodiments, one of R 10 and R 11 is pyrazolyl, triazolyl or tetrazolyl, each optionally substituted with 1 or 2 independently chosen C M alkyl (e.g., methyl or ethyl). In any of these embodiments, the other of R 10 and R 11 can be hydrogen or an optionally substituted alkyl (e.g., a C M alkyl such as methyl or ethyl). Other suitable R 10 and R 11 are described herein. [37] In some embodiments, R 1 can be a carboxylic acid derivative such as -COOR 12 or - CONR 13 R 14 , wherein R 12 , R 13 and R 14 are defined herein. For example, in some

embodiments, R 1 is -COOR 12 , and R 12 can be hydrogen or an optionally substituted alkyl. In some embodiments, R 12 can be hydrogen or an optionally substituted Ci- 6 alkyl, such as a C M alkyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and C1-4 alkyl. In some embodiments, R 1 is -CONR 13 R 14 , and R 13 and R 14 are each independently hydrogen or an optionally substituted alkyl. In some embodiments, R 13 and R 14 are each independently hydrogen or an optionally substituted Ci- 6 alkyl, such as a C M alkyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and C1-4 alkyl.

[38] In some embodiments, R 1 can be -S(0) n R 15 , wherein n and R 15 are defined herein. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, R 15 is an optionally substituted alkyl. In some embodiments, R 15 is an optionally substituted Ci- 6 alkyl, such as a C14 alkyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and Ci4 alkyl. In some embodiments, R 15 is an optionally substituted cycloalkyl. In some embodiments, R 15 is an optionally substituted C3-6 cycloalkyl, such as a C3-6 cycloalkyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen and C 14 alkyl. In some embodiments, R 15 is -NR 10 R n , suitable groups for R 10 and R 11 are described herein.

[39] In some embodiments, R 1 can be -OR 16 , wherein R 16 is defined herein. In some

embodiments, R 16 is H. In some preferred embodiments, R 16 is an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl) or an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), thus R 1 is an optionally substituted alkoxy or cycloalkoxy as described herein. Other suitable R 16 is described herein.

[40] Suitable R 3 for compounds of Formula I (e.g., compounds of Formula 1-1, Formula la, or Formula lb) are described herein. In some embodiments, R 3 is an optionally substituted aryl, for example, an optionally substituted C 6 -ioaryl. In some embodiments, R 3 is a C 6 -ioaryl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C 14 alkyl, optionally substituted C1-4 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy. In some embodiments, R 3 can be an optionally substituted phenyl. In some embodiments, R 3 can be an optionally substituted bicyclic aryl. For example, in some preferred embodiments, R 3 is an optionally substituted naphthyl. In some embodiments, R 3 is a naphthyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, CM alkyl, C 1-4 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C 1-4 alkyl. In some embodiments, R 3 is a naphthyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, C 14 alkyl optionally substituted with 1- 3 halogens, C 14 alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens. In some embodiments, R 3 is a naphthyl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, ethyl,

trifluoromethyl, trifluoromethoxy, methoxy, ethoxy, and cyclopropyl.

[41] In some embodiments, R 3 can also be an optionally substituted heteroaryl. For example, in some embodiments, R 3 is an optionally substituted 8-10 membered bicyclic heteroaryl. In some embodiments, R 3 is a 8-10 membered bicyclic heteroaryl optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, hydroxyl, cyano, optionally substituted C 14 alkyl, optionally substituted C1-4 alkoxy, optionally substituted C3-6 cycloalkyl, and optionally substituted C3-6 cycloalkoxy. In some embodiments, R 3 is an optionally substituted benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl. In some embodiments, R 3 is benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, cyano, hydroxyl, C 1-4 alkyl, C 14 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C 14 alkyl. In some embodiments, R 3 is benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, cyano, hydroxyl, C 14 alkyl optionally substituted with 1-3 halogens, C 1-4 alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens. In some embodiments, R 3 is benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with one or more (e.g., 1-3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, ethyl, trifluoromethyl, trifluoromethoxy, methoxy, ethoxy, and cyclopropyl.

Preferably, substitutions of the heteroaryls herein do not result in a halogen, cyano, or oxygen atom (from a hydroxyl, alkoxy or cycloalkoxy group) directly attach to a ring heteroatom, such as a ring nitrogen atom, of the heteroaryls. Also, those skilled in the art would understand that when a compound has a hydroxyl group attached to a carbon next to a nitrogen ring atom, the compound may exist predominantly in one or more tautomeric forms. For example, a 2-hydroxyl substituted 1 -methyl-benzimidazole may exist predominantly in Form B as shown below

Form B

Form A . Thus, as used herein, a heteroaryl substituted with a hydroxyl group should be understood as encompassing all tautomeric forms when possible, e.g., Forms A and B above.

[42] Various positions of the bicyclic aryls or heteroaryls suitable for R 3 can be used to attach to the benzodioxinone core, for example, a position that is not immediately adjacent to the distal ring. For example, both 1- and 2-naphthyl are suitable R 3 groups. In some

embodiments, R 3 is preferably 2-naphthyl. Similarly, quinolinyl can have seven different

attach points at one of positions some embodiments, R 3 is preferably 2-

, 3-, 6-, or 7-quinolinyl. In some embodiments, R 3 is 4-, 5-, or 8-quinolinyl.

[43] In any of the embodiments described herein, R 3 can be optionally substituted naphthyl, quinolinyl, benzimidazolyl, pyrrolopyridinyl, or indazolyl:

In some embodiments, the naphthyl, quinolinyl, benzimidazolyl, pyrrolopyridinyl, or indazolyl is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, cyano, hydroxyl, C1-4 alkyl, C1-4 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen (e.g., F) and C14 alkyl. In some embodiments, the naphthyl, quinolinyl, benzimidazolyl, pyrrolopyridinyl, or indazolyl is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, cyano, hydroxyl, C14 alkyl optionally substituted with 1-3 halogens, C1-4 alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens. In some embodiments, the naphthyl, quinolinyl,

benzimidazolyl, pyrrolopyridinyl, or indazolyl is optionally substituted with one or more (e.g., 1-3) substituents independently chosen from fluoro, chloro, hydroxyl, methyl, trifluoromethyl, ethyl, methoxy, trifluoromethoxy, ethoxy, and cyclopropyl. Preferably, substitutions of the benzimidazolyl, pyrrolopyridinyl, or indazolyl herein do not result in a halogen, cyano, or oxygen atom (from a hydroxyl, alkoxy or cycloalkoxy group) directly attach to a ring nitrogen atom.

[44] In some embodiments, R 3 can be an optionally substituted benzo[b]thienyl (abbreviated benzothienyl). The benzo[b]thienyl can be connected to the remainder of the molecule via various attaching points, for example, through the 2- or 3- position. In some embodiments,

R 3 can be

optionally substituted with one or more (e.g., 1-3) substituents independently chosen from halogen, -NR 30 R 31 , cyano, hydroxyl, C 14 alkyl optionally substituted with 1-3 halogens, C 14 alkoxy optionally substituted with 1-3 halogens, C 3-6 cycloalkyl optionally substituted with 1-3 halogens, and C 3-6 cycloalkoxy optionally substituted with 1-3 halogens, wherein R 30 and R 31 are each independently hydrogen, an optionally substituted alkyl (e.g., an optionally substituted C 1-6 alkyl), an optionally substituted cycloalkyl (e.g., an optionally substituted C3-6 cycloalkyl), an optionally substituted alkanoyl (e.g., an optionally substituted C1-6 alkanoyl), an optionally substituted cycloalkanoyl (e.g., an optionally substituted C3-6 cycloalkanoyl), an optionally substituted aryl (e.g., an optionally substituted C6-10 aryl), an optionally substituted heteroaryl (e.g., an optionally substituted 5-10 membered heteroaryl), an optionally substituted heterocyclyl (e.g., an optionally substituted 4-6 membered heterocyclyl), -COOR 32 , or -CONR 33 R 34 ; wherein R 32 , R 33 and R 34 are each independently hydrogen or an optionally substituted alkyl (e.g., an optionally substituted Ci- 6 alkyl). In some embodiments, the benzo|b| thienyl can be substituted with -NR 30 R 31 , and optionally substituted with other suitable substituents described herein. In some embodiments, the substituent -NR 30 R 31 can be selected from - NH 2 , -NHCOOR 32 or -NHCONR 33 R 34 , wherein R 32 is a C 1-6 alkyl, R 33 and R 34 are each independently hydrogen or a C alkyl optionally substituted with 1-3 fluorines. In some embodiments, one of R 33 and R 34 is hydrogen and the other of R 33 and R 34 is hydrogen or a C 1-6 alkyl optionally substituted with 1-3 fluorines, for example, a C 14 alkyl (e.g., methyl, ethyl, etc.).

[45] In any of the embodiments described herein, R 3 can be

|46| In any of the embodiments described herein, R 3 can be

Exemplary Benzodioxinone Compounds

[47] Certain embodiments of the present disclosure are directed to some exemplary

benzodioxinone compounds.

[48] In some embodiments, the present disclosure provides a compound of Formula II-1 to Formula II -4, or a pharmaceutically acceptable salt thereof:

Formula II-3 Formula I I-4 wherein R la is H or CM alkyl optionally substituted with 1-3 fluorines (e.g., CF3, ethyl, isopropyl, tert-butyl); R lb is C1-4 alkoxyl optionally substituted with 1-3 fluorines (e.g., methoxy, trifluoromethoxy, ethoxy or isopropoxy); R lc is F, Cl, Br, -CN, or -C0 2 (Ci 4 alkyl) (e.g., -CC Me); R 3 can be any of those defined herein. For example, in some preferred embodiments, R 3 is an optionally substituted naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl. In some embodiments, R 3 is naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with 1-3 substituents independently chosen from halogen, cyano, hydroxyl, C1-4 alkyl, C1-4 alkoxy, C3-6 cycloalkyl, and C3-6 cycloalkoxy, wherein each of the alkyl, alkoxy, cycloalkyl, and cycloalkoxy is optionally substituted with one to three substituents independently chosen from halogen (e.g., F) and Ci- 4 alkyl. In some embodiments, R 3 is naphthyl, benzimidazolyl, pyrrolopyridinyl,

pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with 1-3 substituents independently chosen from halogen, cyano, hydroxyl, C14 alkyl optionally substituted with 1-3 halogens, C1-4 alkoxy optionally substituted with 1-3 halogens, C3-6 cycloalkyl optionally substituted with 1-3 halogens, and C3-6 cycloalkoxy optionally substituted with 1-3 halogens. In some embodiments, R 3 is naphthyl, benzimidazolyl, pyrrolopyridinyl, pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl, each optionally substituted with 1-3 substituents independently chosen from fluoro, chloro, hydroxyl, methyl, ethyl, trifluoromethyl, trifluoromethoxy, methoxy, ethoxy, and

cyclopropyl. Preferably, substitutions of the benzimidazolyl, pyrrolopyridinyl,

pyrazolopyridinyl, indazolyl, indolyl, quinolinyl or isoquinolinyl herein do not result in halogen, cyano, or oxygen atom (from a hydroxyl, alkoxy or cycloalkoxy group) directly attach to a ring nitrogen atom.

[49] In some embodiments, R 3 in Formula II-1 to Formula II-4 can be optionally substituted naphthyl, quinolinyl, benzimidazolyl, pyrrolopyridinyl, or indazolyl:

For example, each of the naphthyl, quinolinyl, benzimidazolyl, pyrrolopyridinyl, or indazolyl can be optionally substituted with 1-3 substituents independently chosen from fluoro, chloro, hydroxyl, methyl, trifluoromethyl, ethyl, methoxy, trifluoromethoxy, ethoxy, and cyclopropyl. In some embodiments, R 3 in Formula II-1 to Formula II-4 can be optionally substituted benzothienyl

For example, the benzothienyl can be substituted with -NR 30 R 31 , and optionally substituted with other suitable substituents described herein. In some embodiments, the substituent - NR 30 R 31 can be selected from -NFh, -NHCOOR 32 or -NHCONR 33 R 34 , wherein R 32 is a C 1-6 alkyl, R 33 and R 34 are each independently hydrogen or a C 1-6 alkyl optionally substituted with 1-3 fluorines. In some embodiments, one of R 33 and R 34 is hydrogen and the other of R 33 and R 34 is hydrogen or a C 1-6 alkyl optionally substituted with 1-3 fluorines, for example, a C 14 alkyl (e.g., methyl, ethyl, etc.).

[50] In some embodiments, the present disclosure provides a compound of Formula III-1 to Formula III- 10, or a pharmaceutically acceptable salt thereof:

wherein R 1 and R 2 can be any of those defined herein, and R 30 and R 31 , as applicable, can be any of those defined herein. In some embodiments, R 2 in Formula III-1 to Formula III-10 is hydrogen. In some embodiments, both R 30 and R 31 are hydrogen. In some embodiments, the substituent -NR 30 R 31 can be -NH 2 , -NH-C(0)-0-(CI-4 alkyl) (e.g., NHBoc) or -NH-C(O)- NH-(C i alkyl) (e.g., -NHCONHEt).

[51] In some embodiments, the present disclosure provides a compound of Formula IV-l to Formula IV- 10, or a pharmaceutically acceptable salt thereof:

wherein R 1 and R 2 , and R 30 and R 31 , as applicable, can be any of those defined herein. In some embodiments, R 2 in Formula IV-l to Formula IV-10 is hydrogen. In some

embodiments, both R 30 and R 31 are hydrogen. In some embodiments, the substituent - NR 30 R 31 can be -NH 2 , -NH-C(0)-0-(Ci4 alkyl) (e.g., NHBoc) or -NH-C(0)-NH-(Ci4 alkyl) (e g., -NHCONHEt).

[52] In some embodiments, R 1 in any of Formula III-1 to III- 10 and IV-l to IV- 10 can be hydrogen or an optionally substituted C M alkyl, for example, methyl, ethyl, isopropyl, or tert- butyl. In some embodiments, R 1 in any of Formula III-1 to III-10 and IV-l to IV-10 can be an optionally substituted C M alkoxy, for example, methoxy, trifluoromethoxy, ethoxy or isopropoxy. In some embodiments, R 1 in any of Formula III-1 to III-10 and IV-l to IV-10 can be -NR 10 R n . In some embodiments, one of R 10 and R 11 is an optionally substituted 5 or 6 membered heteroaryl. In some embodiments, one of R 10 and R 11 is a 5-membered heteroaryl having 2-4 ring nitrogen atoms (e.g., pyrazolyl, triazolyl or tetrazolyl), which is optionally substituted with 1 or 2 substituents independently chosen from halogen, hydroxyl, cyano and Ci- 4 alkyl. In some embodiments, one of R 10 and R 11 is a pyrazolyl, triazolyl or tetrazolyl, each optionally substituted with a CM alkyl. In any of these embodiments, the other of R 10 and R 11 can be hydrogen or C M alkyl (e.g., methyl). For example, in some embodiments,

N

one of R 10 and R 11 is , and the other of R 10 and R 11 is hydrogen or methyl. In some embodiments, R 2 in any of Formula III-1 to III-10 and IV-l to IV-10 can be hydrogen. Other suitable R 1 and R 2 are described herein.

[53] In some embodiments, the present disclosure also provides the following compounds:

or a pharmaceutically acceptable salt thereof.

Method of Synthesis

[54] Certain embodiments of the present disclosure are directed to methods of synthesizing the benzodioxinone compounds described herein.

[55] In some embodiments, the present disclosure provides a method of synthesizing a

compound of Formula I,

Formula I,

wherein R 1 , R 2 , and R 3 are as defined herein. In some embodiments, the method comprises reacting a compound of Formula S-l, or a salt thereof, with an acid of Formula S-2, or an activated form thereof (e.g., acyl chloride, anhydride, etc.) to form a compound of Formula I,

wherein R 1 , R 2 , and R 3 are as defined herein. Suitable reagents and conditions for this transformation are known in the art and exemplified in the Examples section, e.g., using HATU ((l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]py ridinium 3-oxide hexafluorophosphate)) and diisopropyl amine. It should be noted that compounds of Formula S-l, including all variations corresponding to those as described herein for Formula I such as Formula 1-1, la, lb, Formula II-1 to II-4, Formula III-1 to III-10, Formula IV-l to IV-10, etc., or salts thereof are also novel compounds and embodiments of the present invention.

[56] In some embodiments, the method comprises reacting a compound of Formula S-3 with R'G 2 to form a compound of Formula I,

wherein G 1 is halo (preferably Br or I), or B(OH) 2 (or a corresponding boronic ester, such as a pinacol boronate), and R 1 , R 2 , and R 3 are as defined herein, wherein R'G 2 is a reagent capable of reacting with Formula S-3 to form Formula I under suitable conditions. For example, in some embodiments, R'G 2 is an amino compound with G 2 being H, and G 1 is B(OH) 2 (or a corresponding boronic ester), and R'G 2 can react with the compound of Formula S-3, for example, under copper catalyzed (such as in the presence of Cu(OAc) 2 ) coupling reaction conditions to form the compound of Formula I. In some embodiments, R 2 is H. In some embodiments, the compound of Formula S-3 is Formula S-3a, wherein G 1 is B(OH) 2 (or a corresponding boronic ester), which can couple with R'G 2 to form a compound of Formula la:

In some embodiments, Suitable coupling reaction conditions are known in the art and are exemplified in the Examples section.

[57] In some embodiments, the method comprises reacting a compound of Formula S-3b with R'G 2 to form a compound of Formula S-4, which can be deprotected to form a compound of Formula S-l,

Formula S-4 Formula S-1

Formula S-3b

wherein G 1 is halo (preferably Br or I) or B(OH) 2 (or a corresponding boronic ester such as pinacol boronate), Pg 1 is a nitrogen protecting group, such as Boc, benzyl, etc., and R 1 , R 2 , and R 3 are as defined herein, wherein R'G 2 is a reagent capable of reacting with Formula S- 3b to form Formula S-4 under suitable conditions. For example, in some embodiments, R'G 2 is an amino compound with G 2 being H, and G 1 is B(OH) 2 (or a corresponding boronic ester), and R'G 2 can react with the compound of Formula S-3b, for example, under copper catalyzed coupling reaction conditions to form the compound of Formula S-4. In some embodiments, R 2 is H. For example, in some embodiments, the compound of Formula S-3b is a compound of Formula S-3c, wherein G 1 is B(OH) 2 (or a corresponding boronic ester), which can couple with R'G 2 to form a compound of Formula S-4a, which upon deprotection can form a compound of Formula S-l a.

In some embodiments, Suitable coupling reaction conditions are known in the art and are exemplified in the Examples section. Suitable nitrogen protecting groups and deprotection conditions are known in the art, for example, as described in “Protective Groups in Organic Synthesis”, 4 th ed. P. G. M. Wuts; T. W. Greene, John Wiley, 2007, and references cited therein.

[58] Compounds of Formula S-3, S-3a, S-3b, and S-3c can be prepared by following the methods described herein. For example, in some embodiments, the benzodioxinone compound of Formula S-3c, wherein G 1 is B(OH) 2 , can be prepared from the corresponding bromide by following the following scheme:

Suitable conditions for this transformation are known in the art and are exemplified in the Examples section. By following similar methods, the benzodioxinone compound of Formula S-3a, wherein G 1 is B(OH) 2 , can also be prepared from the corresponding bromide compound:

The halogenated compound, e.g., compound of Formula S-3 or S-3b, wherein G 1 is Br, can be prepared through oxidation, for example, as follows: , wherein the piperidine nitrogen is either attached to a nitrogen protecting group or -COR 3 . Suitable oxidation conditions are known in the art, for example, by using KMn0 4 , which is also exemplified in the Examples section.

[59] In some embodiments, R 1 is -COOR 12 and the method comprises reacting a compound of Formula S-3 or S-3b under palladium catalyzed coupling condition in the presence of carbon monoxide and R 12 OH (e.g., MeOH).

[60] Compounds of Formula S-l can be readily accessible by those skilled in the art in view of the present disclosure. For example, in some embodiments, compounds of Formula S-l can be prepared by following Scheme 1:

Scheme 1

Formula S-1

wherein Pg 2 is a nitrogen protecting group, such as Boc, benzyl, etc., and R 1 and R 2 are defined herein. Thus, the compound of Formula S-5 can, for example, react with the compound of Formula S-6 under ketal forming condition to form a spirocyclic compound of Formula S-7. The spirocyclic compound of Formula S-7 can then be oxidized (e.g., by KMnCri) to form a compound of Formula S-8, which can then be deprotected to form the compound of Formula S-l. Suitable nitrogen protecting groups and deprotection conditions are known in the art, for example, as described in“Protective Groups in Organic Synthesis”, 4 th ed. P. G. M. Wuts; T. W. Greene, John Wiley, 2007, and references cited therein. [61] In some embodiments, certain compounds of Formula S-l (such as Formula S-13 below) can also be prepared through an ester intermediate Formula S-9 by following Scheme 2 below:

Scheme 2

wherein R 12 and Pg 2 are as defined herein, G 3 and G 4 are independently an optionally substituted alkyl (e.g., a C M alkyl). Thus, the bromide of Formula S-7a can be converted into a compound of Formula S-9 under palladium catalyzed coupling condition in the presence of carbon monoxide and R 12 OH (e.g., MeOH). The ester of Formula S-9 can then be functionalized, e.g., through reaction with Grignard reagent (e.g., MeMgCl) to form an alcohol of Formula S-10. It should be noted that for the synthesis of compounds with different G 3 and G 4 groups, a selective reaction with the ester of Formula S-9 can be carried out first to form a ketone intermediate, which can then be further converted into the compound of Formula S-10. Alternatively, the ketone intermediate can be obtained from the bromide of Formula S-7a through appropriate coupling reactions. In some embodiments, both G 3 and G 4 groups are methyl. The ketal of Formula S-10 can then be oxidized (e.g., by KMnCri) to form a benzodioxinone compound of Formula S-l l. The alcohol function of Formula S-l 1 can then be reduced, e.g., through hydrogenation in the presence of Pd/C to form a compound of Formula S-12, which can then be deprotected to form an amine of Formula S-l 3.

[62] As will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in“Protective Groups in Organic Synthesis”, 4 th ed. P. G. M. Wuts; T. W. Greene, John Wiley, 2007, and references cited therein. The reagents for the reactions described herein are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the reagents are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA), Sigma (St. Louis, Missouri, USA). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-15 (John Wiley and Sons, 1991), Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1989), Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (Wiley, 7 th Edition), and Larock's Comprehensive Organic Transformations (Wiley -VCH, 1999).

Pharmaceutical Compositions

[63] Certain embodiments are directed to a pharmaceutical composition comprising one or more of the benzodioxinone compounds of the present disclosure.

[64] The pharmaceutical composition can optionally contain a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises a

benzodioxinone compound of the present disclosure and a pharmaceutically acceptable excipient. Pharmaceutically acceptable excipients are known in the art. Non-limiting suitable excipients include, for example, encapsulating materials or additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents and mixtures thereof. See also Remington's The Science and Practice of Pharmacy, 2lst Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, Md., 2005; incorporated herein by reference), which discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. [65] The pharmaceutical composition can include any one or more of the benzodioxinone compounds of the present disclosure. For example, in some embodiments, the

pharmaceutical composition comprises a compound of any of Formula I, Formula 1-1, Formula la, Formula lb, Formula II- 1 to II-4, Formula III-1 to III- 10 and IV- 1 to IV- 10, or a pharmaceutically acceptable salt thereof. In any of the embodiments described herein, the pharmaceutical composition comprises a compound of

or a pharmaceutically acceptable salt thereof.

[66] The pharmaceutical composition can also be formulated for delivery via different routes, such as oral, parenteral, inhalation, topical, etc.

[67] In some embodiments, the pharmaceutical composition is formulated for oral

administration. The oral formulations can be presented in discrete units, such as capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension in an aqueous or non- aqueous liquid; or as an oil-in-water or water-in-oil emulsion. Excipients for the preparation of compositions for oral administration are known in the art. Non-limiting suitable excipients include, for example, agar, alginic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, 1, 3-butylene glycol, carbomers, castor oil, cellulose, cellulose acetate, cocoa butter, com starch, com oil, cottonseed oil, cross-povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, groundnut oil, hydroxypropylmethyl cellulose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mannitol, monoglycerides, olive oil, peanut oil, potassium phosphate salts, potato starch, povidone, propylene glycol, Ringer's solution, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium phosphate salts, sodium lauryl sulfate, sodium sorbitol, soybean oil, stearic acids, stearyl fumarate, sucrose, surfactants, talc, tragacanth, tetrahydrofurfuryl alcohol, triglycerides, water, and mixtures thereof.

[68] In some embodiments, the pharmaceutical composition is formulated for parenteral

administration (such as intravenous injection or infusion). The parenteral formulations can be, for example, an aqueous solution, a suspension, or an emulsion. Excipients for the preparation of parenteral formulations are known in the art. Non-limiting suitable excipients include, for example, l,3-butanediol, castor oil, com oil, cotonseed oil, dextrose, germ oil, groundnut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, U.S.P. or isotonic sodium chloride solution, water and mixtures thereof.

[69] In some embodiments, the pharmaceutical composition is formulated for inhalation. The inhalable formulations can be, for example, formulated as a nasal spray, dry powder, or an aerosol administrable through a metered-dose inhaler. Excipients for preparing formulations for inhalation are known in the art. Non-limiting suitable excipients include, for example, lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, and mixtures of these substances. Sprays can additionally contain propellants, such as

chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

[70] In some embodiments, the pharmaceutical composition is formulated for topical

administration. The topical formulations can be, for example, in the form of a topical solution, lotion, shampoo, transdermal spray, topical film, foam, powder, paste, sponge, transdermal patch, tincture, tape, cream, gel, or ointment. Excipients for preparing topical formulations are known in the art. Non-limiting suitable excipients include, for example, animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, and mixtures thereof.

[71] For example, the topical formulations described herein can have one or more“lipophilic solvent(s)” that acts as a carrier into the pilosebaceous unit. A lipophilic solvent useful in the invention can be miscible with water and/or lower chain (e.g., Ci- 6 ) alcohols. In some embodiments, the lipophilic solvent can be a glycol, e.g., propylene glycol. In some embodiments, the lipophilic solvent can be a polyethylene glycol, e.g., with average molecular weight (e.g., M n or M w ) ranging from 200 to 20000 Dalton. In some embodiments, the lipophilic solvent is a glycol ether, e.g., diethylene glycol monoethyl ether (transcutol).

[72] The topical formulations described herein can also have one or more“filler(s)”. Non limiting useful fillers include water and lower (e.g., Ci- 6 ) alcohols, including ethanol, 2- propanol and n-propanol. In some embodiments, the filler is water, ethanol and/or 2- propanol. [73] The topical formulations described herein can also have one or more“humectant(s)” used to provide a moistening effect. Non-limiting useful humectants include glycerin, polyhydric alcohols and silicone oils. In some embodiments, the topic formulation comprises one or more humectants chosen from glycerin, propylene glycol and cyclomethicone.

[74] The topical formulations described herein can also have a gelling agent that increases the viscosity of the final formulation. In some embodiments, the gelling agent can also act as an emulsifying agent. Non-limiting useful gelling agents include classes of celluloses, acrylate polymers and acrylate copolymers, for example, hydroxypropyl cellulose, hydroxymethyl cellulose, Pluronic PF127 polymer, carbomer 980, carbomer 1342 and carbomer 940. In some embodiments, the topic formulation comprises one or more gelling agents chosen from hydroxypropyl cellulose (Klucel® EF, GF and/or HF), Pluronic PF127, carbomer 980 and carbomer 1342 (Pemulen® TR-l, TR-2 and/or Carbopol® ETD 2020).

[75] The topical formulations described herein can also have one or more anti-oxidants,

radical scavengers, and/or stabilizing agents. Non-limiting useful examples include butylatedhydroxytoluene, butylatedhydroxyanisole, ascorbyl palmitate, citric acid, vitamin E, vitamin E acetate, vitamin E-TPGS, ascorbic acid, tocophersolan and propyl gallate.

[76] The topical formulations described herein can also have one or more preservatives that exhibit anti-bacterial and/or anti-fungal properties. Non-limiting useful examples include diazolidinyl urea, methylparaben, propylparaben, tetrasodium EDTA, and ethylparaben.

[77] The topical formulations described herein can also have one or more chelating agents.

Non-limiting examples for use herein include EDTA, disodium edeate, dipotassium edeate, cyclodextrin, trisodium edetate, tetrasodium edetate, citric acid, sodium citrate, gluconic acid and potassium gluconate.

[78] The pharmaceutical composition can include various amounts of the benzodioxinone compounds of the present disclosure, depending on various factors such as the intended use and potency of the compound. For example, in some embodiments, the pharmaceutical composition can comprise a benzodioxinone compound of the present disclosure in an amount effective for inhibiting ACC1 and/or ACC2 activities in a cell, which is in vitro, in vivo, or ex vivo. In some embodiments, the amount is effective to achieve about 10%, about 20%, about 50%, about 70%, about 90%, about 99% inhibition of the ACC1 and/or ACC2 activities, compared to a control, or any ranges between the recited values. [79] In some embodiments, the pharmaceutical composition can comprise the benzodioxinone compound of the present disclosure in an amount, when administered to a subject (e.g., a human) in need thereof, effective to inhibit one or more activities in the subject. In some embodiments, the one or more activities are chosen from acetyl-CoA carboxylases ACC1 and/or ACC2 activities, lipogenesis, proliferation of cells (e.g., adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, or skin stem cells) in epidermis, dermis, and/or hypodermis, proliferation of human sebocytes, proliferation of human keratinocytes, differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers, sebum production, inflammation, and combinations thereof. In some embodiments, the pharmaceutical composition can comprise the benzodioxinone compound of the present disclosure in an amount, when administered to a subject (e.g., a human) in need thereof, (a) effective to inhibit acetyl-CoA carboxylases ACC1 and/or ACC2 activities in a cell (e.g., a sebocyte, adipocyte) of the subject; (b) effective to inhibit lipogenesis (e.g., lipogenesis of sebocytes, lipogenesis of adipocytes, etc.) in the subject; (c) effective to inhibit proliferation of cells, such as adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, or skin stem cells, in epidermis, dermis, and/or hypodermis in the subject; (d) effective to inhibit proliferation of human sebocytes in the subject; (e) effective to inhibit proliferation of human keratinocytes in the subject; (f) effective to inhibit

differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers in the subject; (g) effective to inhibit sebum production in the subject; (h) effective to inhibit inflammation in the subject, or any combinations thereof. In some embodiments, the subject is characterized as having a disease or disorder chosen from acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea,

fibrofolliculomas in Birt-Hogg-Dube syndrome, and combinations thereof.

[80] In some embodiments, the pharmaceutical composition comprises a therapeutically

effective amount of a benzodioxinone compound of the present disclosure. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of the benzodioxinone compound of the present disclosure and a pharmaceutically acceptable excipient. As used herein, a therapeutically effective amount of a benzodioxinone compound of the present disclosure is an amount effective to treat a disease or disorder as described herein, which can depend on the recipient of the treatment, the disease or disorder being treated and the severity thereof, the composition containing the compound, the time of administration, the route of administration, the duration of treatment, the compound potency, its rate of clearance and whether or not another drug is co-administered.

[81] The pharmaceutical composition can be used for treating various diseases or disorders.

Non-limiting diseases or disorders suitable for such treatment include skin diseases (such as diseases or disorders associated with aberrant sebocyte and/or keratinocyte activity), non alcoholic fatty liver diseases or disorders, metabolic diseases or disorders, and proliferative diseases such as cancer (e.g., non-small cell lung cancer). Non-limiting suitable diseases or disorders associated with aberrant sebocyte and/or keratinocyte activity include, for example, acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea, fibrofolliculomas in Birt-Hogg-Dube syndrome, and a combination thereof. Non-limiting suitable non-alcoholic fatty liver diseases or disorders include, for example, non-alcoholic steatohepatitis (NASH). Non-limiting suitable metabolic diseases or disorders include, for example, obesity and/or diabetes.

[82] The pharmaceutical composition can also include one or more active agents in addition to the benzodioxinone compounds of the present disclosure. The benzodioxinone compound of the present disclosure and the one or more active agents can be present in a single dosage form (e.g., in a single pill, tablet, capsule, topic ointment, gel, paste, cream, etc.) or in separate dosage forms. For example, the benzodioxinone compound of the present disclosure can be in an oral or topical formulation, whereas the one or more active agents can be in the same formulation or a different oral or topical formulation. When in different dosage forms, the benzodioxinone compound of the present disclosure and one or more active agents can be included in a kit.

[83] Various active agents can be combined with the benzodioxinone compound of the present disclosure in the pharmaceutical composition. For example, in some embodiments, the one or more active agents are suitable for treating diseases or disorders associated with aberrant sebocyte and/or keratinocyte activity, such as acne. Non-limiting useful examples include antibiotics (e.g., clindamycin, erythromycin, metronidazole, sulfacetamide, or tetracyclines such as doxy cy cline and minocycline), retinoids (e.g., adapalene, isotretinoin, retinol, tazarotene, or tretinoin), and combinations thereof. In some embodiments, the one or more active agents are present in an amount effective for treating a disease or disorder associated with aberrant sebocyte and/or keratinocyte activity such as acne.

[84] In some embodiments, the one or more active agents are suitable for treating metabolic diseases or non-alcoholic fatty liver diseases. Non-limiting useful examples of such agents include angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacyl glycerol O- acyltransferase 1 or 2 (DGAT1 or DGAT2) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, famesoid X receptor (FXR) agonists, galectin-3 inhibitors, glucagon-like peptide 1 (GLP1) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 11 b-hydroxysteroid dehydrogenase ( 1 1 b-HSDl) inhibitors, IL-Ib antagonists, IL-6 antagonists, IL-10 agonists, IL-17 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors,

phospholipase C (PLC) inhibitors, PPAR-alpha agonists, PPAR-gamma agonists, PPAR-delta agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturase-l inhibitors, thyroid hormone receptor b agonists, tumor necrosis factor alpha (TNF-alpha) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, and combinations thereof. In some specific embodiments, the one or more active agents can be chosen from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX-1002, cenicriviroc, cobiprostone, colesevelam, emricasan, enalapril, GFT-505, GR-MD-02, hydrochlorothiazide, icosapent ethyl ester (ethyl eicosapentaenoic acid), IMM-124E, KD-025, linagliptin, liraglutide, mercaptamine, MGL-3196, obeti cholic acid, olesoxime, peg-ilodecakin, pioglitazone, PX- 102, remogliflozin etabonate, SHP-626, solithromycin, tipelukast, TRX-318, ursodeoxycholic acid, VBY-376, and combinations thereof. In some embodiments, the one or more active agents are present in a therapeutically effective amount for treating a non-alcoholic fatty liver disease (e.g., NASH). In some embodiments, the one or more active agents are present in a therapeutically effective amount for treating a metabolic disease (e.g., obesity and/or diabetes). Method of Treatment

[85] The benzodioxinone compounds of the present disclosure are useful for inhibiting ACC1 and/or ACC2 activities in a cell and for treating diseases or disorders associated with ACC1 and/or ACC2 activities (e.g., a metabolic disease, a disease associated with aberrant sebum production, or a non-alcoholic fatty liver disease).

[86] In some embodiments, the present disclosure provides a method of inhibiting ACC1 and/or ACC2 activities in a cell. In some embodiments, the method comprises contacting the cell with an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the cell can be in vitro (e.g., a human cell line), in vivo (e.g., a human cell in a human subject), or ex vivo (a human cell from a human subject).

[87] In some embodiments, inhibition of ACC1 and/or ACC2 activities can also lead to

inhibition of malonyl-CoA production in a cell, lipogenesis, proliferation of cells, differentiation of fibroblast to adipocytes, sebum production, and/or inflammation, which is implicated in diseases or disorders such as diseases or disorders associated with aberrant sebocyte and/or keratinocyte activity (e.g., acne, and others as described herein), metabolic diseases (e.g., obesity, diabetes, and others as described herein), non-alcohol fatty liver disease (e.g., NASH), and cancer (e.g., non-small cell lung cancer).

[88] Thus, in some embodiments, the present disclosure provides a method of inhibiting

lipogenesis in a cell. In some embodiments, the method comprises contacting the cell (e.g., a human cell) with an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the cell is a sebocyte, adipocyte, or a hepatocyte.

[89] In some embodiments, the present disclosure provides a method of inhibiting proliferation of cells. In some embodiments, the method comprises contacting the cells (e.g., human cells) with an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the cells are adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, and/or skin stem cells. In some embodiments, the cells are adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, and/or skin stem cells in epidermis, dermis, and/or hypodermis. In some embodiments, the cells are human sebocytes. In some embodiments, the cells are human keratinocytes. [90] In some embodiments, the present disclosure provides a method of inhibiting

differentiation of fibroblast (e.g., human fibroblast) to adipocytes. In some embodiments, the method comprises contacting the fibroblast with an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the method inhibits the differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers.

[91] In some embodiments, the present disclosure provides a method of inhibiting sebum production. In some embodiments, the method comprises contacting a sebocyte (e.g., a human sebocyte) with an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein.

[92] In some embodiments, the present disclosure also provides a method of inhibiting ACC1 and/or ACC2 activities and one or more (e.g., 1, 2, 3, 4, 5, 6, or all) activities in a subject (e.g., a human subject) in need thereof, wherein the one or more activities are chosen from (a) lipogenesis (e.g., lipogenesis of sebocytes, lipogenesis of adipocytes, lipogenesis of hepaocytes, etc.) in the subject; (b) proliferation of cells, such as adipocytes, melanocytes, keratinocytes, squamous cells, Merkel cells, Langerhans cells, or skin stem cells, in epidermis, dermis, and/or hypodermis in the subject; (c) proliferation of human sebocytes in the subject; (d) proliferation of human keratinocytes in the subject; (e) differentiation of fibroblast to adipocytes in cutaneous and/or subcutaneous layers in the subject; (f) sebum production in the subject; (g) inflammation in the subject, and combinations thereof, the method comprises administering to the subject an effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the subject is characterized as having a disease or disorder chosen from acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea, fibrofolliculomas in Birt-Hogg-Dube syndrome, and combinations thereof.

[93] In some embodiments, the present disclosure also provides a method of treating a disease or disorder associated with ACC1 and/or ACC2 in a subject in need thereof. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. Various diseases or disorders are associated with undesired or aberrant (e.g., hyperactive) ACC1 and/or ACC2 activities and can be treated by the methods described herein.

Method of Treating Skin Diseases

[94] Skin is the largest organ of human body that provides the interface between the external environment and the host. Lipids play an essential role in the formation and maintenance of both the permeability and antimicrobial barriers of skin. The normal function of skin is maintained by the hydrophobic extracellular lipid matrix in the stratum comeum composed primarily of ceramides, cholesterol, and free fatty acids, which prevents loss of water and electrolytes in the body. Sebaceous gland is the organ producing and secreting sebum into the follicular duct, which reaches the skin’s surface to maintain the normal function of the skin as barriers.

[95] Acne vulgaris (acne) is a common dermatological condition affecting many people. Acne is a chronic inflammatory dermatosis notable for open or closed comedones (blackheads and whiteheads) and inflammatory lesions, including papules, pustules, or nodules (also known as cysts). Zaenglein, A. et al, J. Am. Acad. Dermatol, 74: 945-73 (2016). Key pathogenic factors that play an important role in the development of acne are follicular

hyperkeratinization, microbial colonization with Propionibacterium acnes, sebum

production, and complex inflammatory mechanisms involving both innate and acquired immunity. Acne can have different severities, such as mild, moderate, and severe. The severity of the condition is affected by multiple factors including seasonal and psychological influences.

[96] A wide array of therapies for treating acne such as moderate to severe acne is available.

See e.g., Zaenglein, A. et al, J. Am. Acad. Dermatol, 74: 945-73 (2016). These therapies include, for example, (1) antibiotics (oral or topical), such as tetracycline, doxy cy cline, minocycline, trimethoprim/sulfamethoxazole (TMP/SMX), trimethoprim, erythromycin, clindamycin, azelaic acid, dapsone, azithromycin, amoxicillin, and cephalexin; (2) hormonal agents, such as combined oral contraceptives with an estrogen and a progestin, for example, ethinyl estradiol/norgestimate, ethinyl estradiol/norethindrone acetate/ferrous fumarate, ethinyl estradiol/drospirenone, and ethinyl estradiol/drospirenone/levomefolate; (3) oral retinoid, such as oral isotretinoin, (Accutane®, l3-cis-retinoic acid); (4) topical treatment, such as benzoyl peroxide (BP), salicylic acid, antibiotics, combination of antibiotics with BP, retinoids (e.g., tretinoin, adapalene, tazarotene), retinoid with BP, retinoid with antibiotic, azelaic acid, and sulfone agents (e.g., dapsone) etc.; and (5) others, such as spironolactone, flutamide, intralesional corticosteroid (triamcinolone acetonide), glycolic acid peels, salicylic acid peels, and resorcinol and salicylic acid. However, there are significant deficiencies in the currently available therapies for acne. Dermatological therapies are not fully effective against acne such as mild to moderate acne and many of the agents employed in these therapies produce skin irritation or have other side effects. See e.g., Zaenglein, A. et al., J.

Am. Acad. Dermatol, 74: 945-73 (2016). For example, Accutane was removed from the U.S. market in 2009 among lawsuits for potential inflammatory bowel disease side effects.

[97] Reducing sebum production as a means to treat acne has also been described, with results indicating that acne could significantly improve with a non-retinoid that inhibits sebum production. See, e.g., U.S. Patent No. 8,884,034 to Daynard, T. et al. (2014), quoting Zouboulis, C. C. et al,“Zileuton, an oral 5-lipoxygenase inhibitor, directly reduces sebum production”, Dermatology (2005), Vol. 210, pp. 36-38; and Zouboulis, C. C. et al,“A new concept for acne therapy: a pilot study with zileuton, an oral 5-lipoxygenase inhibitor”, Arch. Dermatol. (2003), Vol. 139, pp. 668-670.

[98] Sebaceous gland is filled with mature sebocytes, which is mainly occupied by lipid

droplets in cytosol and undergoes holocrine secretion. To maintain the normal function of sebaceous gland, mature sebocytes is constantly regenerated from epidermal stem cells. Overactivity of sebocytes, including proliferation, differentiation, and production of sebum can cause different skin diseases including acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea, and fibrofolliculomas in Birt- Hogg-Dube syndrome, etc.

[99] Although lipid metabolism is active in the skin, expression level and enzymatic activity of ACC in the skin, especially in sebaceous gland is not fully elucidated. It was unknown whether inhibiting ACC1 and/or ACC2 would interfere with the development and progression of the skin diseases mentioned above. Although it was proposed that olumacostat glasaretil acts as a prodrug of (5-(tetradecyloxy)-2-furancarboxylic acid, a purported ACC inhibitor, the observed effect of olumacostat glasaretil may not be due to ACC inhibition. [100] The present inventors have found that ACC proteins are abundantly and specifically expressed in sebaceous gland in human skin. This result first shows the localization of ACC proteins in human skin structure and provides evidence suggesting that administering ACC inhibitors to sebaceous gland, e.g., through topical administration of ACC inhibitors, can suppress lipid accumulation and progression of diseases related to overproduction of lipids in sebacytes by inhibiting ACCs in sebaceous gland. Thus, in some embodiments, the benzodioxinone compounds of the present disclosure can also be used for treating diseases or disorders associated with aberrant sebocyte and/or keratinocyte activities, e.g., through topical administration. As shown in the Examples section, exemplary representative benzodioxinone compounds of the present disclosure are inhibitors of ACC1 and/or ACC2. Further, exemplary representative benzodioxinone compounds of the present disclosure were shown to inhibit sebocytes activities such as cellular levels of malonyl-CoA of sebocytes.

[101] Accordingly, in some embodiments, the present disclosure provides a method of treating a disease or disorder associated with aberrant sebocyte and/or keratinocyte activity in a subject in need thereof. In some embodiments, the method comprises administering to the subject in need thereof a therapeutically effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. Non-limiting suitable diseases or disorders associated with aberrant sebocyte and/or keratinocyte activity include, for example, acne, seborrhea, sebaceous hyperplasia, seborrheic keratosis, sebaceous adenoma, sebaceous cyst, actinic keratosis, sebaceous carcinoma, squamous cell carcinoma, melanoma, phymatous rosacea, fibrofolliculomas in Birt-Hogg-Dube syndrome, and a combination thereof. Various routes of administration are suitable, for example, orally or topically. The method can use the benzodioxinone compound of the present disclosure as the active agent or use it in combination with another therapy. In some embodiments, the benzodioxinone compound of the present disclosure is the only active agent administered to the subject for treating the disease or disorder associated with aberrant sebocyte and/or keratinocyte activity. In some embodiments, the method is a combination therapy and further comprises treating the subject with one or more additional therapies effective for the treatment of the disease or disorder associated with aberrant sebocyte and/or keratinocyte activity. When used in a combination therapy, the benzodioxinone compound of the present disclosure can be administered to the subject concurrently with, prior to, or subsequent to the one or more additional therapies. The benzodioxinone compound of the present disclosure and the one or more additional therapies can be administered to the subject through the same or different routes. For example, in some embodiments, the benzodioxinone compound of the present disclosure can be administered topically, whereas the one or more additional therapies (e.g., additional active agents) can be administered orally.

[102] In some specific embodiments, the present disclosure provides a method of treating acne in a subject in need thereof. In some embodiments, the method comprises administering (e.g., topically) to the subject in need thereof a therapeutically effective amount of a benzodioxinone compounds of the present disclosure or a pharmaceutical composition described herein. In some embodiments, the method is a combination therapy and further comprises treating the subject with an additional therapy for acne. Any of the known therapies for treating acne can be part of the combination therapy, some of which are exemplified herein. For example, in some embodiments, the method further comprises administering to the subject an antibiotic, either topically or systemically, a retinoid, either topically or systemically, or a combination thereof. In some embodiments, the antibiotic is clindamycin, erythromycin, metronidazole, sulfacetamide, a tetracycline such as doxycycline and minocycline, or a combination thereof. In some embodiments, the retinoid is adapalene, isotretinoin, retinol, tazarotene, tretinoin, or a combination thereof. Other suitable antibiotics and retinoids are known in the art and exemplified herein.

Method of Treating Other Diseases

[103] Inhibitions of ACC1 and/or ACC2 activities have also been found to be associated with and/or useful for treating various metabolic diseases, non-alcoholic fatty liver diseases, and/or cancer (e.g.,. non-small cell lung cancer). See e.g., U.S. Pat. No. 8,288,405 and Griffith et al, Journal of medicinal chemistry 57: 10512-10526(2014) (obesity, diabetes); Harriman et al, Proceedings of the National Academy of Sciences of the United States of America 773:El796-l 805(2016) (nonalcoholic steatohepatitis (NASH), and Svensson et al, Nature medicine 22, 1108-1119(2016) ( non-small cell lung cancer).

[104] Accordingly, certain embodiments of the present disclosure are directed to the use of the benzodioxinone compounds of the present disclosure or pharmaceutical composition described herein, for the treatment of metabolic diseases, non-alcoholic fatty liver diseases, and/or cancer (e.g., non-small cell lung cancer). Various routes of administration are suitable, for example, orally or topically. The method can use the benzodioxinone compound of the present disclosure as the active agent or use it in combination with another therapy. In some embodiments, the benzodioxinone compound of the present disclosure is the only active agent administered to the subject for treating the metabolic diseases, non-alcoholic fatty liver diseases, and/or cancer (e.g., non-small cell lung cancer). In some embodiments, the method is a combination therapy and further comprises treating the subject with one or more additional therapies effective for the treatment of the metabolic diseases, non-alcoholic fatty liver diseases, and/or cancer (e.g., non-small cell lung cancer). When used in a combination therapy, the benzodioxinone compound of the present disclosure can be administered to the subject concurrently with, prior to, or subsequent to the one or more additional therapies. The benzodioxinone compound of the present disclosure and the one or more additional therapies can be administered to the subject through the same or different routes.

[105] In some specific embodiments, the present disclosure provides a method of treating

obesity and/or obesity-related disorders (e.g., overweight, weight gain, or weight

maintenance) in a subject (e.g., human subject) in need thereof. Obesity and overweight are generally defined by body mass index (BMI), which is correlated with total body fat and estimates the relative risk of disease. BMI is calculated by weight in kilograms divided by height in meters squared (kg/m 2 ). Overweight is typically defined as a BMI of 25-29.9 kg/m 2 , and obesity is typically defined as a BMI of 30 kg/m 2 . See, e.g., National Heart, Lung, and Blood Institute, Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults, The Evidence Report, Washington, D.C.: U.S.

Department of Health and Human Services, NIH publication no. 98-4083 (1998). In some embodiments, the method comprises administering (e.g., orally) to the subject in need thereof a therapeutically effective amount of a benzodioxinone compounds of the present disclosure or a pharmaceutical composition described herein.

[106] In some embodiments, the present disclosure also provides a method of treating diabetes and/or diabetes-related disorders in a subject (e.g., human subject) in need thereof, such as Type 1 (insulin-dependent diabetes mellitus, also referred to as“IDDM”) and Type 2 (noninsulin-dependent diabetes mellitus, also referred to as“NIDDM”) diabetes etc. In some embodiments, the method comprises administering (e.g., orally) to the subject in need thereof a therapeutically effective amount of a benzodioxinone compounds of the present disclosure or a pharmaceutical composition described herein. [107] In some embodiments, the present disclosure also provides a method of treating a non alcoholic fatty liver disease in a subject (e.g., human subject) in need thereof, for example, NASH. In some embodiments, the method comprises administering (e.g., orally) to the subject in need thereof a therapeutically effective amount of a benzodioxinone compounds of the present disclosure or a pharmaceutical composition described herein.

[108] In some embodiments, the methods of treating obesity, obesity-related disorders, diabetes, diabetes-related disorders, and/or non-alcoholic fatty liver diseases can be a combination therapy. For example, the methods can include administering to the subject one or more additional active agents in a therapeutically effective amount for treating the respective disease or disorder. Non-limiting useful agents for the combination therapy include angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacyl glycerol O- acyltransferase 1 or 2 (DGAT1 or DGAT2) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, famesoid X receptor (FXR) agonists, galectin-3 inhibitors, glucagon-like peptide 1 (GLP1) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 11 b-hydroxysteroid dehydrogenase ( 1 1 b-HSDl) inhibitors, IL-Ib antagonists, IL-6 antagonists, IL-10 agonists, IL-17 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors,

phospholipase C (PLC) inhibitors, PPAR-alpha agonists, PPAR-gamma agonists, PPAR-delta agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturase-l inhibitors, thyroid hormone receptor b agonists, tumor necrosis factor alpha (TNF-alpha) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, and combinations thereof. For example, in some embodiments, the one or more additional agents can be chosen from acetylsalicylic acid, alipogene tiparvovec, aramchol, atorvastatin, BLX-1002, cenicriviroc, cobiprostone, colesevelam, emricasan, enalapril, GFT-505, GR-MD-02, hydrochlorothiazide, icosapent ethyl ester (ethyl eicosapentaenoic acid), IMM-124E, KD-025, linagliptin, liraglutide, mercaptamine, MGL-3196, obeti cholic acid, olesoxime, peg-ilodecakin, pioglitazone, PX- 102, remogliflozin etabonate, SHP-626, solithromycin, tipelukast, TRX-318, ursodeoxycholic acid, VBY-376, and combinations thereof. [109] In some embodiments, the present disclosure also provides a method of treating cancer (e.g., non-small cell lung cancer), which comprises administering (e.g., orally) to a subject in need thereof a therapeutically effective amount of a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein.

[110] In some embodiments, the present disclosure also provides a method of delivering

salicyclic acid or a derivative thereof to a subject in need thereof, the method comprising administering to the subject a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. In some embodiments, where R 1 and R 2 in Formula I of the benzodioxinone compound are both hydrogen, administering the benzodioxinone compound can deliver salicyclic acid to the subject. In some embodiments, the administering delivers an anti-inflammatory amount of salicyclic acid to the subject. In some embodiments, the benzodioxinone compounds administered are hydrolyzed into salicyclic acid slowly in vivo, and thus the administering provides an extended release of salicyclic acid. In other words, both the benzodioxione compounds and salicyclic acid can be present over a period of time after administration. In such embodiments, the benzodioxinone compounds can have at least a dual function in vivo : inhibiting ACC activity and providing salicyclic acid, which can for example reduce inflammation. As shown in the Examples section, a few benzodioxinone compounds with both R 1 and R 2 as hydrogens were prepared and tested, see Examples 12-15. These compounds showed inhibition of ACC with an IC50 less than 10 uM and can be converted into salicyclic acid through hydrolysis. In some embodiments, the benzodioxinone compounds are administered to deliver salicyclic acid either locally and/or systemically. In some embodiments, the benzodioxinone compounds are administered topically. In some embodiments, the subject is characterized as having inflammation associated with a skin disease, e.g., as described herein.

[111] In some embodiments, the present disclosure also provides a method of treating

inflammation in a subject in need thereof, the method comprising administering to the subject a benzodioxinone compound of the present disclosure or a pharmaceutical composition described herein. As discussed herein, the compounds herein can be hydrolyzed into salicyclic acid or derivative, and can have anti-inflammatory effect independent of the inhibition of ACC. In some embodiments, where R 1 and R 2 in Formula I of the

benzodioxinone compound are both hydrogen, administering the benzodioxinone compound can deliver salicyclic acid to the subject. In some embodiments, the administering delivers an anti-inflammatory amount of salicyclic acid to the subject. In some embodiments, the administering provides an extended release of salicyclic acid. In some embodiments, the benzodioxinone compounds are administered to deliver salicyclic acid either locally and/or systemically. In some embodiments, the benzodioxinone compounds are administered topically. In some embodiments, the inflammation is associated with a skin disease, e.g., as described herein.

Definitions

[112] It is meant to be understood that proper valences are maintained for all moieties and

combinations thereof.

[113] It is also meant to be understood that a specific embodiment of a variable moiety herein may be the same or different as another specific embodiment having the same identifier.

[114] Suitable groups for R 1 , R 2 , and R 3 in compounds of Formula I (e.g., Formula 1-1, Formula la, Formula lb, Formula II-1 to Formula II -4, Formula III-1 to Formula III-10 or Formula IV- 1 to Formula IV-10) are independently selected. The described embodiments of the present invention may be combined. Such combination is contemplated and within the scope of the present invention. For example, it is contemplated that embodiments for any of R 1 , R 2 , and R 3 can be combined with embodiments defined for any other of R 1 , R 2 , and R 3 .

[115] The symbol, whether utilized as a bond or displayed perpendicular to a bond,

indicates the point at which the displayed moiety is attached to the remainder of the molecule.

[116] Throughout this application, various embodiments of this invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range, such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.

[117] As used herein, the term“benzodioxinone compound(s) of the present disclosure” refers to any of the compounds described herein according to Formula I (e.g., Formula la, Formula lb, Formula II- 1 to Formula II-4, Formula III-1 to Formula III- 10 or Formula IV- 1 to Formula IV-10, Examples 2-15), isotopically labeled compound(s) thereof (e.g., deuterated analogs thereof), possible stereoisomers thereof (including diastereoisomers, enantiomers, and racemic mixtures), tautomers thereof, conformational isomers thereof, and/or

pharmaceutically acceptable salts thereof (e.g., acid addition salt such as HC1 salt or base addition salt such as Na salt). Hydrates and solvates of the benzodioxinone compounds of the present disclosure are considered compositions of the present disclosure, wherein the compound(s) is in association with water or solvent, respectively.

[118] As used herein, the phrase“administration” of a compound,“administering” a compound, or other variants thereof means providing the compound or a prodrug of the compound to the individual in need of treatment.

[119] As used herein, the term "alkyl" as used by itself or as part of another group refers to a straight- or branched-chain aliphatic hydrocarbon containing one to twelve carbon atoms (i.e., C M 2 alkyl) or the number of carbon atoms designated (i.e., a Ci alkyl such as methyl, a C 2 alkyl such as ethyl, a C3 alkyl such as propyl or isopropyl, etc.). In one embodiment, the alkyl group is a straight chain Ci-io alkyl group. In another embodiment, the alkyl group is a branched chain C3-10 alkyl group. In another embodiment, the alkyl group is a straight chain C1-6 alkyl group. In another embodiment, the alkyl group is a branched chain C3-6 alkyl group. In another embodiment, the alkyl group is a straight chain C14 alkyl group. In another embodiment, the alkyl group is a branched chain C34 alkyl group. In another embodiment, the alkyl group is a straight or branched chain C34 alkyl group. Non-limiting exemplary Ci-io alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, iso-butyl, 3- pentyl, hexyl, heptyl, octyl, nonyl, decyl, and the like. Non-limiting exemplary C14 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, and iso-butyl.

[120] As used herein, the term "optionally substituted alkyl" as used by itself or as part of

another group means that the alkyl as defined above is either unsubstituted or substituted with one or more (e.g., one, two, or three) substituents each independently chosen from, e.g., halo (e.g., F), oxo, nitro, cyano, -OR a , -C0 2 R a , -0C0 2 R a , -0S0 2 NR b R c , -SCbR 3 , -OSCbR 3 , - 0S(0) n R e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , -C(=0)R d , - S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R g , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein. In one embodiment, the optionally substituted alkyl is substituted with three substituents, e.g., three fluorines. In one embodiment, the optionally substituted alkyl is substituted with one substituent. In one embodiment, the optionally substituted alkyl is substituted with two substituents.

[121] As used herein, each of R a , R d and R e at each occurrence is independently selected from hydrogen, C i-io alkyl, C i-io haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, 3-14 membered heterocyclyl, C 6- i4 aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 8 groups.

[122] As used herein, each of R b , R c and R f at each occurrence is independently selected from hydrogen, nitro, cyano, -OR aa , -CC R 33 , -S0 3 R aa , -NR bb R cc , -C(=0)NR bb R cc ,

-S0 2 NR bb R cc , -0C(=0)R dd , -C(=0)R dd , -S(0) n R ee , -C (=NR ff )NR bb R cc , C i-10 alkyl, C i-10 haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, 3-14 membered heterocyclyl, C 6- i4 aryl, and 5-14 membered heteroaryl, or R b and R c , or R f and one of R b and R c , are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 8 groups, wherein n is 0, 1, or 2, and R aa , R bb , R cc , R dd , R ee and R ff are defined herein. In some embodiments, at least one of R b and R c is chosen from hydrogen, Ci 10 alkyl, C i-10 haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 8 groups. In some embodiments, both R b and R c are independently chosen from hydrogen, C i-10 alkyl, C i-10 haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, 3-14 membered heterocyclyl, C 6-i4 aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 8 groups.

[123] As used herein, R 8 at each occurrence is independently selected from halo (e.g., F), nitro, cyano, -OR aa , -C0 2 R aa , -NR bb R cc , -C(=0)NR bb R cc , -SC> 2 NR bb R cc , -NR bb (SC> 2 NR bb R cc ), - OS0 2 NR bb R cc , -NR bb (S0 3 R aa ), -S0 3 R aa , -OS0 3 R aa . -NR bb (S(0) n R ee ), -0(S(0) n R ee ), - OC(=0)R dd , -OC0 2 R aa , -NR bb C0 2 R aa , -OC(=0)NR bb R cc , -NR bb (C(=0)R dd ), -C(=0)R dd , - S(0) n R ee , -C (=NR ff )NR bb R cc , -NR hh -C(=0)NR bb R cc , -NR hh -C(=NR ff )NR bb R cc , Ci 6 alkyl, Ci 6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-i o cycloalkyl, Ce io aryl, 3-10 membered heterocyclyl, 5-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 88 groups, or two geminal R 88 substituents can be joined to form =0 or =S, wherein n is 0, 1, or 2, and R 33 , R bb , R cc , R dd , R ee , R ff , R 88 and R 1 * are defined herein.

[124] As used herein, each of R 33 , R bb , R cc , R dd , R ee , R ff and R 1 * at each occurrence is

independently selected from hydrogen, Ci-6 alkyl, C i r, haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, 4-6 membered heterocyclyl, C 6-i o aryl, and 5-10 membered heteroaryl, or R bb and R cc , R ff and R 1 *, R ff and one of R bb and R cc , or R 1 * and one of R bb and R cc , are j oined to form a 4-6 membered heterocyclyl or 5-10 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 R 88 groups.

[125] As used herein, R 88 at each occurrence is independently selected from halo (e.g., F), nitro, cyano, hydroxy, -NH2, -N(H)(CI-6 alkyl), -N(CI-6 alkyl)( C1-6 alkyl), -N(H)(C3-6 cycloalkyl), -N(CI_6 alkyl)(C 3- 6 cycloalkyl), -N(C 3- 6 cycloalkyl)( C3-6 cycloalkyl), -O-C1-6 alkyl, C1-6 alkyl, C 1-6 haloalkyl, -O-C 1-6 haloalkyl, C 3-6 cycloalkyl optionally substituted with 1-5 substituents independently selected from halogen, C1-4 alkyl, and C3-6 cycloalkyl, -O-C3-6 cycloalkyl optionally substituted with 1-5 substituents independently selected from halogen, C1-4 alkyl, and C 3-6 cycloalkyl, 4-6 membered heterocyclyl optionally substituted with one or more (e.g., 1-3) substituents independently selected from halogen, oxo, C1-4 alkyl, and C 3-6 cycloalkyl, Ce-io aryl optionally substituted with 1-5 substituents independently selected from halogen, cyano, C14 alkyl, and C3-6 cycloalkyl, and 5-10 membered heteroaryl optionally substituted with 1-5 substituents independently selected from halogen, cyano, C M alkyl, and C 3-6 cycloalkyl, or two geminal R 88 substituents can be joined to form =0 or =S.

[126] As used herein, the term "cycloalkyl" as used by itself or as part of another group refers to saturated and partially unsaturated (containing one or two double bonds) cyclic aliphatic hydrocarbons containing one to three rings having from three to twelve carbon atoms (i.e., C3-12 cycloalkyl) or the number of carbons designated. In one embodiment, the cycloalkyl group has two rings. In one embodiment, the cycloalkyl group has one ring. In another embodiment, the cycloalkyl group is a C3-8 cycloalkyl group. In another embodiment, the cycloalkyl group is a C3-6 cycloalkyl group. Non-limiting exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbomyl, decalin, adamantyl, cyclopentenyl, and cyclohexenyl. [127] As used herein, the term "optionally substituted cycloalkyl" as used by itself or as part of another group means that the cycloalkyl as defined above is either unsubstituted or substituted with one or more (e.g., one, two, or three) substituents each independently chosen from, e.g., halo (e.g., F), oxo, nitro, cyano, -OR a , -CC R 3 , -OCC R 3 , -0S0 2 NR b R c , -S0 3 R a , - 0S0 3 R a , -0S(0)nR e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , - C(=0)R d , -S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R 8 , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein. In one embodiment, the optionally substituted cycloalkyl is substituted with three substituents. In one embodiment, the optionally substituted cycloalkyl is substituted with two substituents. In another embodiment, the optionally substituted cycloalkyl is substituted with one substituent.

[128] As used herein, the term "alkenyl" as used by itself or as part of another group refers to an alkyl group as defined above containing one, two or three carbon-to-carbon double bonds. In one embodiment, the alkenyl group is a C2-6 alkenyl group. In another embodiment, the alkenyl group is a C2-4 alkenyl group. Non-limiting exemplary alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, sec-butenyl, pentenyl, and hexenyl.

[129] As used herein, the term "optionally substituted alkenyl" as used herein by itself or as part of another group means the alkenyl as defined above is either unsubstituted or substituted with one or more (e.g., one, two, or three) substituents each independently chosen from, e.g., halo (e.g., F), oxo, nitro, cyano, -OR a , -C0 2 R a , -0C0 2 R a , -0S0 2 NR b R c , -S0 3 R a ,

-0S0 3 R a , -0S(0) n R e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , -C(=0)R d , -S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R 8 , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein.

[130] As used herein, the term "alkynyl" as used by itself or as part of another group refers to an alkyl group as defined above containing one to three carbon-to-carbon triple bonds. In one embodiment, the alkynyl has one carbon-carbon triple bond. In one embodiment, the alkynyl group is a C2-6 alkynyl group. In another embodiment, the alkynyl group is a C24 alkynyl group. Non-limiting exemplary alkynyl groups include ethynyl, propynyl, butynyl, 2- butynyl, pentynyl, and hexynyl groups. [131] As used herein, the term "optionally substituted alkynyl" as used herein by itself or as part of another group means the alkynyl as defined above is either unsubstituted or substituted with one or more (e.g., one, two, or three) substituents each independently chosen from, e.g., halo (e.g., F), oxo, nitro, cyano, -OR a , -C0 2 R a , -0C0 2 R a , -0S0 2 NR b R c , -S0 3 R a ,

-0S0 3 R a , -0S(0)nR e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , -C(=0)R d , -S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R 8 , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein.

[132] As used herein, the term "haloalkyl" as used by itself or as part of another group refers to an alkyl substituted with one or more fluorine, chlorine, bromine and/or iodine atoms. In preferred embodiments, the haloalkyl is an alkyl group substituted with one, two, or three fluorine atoms. In one embodiment, the haloalkyl group is a Ci-io haloalkyl group. In one embodiment, the haloalkyl group is a Ci- 6 haloalkyl group. In one embodiment, the haloalkyl group is a C i4 haloalkyl group.

[133] As used herein, the term "alkoxy" as used by itself or as part of another group refers to a radical of the formula -OR al , wherein R al is an alkyl. As used herein, the term“optionally substituted alkoxy” refers to a radical of the formula -OR al , wherein R al is an optionally substituted alkyl.

[134] As used herein, the term "cycloalkoxy" as used by itself or as part of another group refers to a radical of the formula -OR al , wherein R al is a cycloalkyl. As used herein, the term “optionally substituted cycloalkoxy” refers to a radical of the formula -OR al , wherein R al is an optionally substituted cycloalkyl.

[135] As used herein, the term "aryl" as used by itself or as part of another group refers to a monocyclic, bicyclic or tricyclic aromatic ring system having from six to fourteen carbon atoms (i.e., C 6-i 4 aryl). In one embodiment, the aryl group is a C 6-i 2 aryl. In one

embodiment, the aryl group is chosen from phenyl and naphthyl. In one embodiment, the aryl group is naphthyl.

[136] As used herein, the term "optionally substituted aryl" as used by itself or as part of

another group means that the aryl as defined above is either unsubstituted or substituted with one to five substituents each independently chosen from, e.g., halo (e.g., F), nitro, cyano, - OR a , -C0 2 R a , -0C0 2 R a , -0S0 2 NR b R c , -S0 3 R a , -0S0 3 R a , -0S(0) n R e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , -C(=0)R d , -S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R g , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein; or two of the substituents are joined to form an optionally substituted cycloalkyl or optionally substituted heterocyclyl ring fused to the aryl.

[137] As used herein, the term "heteroaryl" or "heteroaromatic" refers to monocyclic, bicyclic or tricyclic aromatic ring systems having 5 to 14 ring atoms (i.e., a 5- to l4-membered heteroaryl) and 1, 2, 3, or 4 heteroatoms independently chosen from oxygen, nitrogen and sulfur. In one embodiment, the heteroaryl has three heteroatoms, e.g., three nitrogen atoms.

In another embodiment, the heteroaryl has two heteroatoms, e.g., two nitrogen atoms, one nitrogen and one oxygen, or one nitrogen and one sulfur. In another embodiment, the heteroaryl has one heteroatom, e.g., one nitrogen. In one embodiment, the heteroaryl has 5 ring atoms, e.g., pyrazolyl. In another embodiment, the heteroaryl has 6 ring atoms, e.g., pyridyl. Non-limiting exemplary heteroaryl groups include thienyl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl, benzofuryl, pyranyl, isobenzofuranyl,

benzooxazonyl, chromenyl, xanthenyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isoindolyl, 3H- indolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, cinnolinyl, quinazolinyl, pteridinyl, 4aH- carbazolyl, carbazolyl, b-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, thiazolyl, isothiazolyl, phenothiazolyl, isoxazolyl, furazanyl, and phenoxazinyl.

In one embodiment, the heteroaryl is chosen from thienyl (e.g., thien-2-yl and thien-3-yl), furyl (e.g., 2-furyl and 3-furyl), pyrrolyl (e.g., lH-pyrrol-2-yl and lH-pyrrol-3-yl), imidazolyl (e.g., 2H-imidazol-2-yl and 2H-imidazol-4-yl), pyrazolyl (e.g., lH-pyrazol-3-yl, 1H- pyrazol- 4-yl, and lH-pyrazol-5-yl), pyridyl (e.g., pyridin-2-yl, pyridin-3-yl, and pyridin- 4-yl), pyrimidinyl (e.g., pyrimidin-2-yl, pyrimidin-4-yl, and pyrimidin-5-yl), thiazolyl (e.g., thiazol- 2-yl, thiazol-4-yl, and thiazol-5-yl), isothiazolyl (e.g., isothiazol-3-yl, isothiazol-4-yl, and isothiazol-5-yl), oxazolyl (e.g., oxazol-2-yl, oxazol-4-yl, and oxazol- 5-yl) and isoxazolyl (e.g., isoxazol-3-yl, isoxazol-4-yl, and isoxazol-5-yl). In one embodiment, the heteroaryl is a bicyclic heteroaryl having 8 to 10 ring atoms, e.g., a bicyclic heteroaryl having 1, 2, or 3 nitrogen ring atoms, such as quinolyl. As used herein, the term "heteroaryl" is also meant to include possible N-oxides. [138] As used herein, the term "optionally substituted heteroaryl" as used by itself or as part of another group means that the heteroaryl as defined above is either unsubstituted or substituted with one or more (e.g., 1, 2, 3, 4, or 5) substituents each independently chosen from, e.g., halo (e.g., F), nitro, cyano, -OR a , -C0 2 R a , -0C0 2 R a , -0S0 2 NR b R c , -SCbR 3 , -OSCbR 3 , - 0S(0)nR e , -NR b R c , -C(=0)NR b R c , -0C(=0)NR b R c , -S0 2 NR b R c , -0C(=0)R d , -C(=0)R d , - S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R 8 , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein; or two of the substituents are joined to form an optionally substituted cycloalkyl or optionally substituted heterocyclyl ring fused to the heteroaryl.

[139] As used herein, the term "heterocycle" or "heterocyclyl" as used by itself or as part of another group refers to saturated and partially unsaturated (e.g., containing one or two double bonds) cyclic groups containing one, two, or three rings having from three to fourteen ring members (i.e., a 3- to l4-membered heterocycle) and at least one heteroatom. Each heteroatom is independently selected from the group consisting of oxygen, sulfur, including sulfoxide and sulfone, and/or nitrogen atoms, which can be quatemized. The term

"heterocyclyl" is meant to include cyclic ureido groups such as imidazolidinyl-2-one, cyclic amide groups such as b-lactam, g-lactam, d-lactam and e-lactam, and cyclic carbamate groups such as oxazolidinyl-2-one. In one embodiment, the heterocyclyl group is a 4-, 5-, 6-, 7- or 8- membered cyclic group containing one ring and one or two oxygen and/or nitrogen atoms. In one embodiment, the heterocyclyl group is a 5- or 6-membered cyclic group containing one ring and one or two nitrogen atoms. In one embodiment, the heterocyclyl group is an 8-, 9-, 10-, 11-, or l2-membered cyclic group containing two rings and one or two nitrogen atoms. The heterocyclyl can be optionally linked to the rest of the molecule through a carbon or nitrogen atom.

[140] As used herein, the term "optionally substituted heterocyclyl" as used herein by itself or part of another group means the heterocyclyl as defined above is either unsubstituted or substituted with one or more (e.g., 1, 2, 3, 4, or 5) substituents each independently chosen from, e.g., halo (e.g., F), oxo, nitro, cyano, -OR a , -C0 2 R a , -OC0 2 R a , -OS0 2 NR b R c , -SChR 3 , - OS0 3 R a , -OS(0) n R e , -NR b R c , -C(=0)NR b R c , -OC(=0)NR b R c , -S0 2 NR b R c , -OC(=0)R d , - C(=0)R d , -S(0) n R e , -C(=NR f )NR b R c , alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl, wherein each alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocyclyl is further optionally substituted with 1-5 R 8 , wherein n is 0, 1, or 2, and R a , R b , R c , R d , R e , R f and R 8 are defined herein; or two of the substituents are joined to form an optionally substituted aryl or optionally substituted heteroaryl ring fused to the heterocyclyl. Substitution may occur on any available carbon or nitrogen atom, and may form a spirocycle.

[141] As used herein, the term "alkanoyl" as used by itself or as part of another group refers to a radical of the formula -C(=0)-R dl , wherein R dl is an alkyl group. As used herein, the term "optionally substituted alkanoyl" as used by itself or as part of another group refers to - C(=0)-R dl , wherein R dl is an optionally substituted alkyl group.

[142] As used herein, the term "cycloalkanoyl" as used by itself or as part of another group refers to a radical of the formula -C(=0)R dl , wherein R dl is a cycloalkyl group. As used herein, the term "optionally substituted cycloalkanoyl" as used by itself or as part of another group refers to -C(=0)R dl , wherein R dl is an optionally substituted cycloalkyl group.

[143] As used herein, the term "salt" includes both internal salt and external salt. In some

embodiments, the salt is an internal salt, i.e., a zwitterion structure. In some embodiments, the salt is an external salt. In some embodiments, the external salt is a pharmaceutically acceptable salt having a suitable counter ion. Suitable counterions for pharmaceutical use are known in the art.

[144] As used herein, the terms "treat," "treating," "treatment," and the like refer to eliminating, reducing, or ameliorating a disease or condition, and/or symptoms associated therewith. Although not precluded, treating a disease or condition does not require that the disease, condition, or symptoms associated therewith be completely eliminated. As used herein, the terms "treat," "treating," "treatment," and the like may include "prophylactic treatment," which refers to reducing the probability of redeveloping a disease or condition, or of a recurrence of a previously-controlled disease or condition, in a subject who does not have, but is at risk of or is susceptible to, redeveloping a disease or condition or a recurrence of the disease or condition. The term "treat" and synonyms contemplate administering a therapeutically effective amount of a compound described herein to a subject in need of such treatment.

[145] The term“inhibition”,“inhibiting”,“inhibit,” or“inhibitor” refer to the ability of a

compound to reduce, slow, halt or prevent activity of a particular biological process ( e.g . , activity of ACC enzyme in a cell relative to vehicle). [146] The term“subject” (alternatively referred to herein as“patient”) as used herein, refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.

Examples

[147] The various starting materials, intermediates, and compounds of the preferred

embodiments can be isolated and purified where appropriate using conventional techniques such as precipitation, filtration, crystallization, evaporation, distillation, and chromatography. Characterization of these compounds can be performed using conventional methods such as by melting point, mass spectrum, nuclear magnetic resonance, and various other

spectroscopic analyses. Exemplary embodiments of steps for performing the synthesis of products described herein are described in greater detail infra.

Example 1. General Synthesis of l'-aryloyl or T-heteraryloyl-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[148] Step 1. To a solution of 5-bromo-2-hydroxybenzyl alcohol (102 g, 0.5mol) and N-Boc-4- piperidone (140 g, 0.7mmol) in chloroform (700 mL) was added p-toluenesulphonic acid (10 g). The mixture was heated at reflux for 18 h, during which time condensate (5x 50 mL) was removed and replaced with equal volume of fresh dry chloroform. The solvent was removed in vacuo. The residue was dissolved in ethyl acetate (500 mL) and washed with a 2M sodium hydroxide solution (100 mL) and brine (300 mL). The organic layer was dried with magnesium sulfate and the solvent removed in vacuo. The residue was purified by column chromatography (silica gel, 20% EtOAc (Ethyl Acetate) in petrol ether to 30% EtOAc in petrol ether) to give the title compound A3 (115 g, 60% yield).

[149] Step 2. tert-butyl 6-bromo-rH,4H-spiro[l,3-benzodioxine-2,4'-piperidine]-r- carboxylate(38.4g, 0.1 mol) was mixed with MeOH(l50ml), Triethylamine(l5g, 0. l5mol) and Pd(PPh 3 ) 4 (l.0g) in an autoclave(lL), the autoclave was purged with CO three times, then filled with CO under lOOPsi and heated at 100 °C for 5h. After the solvent was evaporated, the residue was mixed with MTBE(methyl tert-butyl ether, 200ml) with stirring, followed by filtration through a silica pad. The MTBE solution was evaporated to give pure title compound A4(34g, 95% yield).

[150] Step 3. To a solution of MeMgCl(l00ml, 3M)in THF at 0 °C was added dropwise a

solution of the ester A4 (36.3g, 0. lmol) in THF(200ml) with stirring, the reaction was then stirred at room temperature for another hour. The reaction mixture was poured into a cooled 2N HC1 (500ml) solution, and extracted with Ethyl Acetate(2xl00ml), the combined organic phase was washed with Brine(lOOml) and dried over MgSO4(l0g). After evaporation of the solvent, the residue was purified by column chromatography (silica gel, 10% EtOAc in petrol ether to 20% EtOAc in petrol ether) to give the title compound A5 (36 g, 98% yield).

[151] Step 4. KMn0 4 (l4. lg, lOOmmol) was added to a solution of l8.2g(50mmol) of the

material A5 in DCM (dichloromethane, 200ml) containing 2g of tertabutylammonium chloride at room temperature, the resulting suspension was stirred for overnight. Celite (20g) was added to the reaction, and the resulting mixture was filtered through a Celite pad, washed with another 200ml of DCM. The combined DCM solution was washed by water and dried over MgSCriOOg). After evaporation of the solvent, the residue was used for next step without purification (l8.0g as crude A6).

[152] Step 5. 16. Og of the crude material A6 from step 4 was dissolved in MeOH(lOOml), 0.5g of Pd/C(5%) was added, and the mixture was charged with Hydrogen at 30Psi at room temperature. The reaction was done after 6 hr, as indicated by HPLC. After removal of the catalyst by filtration, the solution was evaporated under vacuum to give the desired product A7 (l5.0g as crude).

[153] Step 6. 15. Og of the crude material A7 from step 5 was dissolved in EtOAc(50ml) and then treated with 7N of a HCl-solution in EtOAc (50ml) at 0 °C. After stirring for 6 hr, the resulting suspension was filtered to collect the white solid as pure product A8 (10.5g as HC1 salt).

[154] Step 7. Compound A8 can be treated with 1 eq. of R 3 COOH in the presence of DIEA (2.5 eq.) and HATU (1.1 eq.) in DMF (0.5 mL for 0.1 mmol of A8) at room temperature for 4 hr. The reaction mixture was diluted with ethyl acetate and washed with 1N HC1, saturated NaHC03, and brine. The crude product was purified by preparative TLC eluting with appropriate solvent to give the desired product A9. Example 2. Synthesis of r-(4,8-dimethoxyquinobne-2-carbonyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[155] Example 2 was prepared according to the general procedure described in Example 1 using ethyl acetate as solvent in prepTLC. 'HNMR (CDCb) d: 7.85 (br s, 1H); 7.80 (d, 1H); 7.51 (d, 1H); 7.46 (m, 1H); 7.22 (s, 1H); 7.11 (d, 1H); 6.98 (d, 1H); 4.12 (s, 3H); 4.05 (s, 3H); 3.9 (m, 4H); 2.94 (pent, 1H); 2.3 (br, 4H); 1.27 (d, 6H). LC-MS: 477.10 (M+H).

Example 3. Synthesis of l'-(4,8-dimethoxy-2-naphthoyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[156] Example 3 was prepared according to the general procedure described in Example 1 using 1:20 ethyl acetate/dichloromethane as solvent in prepTLC. Ή NMR (CDCb) d: 7.91 (s,

1H); 7.84 (m, 2H); 7.5 (d, 1H); 7.45 (t, 1H); 6.98 (d, 1H); 6.92 (s, 1H); 6.90 (d, 1H); 4.04 (s, 3H); 4.00 (s, 3H); 3.7-4.1 (br, 4H); 2.94 (pent, 1H); 2.2 (br, 4H); 1.26 (d, 6H). LC-MS:

476.00 (M+H).

Example 4. Synthesis of 6-isopropyl-r-(2-methyl-lH-benzo[d]imidazole-6-carbonyl)- 4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[157] Example 4 was prepared according to the general procedure described in Example 1 using 1: 10 methanol/ethyl acetate as solvent in prepTLC. 1 HNMR (CDCb) d: 7.83 (s, 1H); 7.60 (s, 1H); 7.48 (m, 2H); 7.30 (s, 1H); 6.98 (d, 1H); 4.0-3.6 (br, 4H); 2.94 (pent, 1H); 2.59 (s, 3H); 2.25-2.10 (br, 4H); 1.27 (d, 6H). LC-MS: 420.2 (M+H).

Example 5. Synthesis of l'-(lH-indazole-5-carbonyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[158] Example 5 was prepared according to the general procedure described in Example 1 using 1: 1 ethyl acetate/dichloromethane as solvent in prepTLC. LC-MS: 406.2 (M+H)

Example 6. Synthesis of l'-(lH-indazole-6-carbonyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[159] Example 6 was prepared according to the general procedure described in Example 1 using l;l ethyl acetate/dichloromethane as solvent in prepTLC. 1 HNMR (CDCb) d: 8.15 (s, 1H); 7.84 (s, 1H); 7.81 (d, 1H); 7.62 (s, 1H); 7.47 (d, 1H); 7.21 (d, 1H); 6.98 (d, 1H); 3.9 (br, 2H); 3.65 (br, 2H); 2.95 (pent, 1H); 2.2 (br, 4H); 1.24 (d, 6H).

Example 7. Synthesis of 6-isopropyl-r-(lH-pyrrolo[3,2-b]pyridine-2-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[160] Example 7 was prepared according to the general procedure described in Example 1 using 1: 1 ethyl acetate/dichloromethane as solvent in prepTLC. 1 HNMR (CDCb) d: 9.58 (br s, 1H); 8.58 (m, 1H); 7.88 (s, 1H); 7.80 (d, 1H); 7.52 (d, 1H); 7.27 (m, 1H); 7.0 (m, 2H); 4.2- 4.0 (br, 4H); 2.98 (pent, 1H); 2.28 (br, 4H); 1.3 (d, 6H). LC-MS: 406.2 (M+H).

Example 8. Synthesis of 6-isopropyl- l'-(6-methoxy quinoline-3 -carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[161] Example 8 was prepared according to the general procedure described in Example 1 using ethyl acetate as solvent in prepTLC. 1 HNMR (CDCb) d: 8.82 (s, 1H); 8.19 (s, 1H); 8.04 (d, 1H); 7.84 (s, 1H); 7.5 (m, 2H); 7.14 (s, 1H); 6.98 (d, 1H); 3.99 (s, 3H); 3.98 (br, 2H); 3.75 (br, 2H); 2.96 (pent, 1H); 2.3-2.1 (br, 4H); 1.25 (d, 6H); LC-MS: 447 (M+H).

Example 9. Synthesis of r-(2-ethyl-lH-benzo[d]imidazole-6-carbonyl)-6-isopropyl-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[162] Example 9 was prepared according to the general procedure described in Example 1 using 1:20 methanol/ethyl acetate as solvent in prepTLC. 1 HNMR (CDCb) d: 7.84 (s, 1H); 7.7.63 (s, 1H); 7.55-7.45 (m, 3H); 7.28 (m, 2H); 6.98 (d, 1H); 3.8 (br, 4H); 2.98 (q, 2H); 2.997 (pent, 1H); 2.2 (br, 4H); 1.43 (t, 3H); 1.27 (d, 6H). LC-MS: 434 (M+H).

Example 10. Synthesis of 6-isopropyl-l'-(3-methyl-2-oxo-2,3-dihydro-lH- benzo[d]imidazole-5-carbonyl)-4H-spiro[benzo[d][l,3]dioxine- 2,4'-piperidin]-4-one

[163] Example 10 was prepared according to the general procedure described in Example 1 using 1:20 methanol/ethyl acetate as solvent in prepTLC. 'HNMR (CDCT) d: 9.36 (s, 1H); 7.83 (s, 1H); 7.46 (d, 1H); 7.18-7.08 (m, 3H); 6.96 (d, 1H); 3.9-3.75 (br, 4H); 3.46 (s, 3H); 2.96 (pent, 1H); 2.2 (br, 4H); 1.28 (d, 6H). LC-MS: 436 (M+H).

Example 11. Synthesis of T-(4,8-dimethoxy-2-naphthoyl)-6-((l-methyl-lH-pyrazol-5- yl)amino)-4H-spiro[benzo[d] [1,3] dioxine-2,4'-piperidin] -4-one

[164] Step 1

[165] To a solution of the starting material A3 (19.2 g, 50 mmole) in DCM (200 mL) containing tertabutylamonium chloride (2 g) was added KMnCh (14.1 g, 100 mmole) at room temperature. The resulting suspension was stirred at room temperature for overnight. Celite (20 g) was added, and this mixture was filtered through a Celite pad, the filter cake was washed with DCM, the combined DCM solution was washed with water and dried over MgSCri. Evaporation of the solvent afforded the crude product B1 (17.0 g).

[166] Step 2:

[167] Step 2. Starting material B1 (4.0g, lOmmol) was mixed with KOAc(6g), 0.2g of

Pd(dppf)Ch, and Bis(pinacolato)diboron(6g) in dioxane(20.0ml) under N 2 . The resulting mixture was heated at 90 °C for 8h. The resulting mixture was treated with water and extracted with EtOAc(2x50ml), the combined EtOAc solution was washed by brine(50ml) and dried over MgSC . Evaporation of the solvent produced a crude product B2(5.0g) as a solid.

[168] Step 3:

[169] Step3. The pinacol boronate B2 (5.0g, lOmmol) was mixed with NaI0 4 (6g) in

acetone(30ml) and water(30ml). The mixture was stirred at room temperature for 4h. The resulting mixture was extracted with EtOAc (2x50ml), the combined EtOAc solution was washed with brine(50ml) and dried over MgSCri. Evaporation of solvent afforded crude product B3 (3.0g) as a solid.

[170] Step 4:

[171] Step 4. The boronic acid B3 (2.9 g, 7 mmole) was mixed with 5-amino-l-methyl-lH- pyrazole (l.2g), Cu(OAc) 2 (2.0g) and Triethylamine(4ml) in DCM(20ml) at room temperature and was stirred for 4h. Water was then added and the resulting mixture was extracted with EtOAc(2x30ml), the combined EtOAc solution was washed by 1N HCl(50ml) and dried over MgS0 4 . After evaporation of the solvent, the residue was purified through flash column chromatography to give the title compound B5 (0. lOg as solid). 1 HNMR (CDC13) d: 7.48 (d, 1H); 7.28 (d, 1H); 6.96 (dd, 1H); 6.90 (d, 1H); 6.02 (d, 1H); 5.40 (s,

1H); 3.73 (s, 3H); 3.62 (m, 2H); 3.53 (m, 2H); 2.05 (m, 4H); 1.46 (s, 9H).

[172] Step 5:

[173] Step 4. Starting material B5 (obtained from step 4) was dissolved in EtOAc and then treated with 7N of HC1 solution in EtOAc at 0 °C. After 6h with stirring, the suspension was filtered to collect the white solid as HC1 salt B6.

[174] Step 6:

[175] Piperidine B6 from step 5 was then coupled with the carboxylic acid B7 following

conditions similar to step 7 described in Example 1. The title compound, compound B8, was purified using ethyl acetate as solvent in prepTLC. 'HNMR (CDCb) d: 7.90 (s, 1H); 7.84 (d, 1H); 7.5 (s, 1H); 7.44 (t, 1H); 7.32 (s, 1H); 7.0-6.9 (m, 4H); 6.05 (s, 1H); 5.4 (s, 1H); 4.03 (s, 3H); 4.01 (s, 3H); 3.75 (s, 3H); 4-3.7 (br, 4H); 2.2 (br, 4H); LC-MS: 529 (M+H).

Example 12. Synthesis of r-(4,8-dimethoxyquinoline-2-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one

[176] Example 12 was prepared according to the general amide coupling procedure described in Example 1. Starting material 4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one hydrochloride was prepared according to literature published procedure. Preparatory TLC using 1:20 MeOEPDCM as solvent provided r-(4,8-dimethoxyquinoline-2-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one as a white powder. 1 HNMR (CDCb) d:

8.00 (dd, 1H); 7.80 (dd, 1H); 7.62 (td, 1H); 7.50 (t, 1H); 7.23 (s, 1H); 7.18 (t, 1H); 7.11 (d, 1H); 7.06 (d, 1H); 4.12 (s, 3H); 4.07 (s, 3H); 4.0 (m, 4H), 2.3 (m, 4H). Example 13. Synthesis oftert-butyl (3-(4-oxo-4H-spiro[benzo[d][l,3]dioxine-2,4'- piperidin]-r-ylcarbonyl)benzo[b]thiophen-2-yl)carbamate

[177] Example 13 was prepared in the same method as Example 12. Preparatory TLC using 1:3 ethyl acetate: hexane as solvent provide Example 3 (tert-butyl (3-(4-oxo-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r-ylcarbonyl)benz o[b]thiophen-2-yl)carbamate) as a white powder after lyophilization. 1 ElNMR. (CDCb) d: 9.10 (d, 1H); 7.00 (d, 1H); 7.76 (d, 1H); 7.62 (m, 1H); 7.52 (d, 1H); 7.40 (m, 1H); 7.28 (m, 1H); 7.18 (m, 1H); 7.06 (dd, 1H); 3.6-4.0 (m, 4H); 2.0-2.4 (m, 4H); 1.6 (s, 9H).

Example 14 and 15. Synthesis of l'-(2-aminobenzo[b]thiophene-3-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one, and l-ethyl-3-(3-(4-oxo-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r-ylcarbonyl)benz o[b]thiophen-2-yl)urea

[178] Step 1. tert-Butyl (3-(4-oxo-4H-spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r- ylcarbonyl)benzo[b]-thiophen-2-yl)carbamate (40 mg) was treated with HC1/EA solution (4N, 4 mL) for 1.5 hr at room temperature. HPLC indicated clean conversion of starting material to Example 14. Evaporation off excess reagent and solvent, followed by lyophilzation, produced 1 '-(2-aminobenzo[b]thiophene-3-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one hydrochloride (Example 14) as a white powder (35 mg). A small portion of which was dissolved in DMSO as 10 uM solution for biological assay.

[179] Step 2. A solution of l'-(2-aminobenzo[b]thiophene-3-carbonyl)-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-4-one hydrochloride (32 mg), DIEA (13 uL) in DCM (300 uL) was added to a solution of triphosgen (8 mg), DIEA (28 uL) in DCM (150 uL). After 30 min stirring at room temperature, a solution of EtNH2 HC1 (6 mg), DIEA (15 uL) in DCM (50 uL) and acetonitrile (60 uL) was added to the reaction mixture. The mixture was stirred for 4 hr, and diluted with ethyl acetate (4 mL), washed with 0.5N HC1, brine. Preparatory TLC (1: 1 ethyl acetate: hexane) yielded l-ethyl-3-(3-(4-oxo-4H- spiro[benzo[d][l,3]dioxine-2,4'-piperidin]-r-ylcarbonyl)benz o[b]thiophen-2-yl)urea

(Example 15) as a white powder after lyophilization. 1 HNMR (CDCh) d: 9.44 (d, 1H); 7.98 (d, 1H); 7.74 (d, 1H); 7.58 (m, 1H); 7.46 (m, 1H); 7.36 (m, 1H); 7.15-7.27 (m, 2H); 6.7-7.2 (m, 1H); 5.32 (br, 1H); 3.6-4.0 (m, 4H); 3.2 (br, 2H); 2.0-2.35 (m, 4H); 1.06 (m, 3H).

Biological Example 1. Human ACC enzymatic activity assay

[180] Human ACC enzymatic activity assays were performed by using purified human ACC1 and ACC2 proteins (BPS Bioscience). ACC1 and ACC2 proteins were diluted in 24.5 pL reaction buffer (50 mM HEPES pH 7.5, 10 mM MgCh, 2.0 mM DTT, 4.0 mM ATP, 15 mM 1 3 C-NaHC0 3 , 0.75 mg/mL BSA, 0.5 mM sodium citrate), and final concentrations of ACC1 and ACC2 were 0.4 pg/mL and 0.48 pg/mL. For each reaction, 0.5 pL of serial diluted compounds were prepared in DMSO in duplicate or triplicate and mixed with the above 24.5 pL reaction mixtures with ACC proteins at 37°C for 10 min. Meanwhile, 25 pL of reaction buffer plus acetyl-CoA (final concentration: 5 pM) was prepared freshly and added to the above 25 pL mixture to initiate the reaction. After incubation at 37°C for 1-2 hours, 100 pl ice-cold 10% TCA with malonyl- 13 C 3 - CoA (final concentration: 40 nM, Sigma-Aldrich) was added to terminate the reaction. For LC-MS/MS analysis, 300 pL quenching buffer (acetonitrile:methanol = 1 : 1, 20 pM carbutamide as internal standard) was added to each tube and mixed well. Samples were then centrifuged at 14,000 rpm for 10 min (Eppendorf) and supernatants were loaded onto LC-MS/MS for quantitation of malonyl- 13 C-CoA (Gao et ak, Journal of chromatography B, Analytical technologies in the biomedical and life sciences 853, 303-313 (2007)).

[181] Examples 12-15 showed IC50 in the range of 0.1-10 uM in this assay.

Biological Example 2. Quantitation of malonyl-CoA by LC-MS/MS

[182] Shimadzu LC system was used for sample analysis consisting of system controller

(Shimadzu CBM-20A), autosampler (Shimadzu SIL-20A), pump A and B (Shimadzu LC- 20AD), column oven (Shimadzu CTO-20 A), and analytical column (Phenomenex Gemini 5u

C18 11 A, 100 X2 mm). LC was running at 200 pL/min in the binary gradient mode with mobile phase A (5 mM Ammonium Acetate and 5 mM DMBA in water (1,3- Dimethylbutylamine)) and B (methanol). The samples were analyzed on an AB Sciex QTrap4000 LC-MS/MS triple quadrupole mass spectrometer (Applied Biosystems).

Malonyl-CoA and malonyl- 13 C-CoA were monitored in positive ion mode following MRM transitions at 854.2/347.1 and 855.1/246.1, respectively. The product and internal standard were coeluted to the mass spectrometer in 3 s with 50% LLO, 25% MeCN, 25% acetone, and 5 mM ammonium acetate. Ion chromatograms were integrated using the Analyst 1.5.2 software. Peak area ratios were compared against a standard curve and final cellular concentration of malonyl-CoA or production rate of malonyl- 13 C-CoA was calculated as pmoles/mg protein per minute.

Biological Example 3. Effect of ACC inhibitors on cellular levels of malonyl-CoA in human sebocytes

[183] Human sebocytes (Celprogen) were grown in growth media (DMEM with 4.5 g/L

glucose, 0.584 g/L L-glutamine (Sigma), 100 units/mL of penicillin and 100 pg/mL of streptomycin (Gibco), 10% fetal bovine serum (Gibco)) at 37°C in a humidified water jacketed incubator (Forma Scientific) supplemented with 5% CO2 till 100% confluency.

Cells grown in 152 cm 2 cell culture dish (Coming) were trypsinized with 0.25% trypsin- EDTA (Gibco), centrifuged at 800 rpm for 10 min (Eppendorf) and resuspended in 4 mL growth media. To 1 pL serial diluted compounds in DMSO, 99 pL cells (around 0.5 million cells suspended in growth media) were added and mixed well followed by incubation in cell culture incubator for 30 min. Cells were then centrifuged at 800 rpm for 10 min and 70 pL 10% trichloroacetic acid was added to each well to lyse the cells and precipitate proteins. Cellular levels of malonyl-CoA were quantitated by LC-MS/MS and effect of compounds on levels of malonyl-CoA in sebocytes was calculated by % of DMSO vehicle control.

[184] Examples 2 to 11 showed IC50 in the range of 0.01- 1 uM in this assay.

Biological Example 4. IHC staining for human skin tissue

[185] Formalin-fixed, Paraffin-embedded slides of skin tissues from humans (Amsbio and ProSci) were baked at 65°C for 30 min and deparaffmized / hydrated by incubating in three washes of xylene for 5 min each, two washes of 100% ethanol for 10 min each, two washes of 95% ethanol for 10 min each, and finally two washes in dEbO for 5 min each. Slides were then boiled in IX antigen unmasking solution (Vector Laboratories) for 10 min. After washing in dFEO. slides were stained with rabbit IgG isotype control antibody or rabbit monoclonal anti-ACC (18 pg/mL, 1:50 dilution, Cell Signaling Technology) at room temperature for 1 hour or at 4°C overnight. Sections were then washed with wash buffer three times for 5 min each, Rabbit on Rodent HRP-Polymer (BioCare Medical) and DAB Quanto Chromagen and Substrate (Thermo Fisher Scientific) were used to develop the dark brown color to reveal the expression of ACC protein. Slides were counterstained with hematoxylin (RICCA Chemical), dehydrated, and sealed with coverslips by using

VectaMount (Vector Laboratories). Pictures were taken by using Zeiss Primo Star and Nikon D800.

[186] As shown in Fig. 1, in the above study of human skin, ACC proteins are abundantly and specifically expressed in sebaceous glands. This represents the first report on the localization of ACC proteins in human skin structure and it suggests that delivering ACC inhibitors to sebocytes, such as by topical administration of ACC inhibitors, will specifically inhibit ACCs in sebaceous gland, thus provides a powerful method of suppressing lipid accumulation and progression of diseases related to overproduction of lipids in sebocytes.

[187] On the other hand, as shown in Fig. 2, ACC proteins are not abundantly expressed in keratinocytes of human normal skin.

[188] Fig. 3 shows images of Immunohistochemical (IHC) staining of human in situ squamous cell carcinoma (indicated by arrows) with adjacent skin tissue including sebaceous gland. As shown in Fig. 3, tumor cells in the squamous cell carcinoma, sebaceous gland and epidermal squamous cells are stained with comparable density of dark color, indicating abundant expression of ACC proteins.

[189] Fig. 4 shows images of Immunohistochemical (IHC) staining of human seborrheic

keratosis and actinic keratosis. As can be seen, keratin layer is apparent in this disease and keratinocytes next to the keratin layer are stained with dark color (indicated by arrows), indicating abundant expression of ACC proteins in keratinocytes in skin disease of keratosis.

[190] With respect to aspects of the invention described as a genus, all individual species are individually considered separate aspects of the invention. If aspects of the invention are described as "comprising" a feature, embodiments also are contemplated "consisting of or "consisting essentially of’ the feature.

[191] The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the skill of the art, readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance.

[192] The breadth and scope of the present invention should not be limited by any of the above- described exemplary embodiments.

[193] All of the various aspects, embodiments, and options described herein can be combined in any and all variations.

[194] All publications, patents, and patent applications mentioned in this specification are

herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent that any meaning or definition of a term in this document conflicts with any meaning or definition of the same term in a document incorporated by reference, the meaning or definition assigned to that term in this document shall govern.