Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OPSIN-BINDING LIGANDS, COMPOSITIONS AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2011/155983
Kind Code:
A1
Abstract:
Compounds are disclosed that are useful for treating ophthalmic conditions related to mislocalization of opsin proteins, the misfolding of mutant opsin proteins and the production of toxic visual cycle products that accumulate in the eye. Compositions of these compounds, either alone or in combination with other therapeutic agents, are also disclosed, along with thereapeutic methods of using such compositions. Methods of synthesizing such compounds are also described.

Inventors:
GARVEY DAVID S (US)
LAROSA GREGORY J (US)
GREENWOOD JEREMY R (US)
FRYE LEAH L (US)
Application Number:
PCT/US2011/001028
Publication Date:
December 15, 2011
Filing Date:
June 07, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BIKAM PHARMACEUTICALS INC (US)
GARVEY DAVID S (US)
LAROSA GREGORY J (US)
GREENWOOD JEREMY R (US)
FRYE LEAH L (US)
International Classes:
A61K31/015; A01N43/38
Domestic Patent References:
WO2006058088A22006-06-01
WO2008013984A22008-01-31
Other References:
SCHEERER ET AL.: "Crystal structure of opsin in its G-protein-interacting conformation", NATURE, vol. 455, September 2008 (2008-09-01), pages 497 - 503
Attorney, Agent or Firm:
GRANT, Alan, J. et al. (Byrne Cecchi, Olstein, Brody & Agnello,5 Becker Farm Roa, Roseland NJ, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound having the structure of Formula I

Formula I wherein X--Y is:

1) C(R )-C(0),

2) C(R )-C(R5)(OR1),

3) C=C(R5),

6) C(R4)-C(H)(R5),

7) C(R4)-C(D)(R5),

6) C(R )-C(F)(R5),

7) C(R4)-0,

8) C(R4)-S(0)n , wherein n

9) C=N,

10) C(R )-N(R6),

11) C(R4)-C(=N-OR7), or

12) C(R4)-C(=N-CN);

R1 and R2 are independently:

1) hydrogen,

2) -CH3, or

3 -CH2CH3;

R3 is:

1) hydrogen, 2) -CH3,

4) deuteron;

R4 is:

1) hydrogen,

2) -CH3,

3) halogen, or

4) deuteron;

R5 is:

1) hydrogen,

2) deuteron,

3) lower alkyl,

4) lower alkenyl,

5) lower alkynyl,

6) aryl,

7) nitrite, or

8) fluoro;

R6 is:

1) -C(0)-R8,

2) -S(0)2-R10,

3) lower alkyl,

4) lower alkenyl,

5) lower alkynyl, c

6) hydrogen;

R7 is:

1) hydrogen, or

2) lower alkyl;

R8 is:

1) hydrogen, 2) lower alkyl,

3) aryl,

4) -O-R9, or

5) -N(R7)(R9);

R9 is:

1) hydrogen,

2) lower alkyl,

3) lower alkenyl

4) lower alkynyl

5) aryl;

R10 is:

1) lower alkyl,

2) phenyl, or 3) -CF3;

A is:

4)

5)

Ri, Rm, Rn and R0 are each independently:

1) C(Re), or

2) nitrogen;

wherein at least one of Ri, Rm, Rn, or R0 is nitrogen;

Ra, and R are each independently:

1) hydrogen,

2) -CH3, or

and wherein if Ri and/or R2 is hydrogen, then Ra and Rb are selected from methyl and ethyl;

Rc and Rd, are each independently:

1) hydrogen,

2) alkoxy,

3) lower alkyl,

4) lower alkenyl,

5) haloalkyl,

6) hydroxy,

7) fluoro, or

8) deuteron;

Ro is:

1) hydrogen,

2) lower alkyl,

3) alkoxyalkyl, or 4) fluoro; and where Rc and R0 may be taken together as:

1) oxo,

2) =CH2) or

3) lower alkenyl;

Re, Rf Rg and Rh are each independently:

1) hydrogen,

2) deuteron,

3) lower alkyl,

4) halogen,

5) nitro,

6) alkoxy,

7) nitrite,

8) carboxamido,

9) urea,

10) alkylcarbonyi,

11) arylcarbonyl,

12) carbamoyl,

13) amidyl, or

14) amino;

Z is:

1) oxygen, or

2) sulfur; including pharmaceutically acceptable salts, solvates and hydrates thereof,

and wherein the compound is not a member of the Excluded

Compound Group.

2. The compound of claim 1 , wherein X^Y is C(R4)-C(0), C(R4)- C(H)OH, C=C(R5), C(R4)-C(=N-OR7) or C(R4)-C(=N-CN).

3. The compound of claim 2, wherein R and R2 are each hydrogen. 4. The compound of claim 2, wherein Ra and Rt are independently methyl or ethyl.

5. The compound of claim 4, wherein both of Ra and R are methyl . 6. The compound of claim 2, wherein R3 is hydrogen or deuteron.

7. The compound of claim 6, wherein R3 is deuteron.

8. The compound of claim 2, wherein R4 and R5 are each independently hydrogen or methyl.

9. The compound of claim 8, wherein at least one of R4 and R5 is hydrogen. 10. The compound of claim 8, wherein both R4 and R5 are hydrogen.

11. The compound of claim 1 , wherein A is an aryl group and one or more of R and R2 is methyl, ethyl or hydrogen. 12. The compound of claim 11 , wherein each of R1 and R2 is methyl or hydrogen.

13. The compound of claim 12, wherein each of R1 and R2 is hydrogen.

14. The compound of claim 1 1 , wherein R3 is hydrogen, deuteron or methyl.

15. The compound of claim 14, wherein R3 is hydrogen or deuteron.

16. The compound of claim 15, wherein R3 is deuteron. 17. The compound of claim 11 , wherein Ra and Rb are independently hydrogen, methyl or ethyl.

18. The compound of claim 17, wherein Ra and R are independently methyl or ethyl.

19. The compound of claim 17, wherein both Ra and Rb are methyl.

20. The compound of claim 11 , wherein Rc and Rd are hydrogen, lower alkyl or alkoxy.

21. The compound of claim 20, wherein Rc and Rd are hydrogen or lower alkyl.

22. The compound of claim 20, wherein Rc and Rd are hydrogen or methyl.

23. A compound having the structure of Formula II

Formula II wherein R, and Rj are each independently:

1) hydrogen,

2) deuteron, 3) fluoro,

4) alkoxy,

5) hydroxyl, or

6) lower alkyl;

Ri and Rj taken together are:

1 ) oxo,

2) =N-OR7, or

3) =N-CN;

Rp and Rq are each independently

1) hydrogen,

2) methyl, or

3) ethyl;

4)

7) cycloalkyl;

Rk is:

1 hydrogen,

2 lower alkyl,

3 halogen,

4 nitro,

5 alkoxy,

6 nitrile,

7 carboxamido,

8 urea,

9 alkylcarbonyl,

10 arylcarbonyl,

11 carbamoyl,

12 amidyl, or

13 amino; and wherein n, R1, R2 R3, Rc, Rd, Re, Rf, Rg, Rh, Ri, Rm, Rn,, R0 and Z are as defined for Formula I

including pharmaceutically acceptable salts, solvates and hydrates thereof,

and wherein the compound is not a member of the Excluded Compound Group.

24. The compound of claim 23, wherein R, and Rj are taken together as oxo, =N-OR7, or =N-CN.

25. The compound of claim 23, wherein Ri is hydroxy and Rj is hydrogen.

26. The compound of claim 23, wherein R1 and R2 are each independently methyl or ethyl.

27. The compound of claim 26, wherein both R1 and R2 are methyl.

28. The compound of claim 23, wherein R3 is hydrogen or methyl. 29. The compound of claim 23, wherein R3 methyl.

30. The compound of claim 23, wherein B is an aryl group.

31. The compound of claim 30, wherein R and R2 are methyl or ethyl.

32. The compound of claim 30, wherein R and R2 are methyl.

33. The compound of claim 30, wherein R3 is hydrogen, methyl, or ethyl.

34. The compound of claim 33, wherein R3 is hydrogen or methyl.

35. The compound of claim 33, wherein each of R1, R2 and R3 is a methyl.

36. The compound of claim 23, wherein Rp and Rq are independently hydrogen or methyl.

37. The compound of claim 23, wherein Rp and Rq are both hydrogen.

38. The compound of claim 23, wherein Rc and Rd are hydrogen, lower alkyl or alkoxy.

39. The compound of claim 38, wherein Rc and Rd are hydrogen or lower alkyl.

40. The compound of claim 23, wherein Rc and Rd are hydrogen or methyl. 41. The compound of claim 23, wherein R, and Rj are each independently hydrogen or hydroxyl.

42. The compound of claim 41 , wherein Rj is hydrogen and Rj is hydroxyl.

43. The compound of claim 41 , wherein Ri and Rj are taken together as oxo.

44. A method of inhibiting the formation or accumulation of a visual cycle product or of treating or preventing an ophthalmic condition in a subject at risk thereof, comprising contacting an opsin protein with a compound having the structure of Formula I

Formula I wherein X--Y is:

1 ) C(R4)-C(0), 2) C(R4)-C(R5)(OR1),

3) C=C(R5),

8) C(R4)-C(H)(R5),

9) C(R )-C(D)(R5),

6) C(R4)-C(F)(R5),

7) C(R4)-0,

8) C(R )-S(0)n , wherein n

9) C=N,

10) C(R )-N(R6),

11) C(R4)-C(=N-OR7), or

12) C(R4)-C(=N-CN);

R1 and R2 are independently:

1) hydrogen,

2) -CH3, or

3) -CH2CH3;

R3 is:

1) hydrogen,

2) -CH3,

4) deuteron;

R4 is:

1) hydrogen,

2) -CH3,

3) halogen, or

4) deuteron;

R5 is:

1) hydrogen,

2) deuteron,

3) lower alkyl,

4) lower alkenyl, 5) lower alkynyl,

6) aryl,

7) nitrite, or

8) fluoro;

1) -C(0)-RB,

2) -S(0)2-R10,

3) lower alkyl,

4) lower alkenyl,

5) lower alkynyl, or

6) hydrogen;

R7 is:

.1) hydrogen, or 2) lower alkyl;

R8 is:

1) hydrogen,

2) lower alkyl,

3) aryl,

4) -O-R9, or

5) -N(R7)(R9);

R9 is:

1) hydrogen,

2) lower alkyl,

3) lower alkenyl,

4) lower alkynyl,

5) aryl;

R10 is:

1) lower alkyl,

2) phenyl, or 3) -CF3;

A is:

1)

2)

3)

4)

5)

Ri, Rm, Rn and R0 are each independently:

1) C(Re), or

2) nitrogen;

wherein at least one of R|, Rm, Rn, or R0 is nitrogen;

Ra, and Rb, are each independently:

1) hydrogen, 2) -CH3, or

3) -CH2CH3; and wherein if Ri and/or R2 is hydrogen, then Ra and Rb are selected from methyl and ethyl;

Rc and Rd are each independently:

1) hydrogen,

2) alkoxy,

3) lower alkyl,

4) lower alkenyl

5) haloalkyl,

6) hydroxy,

7) fluoro, or

8) deuteron;

Ro is:

1) hydrogen,

2) lower alkyl,

3) alkoxyalkyl, or

4) fluoro; and where Rc and R0 may be taken together as:

1) oxo,

2) =CH2, or

3) lower alkenyl;

Re, Rf Rg and Rh are each independently:

1) hydrogen,

2) deuteron,

3) lower alkyl,

4) halogen,

5) nitro, 6) alkoxy,

7) nitrile,

8) carboxamido,

9) urea,

10) alky!carbonyl,

11) arylcarbonyl,

12) carbamoyl,

13) amidyl, or

14) amino;

Z is:

1) oxygen, or

2) sulfur; including pharmaceutically acceptable salts, solvates and hydrates thereof.

45. The method of claim 44, wherein said compound reduces mislocalization of said opsin protein.

46. The method of claim 44, wherein said compound reduces hydration of said opsin protein.

47. The method of claim 44, wherein said opsin protein is present in a cell.

48. The method of claim 47, wherein said cell is present in a mammalian eye. 49. The method of claim 48, wherein said mammal is a human being.

50. The method of claim 44, wherein said visual cycle product is a toxic visual cycle product.

51. The method of claim 50, wherein said visual cycle product is lipofuscin or N-retinylidene-N-retinylethanolamine (A2E).

52. The method of claim 44, wherein said ophthalmic condition is an ocular protein mislocalization disorder.

53. The method of claim 44, wherein said ophthalmic condition is selected from the group consisting of wet or dry age related macular degeneration (ARMD), retinitis pigmentosa (RP), a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity, normal vision loss related aging and normal loss of night vision related to aging .

54. The method of claim 44, wherein ^Y is C(R 4)-C(0), C(R4)- C(H)OH, C=C(R5), C(R )-C(=N-OR7) or C(R )-C(=N-CN).

55. The method of claim 54, wherein R1 and R2 are each hydrogen.

56. The method of claim 54, wherein Ra and Rb are independently methyl or ethyl.

57. The method of claim 56, wherein both of Ra and Rb are methyl.

58. The method of claim 54, wherein R3 is hydrogen or deuteron.

59. The method of claim 58, wherein R3 is deuteron. 60. The method of claim 54, wherein R4 and R5 are each independently hydrogen or methyl.

61. The method of claim 60, wherein at least one of R4 and R5 is hydrogen.

62. The method of claim 60, wherein both R4 and R5 are hydrogen. 63. The method of claim 44, wherein A is an aryl group and one or more of R and R2 is methyl, ethyl or hydrogen.

64. The method of claim 63, wherein each of R1 and R2 is methyl or hydrogen.

65. The method of claim 64, wherein each of R and R2 is hydrogen.

66. The method of claim 63, wherein R3 is hydrogen, deuteron or methyl.

67. The method of claim 66, wherein R3 is hydrogen or deuteron.

68. The method of claim 67, wherein R3 is deuteron. 69. The method of claim 63, wherein Ra and Rb are independently hydrogen, methyl or ethyl.

70. The method of claim 63, wherein Ra and Rb are independently methyl or ethyl.

71. The method of claim 63, wherein both Ra and R are methyl.

72. The method of claim 63, wherein Rc and Rd are hydrogen, lower alkyl or alkoxy.

73. The method of claim 72, wherein Rc and Rd are hydrogen or lower alkyl.

74. The method of claim 72, wherein Rc and Rd are hydrogen or methyl.

75. A method of inhibiting the formation or accumulation of a visual cycle product or of treating or preventing an ophthalmic condition in a subject at risk thereof, comprising contacting an opsin protein with a compound having the structure of Formula II

Formula II wherein Rj and Rj are each independ<

1) hydrogen,

2) deuteron,

3) fluoro,

4) alkoxy,

5) hydroxyl, or

6) lower alkyl;

Ri and Rj taken together are:

1) oxo,

2) =N-OR7, or

3) =N-CN;

Rp and Rq are each independently:

1) hydrogen,

2) methyl, or

3) ethyl;

k is:

1) hydrogen,

2) lower alkyl,

3) halogen, 4) nitro, 5) alkoxy,

6) nitrile,

7) carboxamido,

8) urea,

9) alkylcarbonyl,

10) arylcarbonyl,

11) carbamoyl,

12) amidyl, or

13) amino; wherein n, R , R2 R3, Rc, Rd, Re, Rf, Rg, Rh, Ri, Rm, Rn,, R0 and Z are as defined for Formula I

including pharmaceutically acceptable salts, solvates and hydrates thereof.

76. The method of claim 75, wherein said compound reduces mislocalization of said opsin protein.

77. The method of claim 75, wherein said compound reduces hydration of said opsin protein.

78. The method of claim 75, wherein said opsin protein is present in a cell.

79. The method of claim 78, wherein said cell is present in a mammalian eye.

80. The method of claim 79, wherein said mammal is a human being.

81. The method of claim 75, wherein said visual cycle product is a toxic visual cycle product.

82. The method of claim 81 , wherein said visual cycle product is lipofuscin or N-retinylidene-N-retinylethanolamine (A2E).

83. The method of claim 75, wherein said ophthalmic condition is an ocular protein mislocalization disorder.

84. The method of claim 75, wherein said ophthalmic condition is selected from the group consisting of wet or dry age related macular degeneration (ARMD), retinitis pigmentosa (RP), a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity, normal vision loss related aging and normal loss of night vision related to aging.

85. The compound of claim 75, wherein R, and Rj are taken together as oxo, =N-OR7, or =N-CN.

86. The compound of claim 75, wherein Rj is hydroxy and Rj is hydrogen.

87. The compound of claim 75, wherein R1 and R2 are each independently methyl or ethyl.

88. The compound of claim 87, wherein both R1 and R2 are methyl.

89. The compound of claim 75, wherein R3 is hydrogen or methyl.

90. The compound of claim 75, wherein R3 methyl.

91. The compound of claim 75, wherein B is an aryl group. 92. The compound of claim 91 , wherein R1 and R2 are methyl or ethyl.

93. The compound of claim 91 , wherein R1 and R2 are methyl.

94. The compound of claim 91 , wherein R3 is hydrogen, methyl, or ethyl.

95. The compound of claim 94, wherein R3 is hydrogen or methyl.

96. The compound of claim 94, wherein each of R1, R2 and R3 is a methyl.

97. The compound of claim 75, wherein Rp and Rq are independently hydrogen or methyl.

98. The compound of claim 75, wherein Rp and Rq are both hydrogen.

99. The compound of claim 75, wherein Rc and Rd are hydrogen, lower alkyl or alkoxy.

100. The compound of claim 99, wherein Rc and Rd are hydrogen or lower alkyl. 101. The compound of claim 75, wherein Rc and Rd are hydrogen or methyl.

102. The compound of claim 75, wherein R, and Rj are each independently hydrogen or hydroxyl.

103. The compound of claim 102, wherein R, is hydrogen and Rj is hydroxyl.

104. The compound of claim 102, wherein R, and Rj are taken together as oxo.

105. A method of inhibiting the formation or accumulation of a visual cycle product or of treating or preventing an ophthalmic condition in a subject at risk thereof, comprising contacting an opsin protein with compound 3, 3a, 3b, 5, 7, 11 , 12, 13, 17, 18, 28, 29, 36, 37, 40 41, 43 ,47, 48, 53, 54, 56, 57, 58, 61 , 62, 63, 64a, 64b, 67, 68, 69, 70, 71 , 73, 75, 79, 80, 84, 87, 88, 90, 93, 101 , 105, 109, 110, 112a, 112b, 113, 115, 116a, 116b, 123, 125, 126, 129, 131 , 132, 133, 137, 139, 142, 147, 149, 155, 166, 168, 169, 171 , 182, 187, 189, 190, 191 , 202, 205, 207, 213, 214, 217, 218 or 220.

106. A composition, comprising a therapeutically effective amount of a compound of claim 1 in a pharmaceutically acceptable carrier. 07. A composition, comprising a therapeutically effective amount of a compound of claim 23 in a pharmaceutically acceptable carrier.

Description:
OPSIN-BINDING LIGANDS, COMPOSITIONS AND METHODS OF USE

PRIORITY CLAIM

This application claims priority of U.S. Provisional Application 61/397,120, filed 7 June 2010, the disclosure of which is hereby incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention relates to compounds and compositions thereof for use in the treatment and/or prevention of ophthalmic diseases.

BACKGROUND OF THE INVENTION A diminished visual acuity or total loss of vision may result from a number of eye diseases or disorders caused by dysfunction of tissues or structures in the anterior segment of the eye and/or posterior segment of the eye. Of those that occur as a consequence of a dysfunction in the anterior segment, aberrations in the visual cycle are often involved. The visual cycle (also frequently referred to as the retinoid cycle) comprises a series of light- driven and/or enzyme catalyzed reactions whereby a light-sensitive chromophore (called rhodopsin) is formed by covalent bonding between the protein opsin and the retinoid agent 1-cis-retinal and subsequently, upon exposure to light, the 11-cis-retinal is converted to all-trans-retinal, which can then be regenerated into 11-cis-retinal to again interact with opsin. A number of visual, ophthalmic, problems can arise due to interference with this cycle. It is now understood that at least some of these problems are due to improper protein folding, such as that of the protein opsin.

The main light and dark photoreceptor in the mammalian eye is the rod cell, which contains a folded membrane containing protein molecules that can be sensitive to light, the main one being opsin. Like other proteins present in mammalian cells, opsin is synthesized in the endoplasmic reticulum (i.e., on ribosomes) of the cytoplasm and then conducted to the cell membrane of rod cells. In some cases, such as due to genetic defects and mutation of the opsin protein, opsin can exhibit improper folding to form a conformation that either fails to properly insert into the membrane of the rod cell or else inserts but then fails to properly react with 11-cis-retinal to form native rhodopsin. In either case, the result is moderate to severe interference with visual perception in the animal so afflicted.

Among the diseases and conditions linked to improper opsin folding is retinitis pigmentosa (RP), a progressive ocular-neurodegenerative disease (or group of diseases) that affects an estimated 1 to 2 million people worldwide. In RP, photoreceptor cells in the retina are damaged or destroyed, leading to loss of peripheral vision (i.e., tunnel vision) and subsequent partial or near- total blindness.

In the American population the most common defect occurs as a result of replacement of a proline residue by a histidine residue at amino acid number 23 in the opsin polypeptide chain (dubbed "P23H"), caused by a mutation in the gene for opsin. The result is production of a destabilized form of the protein, which is misfolded and aggregates in the cytoplasm rather than being transported to the cell surface. Like many other protein conformational diseases (PCDs), the clinically common P23H opsin mutant associated with autosomal dominant RP is misfolded and retained intracellular^. The aggregation of the misfolded protein is believed to result in photoreceptor damage and cell death. Recent studies have identified small molecules that stabilize misfolded mutant proteins associated with disease. Some of these, dubbed "chemical chaperones," stabilize proteins non-specifically. Examples of these include glycerol and trimethylamine oxide. These are not very desirable for treating ophthalmic disease because such treatment usually requires high dosages that may cause toxic side effects. Other agents, dubbed "pharmacological chaperones," (which include native ligands and substrate analogs) act to stabilize the protein by binding to specific sites and have been identified for many misfolded proteins, e.g., G-protein coupled receptors. Opsin is an example of a G-protein coupled receptor and its canonical pharmacological chaperones include the class of compounds referred to as retinoids. Thus, certain retinoid compounds have been shown to stabilize mutant opsin proteins (see, for example, U.S. Patent Pub. 2004-0242704, as well as Noorwez et al., J. Biol. Chem., 279(16): 16278-16284 (2004)).

The visual cycle comprises a series of enzyme catalyzed reactions, usually initiated by a light impulse, whereby the visual chromophore of rhodopsin, consisting of opsin protein bound covalently to 11-cis-retinal, is converted to an all-trans-isomer that is subsequently released from the activated rhodopsin to form opsin and the all-trans-retinal product. This part of the visual cycle occurs in the outer portion of the rod cells of the retina of the eye. Subsequent parts of the cycle occur in the retinal pigmented epithelium (RPE). Components of this cycle include various enzymes, such as dehydrogenases and isomerases, as well as transport proteins for conveying materials between the RPE and the rod cells.

As a result of the visual cycle, various products are produced, called visual cycle products. One of these is all-trans-retinal produced in the rod cells as a direct result of light impulses contacting the 11-cis-retinal moiety of rhodopsin. All-trans-retinal, after release from the activated rhodopsin, can be regenerated back into 11-cis-retinal or can react with an additional molecule of all-trans-retinal and a molecule of phosphatidylethanolamine to produce N- retinylidene-N-retinylethanolamine (dubbed "A2E"), an orange-emitting fluorophore that can subsequently collect in the rod cells and in the retina pigmented epithelium (RPE). As A2E builds up (as a normal consequence of the visual cycle) it can also be converted into lipofuscin, a toxic substance that has been implicated in several abnormalities, including ophthalmic conditions such as wet and dry age related macular degeneration (ARMD). A2E can also prove toxic to the RPE and has been associated with dry ARMD.

Because the build-up of toxic visual cycle products is a normal part of the physiological process, it is likely that all mammals, especially all humans, possess such an accumulation to some extent throughout life. However, during surgical procedures on the eye, especially on the retina, where strong light is required over an extended period, for example, near the end of cataract surgery and while implanting the new lens, these otherwise natural processes can cause toxicity because of the build-up of natural products of the visual cycle. Additionally, excessive rhodopsin activation as a result of bright light stimulation can cause photoreceptor cell apoptosis via an AP-1 transcription factor dependent mechanism. Because of this, there is a need for agents that can be administered prior to, during or after (or any combination of these) the surgical process and that has the effect of inhibiting rhodopsin activation as well as reducing the production of visual cycle products that would otherwise accumulate and result in toxicity to the eye, especially to the retina.

The present invention answers this need by providing small molecules which noncovalently bind to opsin or mutated forms of opsin for treating and/or amelioration such conditions, if not preventing them completely. Importantly, such agents are not natural retinoids and thus are not tightly controlled for entrance into the rod cells, where mutated forms of opsin are synthesized and/or visual cycle products otherwise accumulate. Therefore, such agents can essentially be titrated in as needed for facilitating the proper folding trafficking of mutated opsins to the cell membrane or prevention of rhodopsin activation that can lead to the excessive build-up of visual cycle products like all-trans-retinal that in turn can lead to toxic metabolic products. Such compounds may compete with 11-cis-retinal to reduce all-trans-retinal by tying up the retinal binding pocket of opsin to prevent excessive all-trans- retinal build up. Thus, the compounds provided by the present invention have the advantage that they do not directly inhibit the enzymatic processes by which 11-cis-retinal is produced in the eye (thus not contributing to retinal degeneration). Instead, the formation of all-trans-retinal is limited and thereby the formation of A2E is reduced. Finally, by limiting the ability of 11-cis-retinal to combine with opsin to form rhodopsin, rhodopsin activation caused by bright light stimulation especially during ophthalmic surgery is also diminished thus preventing the photocell death that results.

Mislocalization of photoreceptor cell visual pigment proteins (opsins) can occur in various ocular diseases, and also with normal aging. In both cases the accumulation of mislocalized opsin leads to the decline in viability of photoreceptor cells. With time this mislocalized opsin accumulation leads to rod and cone cell death, retinal degeneration, and loss of vision. The present invention solves this problem by providing a method of correcting mislocalized opsin within a photoreceptor cell by contacting a mislocalized opsin protein with an opsin-binding agent that binds reversibly and/or non-covalently to said mislocalized opsin protein, and promotes the appropriate intracellular processing and transport of said opsin protein. This correction of mislocalization relieves photoreceptor cell stress, preventing decline in viability and death of photoreceptor cells in various diseases of vision loss, and in normal age-related decline in dim-light and peripheral rod-mediated vision, central cone-mediated vision, and loss of night vision.

Computer-assisted molecular docking has lead to the successful discovery of novel ligands for more than 30 targets (Shoichet et al., Curr. Opin. in Chem. Biol. 6: 439-46 (2002)). This strategy has been applied primarily to enzymes, such as aldose reductase (Iwata et al., J. Med. Chem. 44: 1718-28 (2001)), Bcl-2, matriptase (Enyedy et al., J. Med. Chem. 44: 1349-55 (2001)), adenovirus protease (Pang et al., FEBS Letters 502: 93-97 (2001)), AmpC fl-lactamase, carbonic anhydrase (Gruneberg et al., J. Med. Chem. 45: 3588-602 (2002)), HPRTase (Freymann et al., Chemistry & Biology 7: 957-68 (2000)), dihydrodipicolinate (Paiva et al., Biochimica Biophysica Acta 1545: 67-77 (2001)) and Cdk4 (Honma et al., J. Med. Chem. 44: 4615-27 (2001)). Improvements in docking algorithms and multiprocessor resources have improved the technique of computer-assisted molecular docking such that it can now be applied to more challenging problems. For example, this approach has recently been applied to defining small molecules that target protein-protein interfaces, which are relatively broad and flat compared to easily targeted enzyme active sites.

More recently, a new computational technique defining the thermodynamic properties and phase behavior of water in confined regions of protein pockets has been developed (Young et. al., PNAS 104: 808-13 (2007)). The algorithm developed has been utilized to characterize the solvation of protein pockets. The molecular dynamics simulations and solvent analysis techniques have characterized the solvation of hydrophobic enclosures and correlated hydrogen bonds as inducing atypical entropic and enthalpic penalties of hydration which stabilize the protein-ligand complex with respect to the independently solvated ligand and protein. These criteria, commonly referred to as the water map, have been used to rationalize Factor Xa ligand binding (Abel et. al., JACS 130: 2817-31 (2008)).

BRIEF SUMMARY OF THE INVENTION

In one aspect, the present invention provides compounds having the structure of Formula I, including pharmaceutically acceptable salts, solvates and hydrates thereof, and compositions of said compounds:

Formula I wherein X-Y , A, R 1 , R 2 , R 3 , R a , R b , R c , R d , and R 0 are as described elsewhere herein.

In another aspect, the present invention provides compounds having the structure of Formula II, including pharmaceutically acceptable salts, compounds:

Formula II wherein B, R 1 , R 2 , R 3 , R a , R b , R c , R d , and R 0 are as described elsewhere herein.

In a related aspect, the present invention relates to a method of inhibiting the formation or accumulation of a visual cycle product, comprising contacting an opsin protein with a compound recited herein to inhibit formation of said visual cycle product relative to when said contacting does not occur.

In a further aspect, the present invention relates to a method to reduce the light toxicity associated with ophthalmic surgery by preventing rhodopsin regeneration during surgery to a mammalian eye and/or prevent or slow the formation of toxic visual cycle products by fractionally preventing rhodopsin formation during periods of light activation thereby providing a treatment of ocular conditions associated with the build up of visual products such as wet or dry ARMD. In yet a further aspect, the present invention relates to a method of correcting the proper folding and trafficking of mutated opsin proteins, comprising contacting a mutated opsin protein with a compound that stabilizes the proper three dimensional conformation of the protein relative to when said contacting does not occur wherein the compound has the structure of Formula I including pharmaceutically acceptable salts, solvates and hydrates thereof.

In one embodiment, the ligand selectively binds reversibly or non- covalently to opsin. In another embodiment, the ligand binds at or near the 11-cis- retinal binding pocket of the opsin protein. In yet another embodiment, the ligand binds to the opsin protein so as to inhibit or slow the covalent binding of 11-cis-retinal to the opsin protein when the 11-cis-retinal is contacted with the opsin protein in the presence of the ligand. In yet another embodiment, the ligand binds to the opsin in the retinal binding pocket of opsin protein or disrupts 11-cis-retinal binding to the retinal binding pocket of opsin. In yet another embodiment, the ligand binds to the opsin protein so as to inhibit covalent binding of 11-cis-retinal to the opsin protein. In yet another embodiment, the mammal is a human being. In yet another embodiment, slowing or halting the progression of wet or dry ARMD is associated with reducing the level of a visual cycle product, for example, a visual cycle product formed from all-trans-retinal, such as lipofuscin or N-retinylidine-N-retinylethanolamine (A2E). In yet another embodiment slowing or halting the progression of RP is associated with correcting the folding of mutated opsins. In another embodiment, the administering is topical administration, local administration (e.g., intraocular or periocular injection or implant) or systemic administration (e.g., oral, injection). In yet another embodiment, the light toxicity is related to an ophthalmic procedure (e.g., ophthalmic surgery). In still another embodiment, the administering occurs prior to, during, or after the ophthalmic surgery.

Mislocalization of photoreceptor cell visual pigment proteins (opsins) can occur in various ocular diseases, and also with normal aging. In such cases the accumulation of mislocalized opsin leads to the decline in viability of photoreceptor cells. With time this mislocalized opsin accumulation leads to rod and cone cell death, retinal degeneration, and loss of vision. In one aspect, the invention provides a method of correcting mislocalized opsin within a photoreceptor cell, comprising contacting a mislocalized opsin protein with an opsin-binding agent that binds reversibly and/or non-covalently to said mislocalized opsin protein to promote the appropriate intracellular processing and transport of said opsin protein. This correction of mislocalization reduces photoreceptor cell stress, preventing photoreceptor cell decline in viability and death in various diseases of vision loss, and in normal age-related decline in dim-light and peripheral rod-mediated vision, central cone-mediated vision, and loss of night vision.

In various embodiments, the ocular protein mislocalization disorder is any one or more of wet or dry form of macular degeneration, retinitis pigmentosa, a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity, retinitis pigmentosa, normal vision loss related aging and normal loss of night vision related to aging .

In still another embodiment, the method further involves administering to a mammal, preferably a human being, an effective amount of at least one additional agent selected from the group consisting of a proteasomal inhibitor, an autophagy inhibitor, a lysosomal inhibitor, an inhibitor of protein transport from the ER to the Golgi, an Hsp90 chaperone inhibitor, a heat shock response activator, a glycosidase inhibitor, and a histone deacetylase inhibitor. In yet another embodiment, the opsin binding ligand and the additional agent are administered simultaneously.

In still another embodiment, the opsin binding ligand and the additional agent are each incorporated into a composition that provides for their long- term release. In another embodiment, the composition is part of a microsphere, nanosphere, nano emulsion or implant. In another embodiment, the composition further involves administering a mineral supplement, at least one anti-inflammatory agent, such as a steroid (e.g., any one or more of cortisone, hydrocortisone, prednisone, prednisolone, methylprednisolone, triamcinolone, betamethasone, beclamethasone and dexamethasone), or at least one anti-oxidant, such as vitamin A, vitamin C and vitamin E. In various embodiments, the opsin binding ligand, the anti-inflammatory agent, and/or the anti-oxidant are administered simultaneously.

BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows predicted hydration of the rod opsin retinal binding pocket as developed from a homology model of human rhodopsin based upon the crystal structure of bovine rhodopsin. As a reference, the surface volume of 11-cis retinal is indicated by general outline. Specific hydration sites are shown as circles where water molecules would be predicted to reside within the pocket in the absence of a ligand. Circles labeled with a "D" designate hydration sites that are in very hydrophobic environments and thus upon displacement by a ligand are predicted to lower the energy of the ligand protein complex relative to the hydrated apoprotein. Circles labeled with an "R" designate hydration sites where the water molecule is forming stable hydrogen bonds with functional groups on the protein and thus signify coordinates within the binding pocket where suitable hydrogen bonding functionality of the ligand should be incorporated to replace the hydrogen bonding interactions that are broken between the water molecule and the protein upon binding of the ligand.

Figure 2 shows the increase in regeneration of 500 nm absorbing pigment upon treatment with 9-cis retinal from P23H opsin that was treated with 20 μΜ of β-ionone during mutant protein production relative to pigment formation in the presence of vehicle (DMSO) alone.

DEFINITIONS

As used throughout the disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings.

By "mislocalization" of a photoreceptor cell visual pigment protein (for example, opsin, especially human opsin) is meant that the synthesized protein is not found at the normal or appropriate cellular location. "Pharmacologic chaperones" refer to small molecular weight chemical compounds that interact with a protein (usually with a mis-folded, or un-folded protein) in such a way as to alter the folding or confirmation of said protein. Such an interaction can have diverse consequences on the cellular fate of the protein, including but not limited to leading to increased stability and increased levels of functional protein, increased stability and increased levels of non-functional protein, or decreased stability and decreased levels of functional or non-functional protein.

"Productive chaperone" refers to a pharmacologic chaperone that when interacting with a protein leads to an increased level of functional protein.

"Counterproductive, shipwreck or destructive chaperone" refers to a pharmacologic chaperone that interacts with a protein (usually with a mis- folded, or un-folded protein) and this interaction leads to a decreased stability and/or decreased levels of functional or non-functional protein.

By "proteasomal inhibitor" is meant a compound that reduces a proteasomal activity, such as the degradation of a ubiquinated protein.

By "autophagy inhibitor" is meant a compound that reduces the degradation of a cellular component by a cell in which the component is located.

By "lysosomal inhibitor" is meant a compound that reduces the intracellular digestion of macromolecules by a lysosome. In one embodiment, a lysosomal inhibitor decreases the proteolytic activity of a lysosome. By "Inhibitor of ER-Golgi protein transport" is meant a compound that reduces the transport of a protein from the ER (endoplasmic reticulum) to the Golgi, or from the Golgi to the ER.

By "HSP90 chaperone inhibitor" is meant a compound that reduces the chaperone activity of heat shock protein 90 (HSP90). In one embodiment, the inhibitor alters protein binding to an HSP90 ATP/ADP pocket.

By "heat shock response activator" is meant a compound that increases the chaperone activity or expression of a heat shock pathway component. Heat shock pathway components include, but are not limited to, HSP100, HSP90, HSP70, HASP60, HSP40 and small HSP family members.

By "glycosidase inhibitor" is meant a compound that reduces the activity of an enzyme that cleaves a glycosidic bond.

By "histone deacetylase inhibitor" is meant a compound that reduces the activity of an enzyme that deacetylates a histone. By "reduces" or "increases" is meant a negative or positive alteration, respectively. In particular embodiments, the alteration is by at least about 10%, 25%, 50%, 75%, or 100% of the initial level of the protein produced in the absence of the opsin binding ligand.

As used herein, the term "wild-type conformation" refers to the three dimensional conformation or shape of a protein that is free of mutations to its amino acid sequence. For opsin, this means a protein free from mutations that cause misfiling, such as the mutation designated P23H (meaning that a proline is replaced by a histidine at residue 23 starting from the N-terminus). Opsin in a "wild-type conformation" is capable of opsin biological function, including but not limited to, retinoid binding, visual cycle function, and insertion into a photoreceptor membrane. By "agent" is meant a small compound (also called a "compound"), polypeptide, polynucleotide, or fragment thereof. The terms compound and agent are used interchangeably unless specifically stated otherwise herein for a particular agent or compound. By "correcting the conformation" of a protein is meant inducing the protein to assume a conformation having at least one biological activity associated with a wild-type protein.

By "misfolded opsin protein" is meant a protein whose tertiary structure differs from the conformation of a wild-type protein, such. that the misfolded protein lacks one or more biological activities associated with the wild-type protein.

By "selectively binds" is meant a compound that recognizes and binds a polypeptide of the invention, such as opsin, but which does not substantially recognize and bind other molecules, especially non-opsin polypeptides, in a sample, for example, a biological sample. By "effective amount" or "therapeutically effective amount" is meant a level of an agent sufficient to exert a physiological effect on a cell, tissue, or organ or a patient. As used herein, it is the amount sufficient to effect the methods of the invention to achieve the desired result.

By "pharmacological chaperone" is meant a molecule that upon contacting a mutant protein is able to facilitate/stabilize the proper folding of the protein such that it acts and functions much more like wild type protein than would be the case in the absence of the molecule.

By "control" is meant a reference condition. For example, where a cell contacted with an agent of the invention is compared to a corresponding cell not contacted with the agent, the latter is the "control" or "control" cell. By "treat" is meant decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease, preferably an ocular disease, such as RP, AMD and/or light toxicity.

By "prevent" is meant reduce the risk that a subject will develop a condition, disease, or disorder, preferably an ocular disease, such as RP, AMD and/or light toxicity.

By "competes for binding" is meant that a compound of the invention and an endogenous ligand are incapable of binding to a target at the same time. Assays to measure competitive binding are known in the art, and include, measuring a dose dependent inhibition in binding of a compound of the invention and an endogenous ligand by measuring ti/2 , for example.

A "pharmaceutically acceptable salt" is a salt formed from an acid or a basic group of one of the compounds of the invention. Illustrative salts include, but are not limited to, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbatc, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesuifonate, and pamoate (i.e., 1 , 1 '-methytene-bis-(2-hydroxy-3-naphthoate)) salts.

The term "pharmaceutically acceptable salt" also refers to a salt prepared from a compound of the invention having an acidic functional group, such as a carboxylic acid functional group, and a pharmaceutically acceptable inorganic or organic base. Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl-N- ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lovyer alkylamines), such as mono-, bis-, or tris-(2-hydroxyethyl)- amine, 2-hydroxy- tert-butylamine, or tris-(hydroxymethyl)methylamine, N, N,-di-lower alkyl-N- (hydroxy lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)-amine, or tri-(2-hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like.

The term "pharmaceutically acceptable salt" also refers to a salt prepared from a compound disclosed herein, e.g., a salt of a compound of Example 1 , having a basic functional group, such as an amino functional group, and a pharmaceutically acceptable inorganic or organic acid. Suitable acids include, but are not limited to, hydrogen sulfate, citric acid, acetic acid, oxalic acid, hydrochloric acid, hydrogen bromide, hydrogen iodide, nitric acid, phosphoric acid, isonicotinic acid, lactic acid, salicylic acid, tartaric acid, ascorbic acid, succinic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucaronic acid, saccharic acid, formic acid, benzoic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, and p- toluenesulfonic acid. The term "pharmaceutically-acceptable excipient" as used herein means one or more compatible solid or liquid tiller, diluents or encapsulating substances that are suitable for administration into a human. The term "excipient" includes an inert substance added to a pharmacological composition to further facilitate administration of a compound. Examples of excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate administration.

The term "parenteral" includes subcutaneous, intrathecal, intravenous, intramuscular, intraperitoneal, or infusion.

The term "visual cycle product" refers to a chemical entity produced as a natural product of one or more reactions of the visual cycle (the reactive cycle whereby opsin protein binds 11-cis-retinal to form rhodopsin, which accepts a light impulse to convert -cis-retinal to all trans-retinal, which is then released from the molecule to regenerate opsin protein with subsequent binding of a new 11-cis-retinal to regenerate rhodopsin). Such visual cycle products include, but are not limited to, all-trans-retinal, lipofuscin and A2E. The term "light toxicity" refers to any condition affecting vision that is associated with, related to, or caused by the production and/or accumulation of visual cycle products. Visual cycle products include, but are not limited to, all-trans-retinal, lipofuscin or A2E. In one particular embodiment, light toxicity is related to exposure of the eye to large amounts of light or to very high light intensity, occurring, for example, during a surgical procedure on the retina.

The term "opsin" refers to an opsin protein, preferably a mammalian opsin protein, most preferably a human opsin protein. In one embodiment, the opsin protein is in the wild-type (i.e., physiologically active) conformation. One method of assaying for physiological activity is assaying the ability of opsin to bind 11-cis-retinal and form active rhodopsin. A mutant opsin, such as the P23H mutant, that is ordinarily misfolded has a reduced ability to bind 11-cis-retinal, and therefore forms little or no rhodopsin. Where the conformation of the mutant opsin has been corrected (for example, by binding to a pharmacological chaperone), the opsin is correctly inserted into the rod cell membrane so that its conformation is the same, or substantially the same, as that of a non-mutant opsin. This allows the mutant opsin to bind 11-cis- retinal to form active rhodopsin. Therefore, the methods of the invention operate to reduce the formation of visual cycle products.

"Alkyl" refers to an unbroken non-cyclic chain of carbon atoms that may be substituted with other chemical groups. It may also be branched or unbranched, substituted or unsubstituted.

"Lower alkyl" refers to a branched or straight chain acyclic alkyl group comprising one to ten carbon atoms, preferably one to eight carbon atoms, more preferably one to six carbon atoms. Exemplary lower alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, pentyl, neopentyl, iso-amyl, hexyl, and octyl.

All alkyl, alkenyl or alkynyl groups disclosed herein may be substituted with one or more of the following: lower alkyl, hydroxy, ester, amidyl, oxo, carboxyl, carboxamido, halo, cyano, nitrate, nitrite, thionitrate, thionitrite sulfhydryl and amino groups (as elsewhere defined herein).

"Haloalkyl" refers to a lower alkyl group, an alkenyl group, an alkynyl group, a bridged cycloalkyl group, a cycloalkyl group or a heterocyclic ring, as defined herein, to which is appended one or more halogens, as defined herein. Exemplary haloalkyl groups include trifluoromethyl, chloromethyl, 2- bromobutyl and 1-bromo-2-chloro-pentyl. "Alkenyl" refers to a branched or straight chain C 2 -Ci 0 hydrocarbon (preferably a C 2 -C 8 hydrocarbon, more preferably a C 2 -C 6 hydrocarbon) that can comprise one or more carbon-carbon double bonds. Exemplary alkenyl groups include propylenyl, buten-1-yl, isobutenyl, penten-1-yl, 2,2- methylbuten-1-yl, 3-methylbuten-1-yl, hexan-1-yl, hepten-1-yl and octen-1-yl.

"Lower alkenyl" refers to a branched or straight chain C2-C4 hydrocarbon that can comprise one or two carbon-carbon double bonds. "Substituted alkenyl" refers to a branched or straight chain C2-C10 hydrocarbon (preferably a C2-C8 hydrocarbon, more preferably a C2-C6 hydrocarbon) which can comprise one or more carbon-carbon double bonds, wherein one or more of the hydrogen atoms have been replaced with one or more R 100 groups, wherein each R 100 is independently a hydroxy, an oxo, a carboxyl, a carboxamido, a halo, a cyano or an amino group, as defined herein.

"Alkynyl" refers to an unsaturated acyclic C2-C10 hydrocarbon (preferably a C2-C8 hydrocarbon, more preferably a C2-C6 hydrocarbon) that can comprise one or more carbon-carbon triple bonds. Exemplary alkynyl groups include ethynyl, propynyl, butyn-1-yl, butyn-2-yl, pentyl-1-yl, pentyl-2- yl, 3-methylbutyn-1-yl, hexyl-1-yl, hexyl-2-yl, hexyl-3-yl and 3,3-dimethyl- butyn-1-yl. "Lower alkynyl" refers to a branched or straight chain C2-C4 hydrocarbon that can comprise one or two carbon-carbon triple bonds

"Bridged cycloalkyl" refers to two or more cycloalkyl groups, heterocyclic groups, or a combination thereof fused via adjacent or non- adjacent atoms. Bridged cycloalkyl groups can be unsubstituted or substituted with one, two or three substituents independently selected from alkyl, alkoxy, amino, alkylamino, dialkylamino, hydroxy, halo, carboxyl, alkylcarboxylic acid, aryl, amidyl, ester, alkylcarboxylic ester, carboxamido, alkylcarboxamido, oxo and nitro. Exemplary bridged cycloalkyl groups include adamantyl, decahydronapthyl, quinuclidyl, 2,6-dioxabicyclo(3.3.0)octane, 7- oxabicyclo(2.2.1 )heptyl and 8-azabicyclo(3,2,1)oct-2-enyl. "Cycloalkyl" refers to a saturated or unsaturated cyclic hydrocarbon comprising from about 3 to about 10 carbon atoms. Cycloalkyl groups can be unsubstituted or substituted with one, two or three substituents independently selected from alkyl, alkoxy, amino, alkylamino, dialkylamino, arylamino, diarylamino, alkylarylamino, aryl, amidyl, ester, hydroxy, halo, carboxyl, alkylcarboxylic acid, alkylcarboxylic ester, carboxamido, alkylcarboxamido, oxo, alkylsulfinyl, and nitro. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl and cyclohepta-1 ,3-dienyl.

"Heterocyclic ring or group" refers to a saturated or unsaturated cyclic or polycyclic hydrocarbon group having about 2 to about 12 carbon atoms where 1 to about 4 carbon atoms are replaced by one or more nitrogen, oxygen and/or sulfur atoms. Sulfur may be in the thio, sulfinyl or sulfonyl oxidation state. The heterocyclic ring or group can be fused to an aromatic hydrocarbon group. Heterocyclic groups can be unsubstituted or substituted with one, two or three substituents independently selected from alkyl, alkoxy, amino, alkylthio, aryloxy, arylthio, arylalkyl, hydroxy, oxo, thial, halo, carboxyl, carboxylic ester, alkylcarboxylic acid, alkylcarboxylic ester, aryl, arylcarboxylic acid, arylcarboxylic ester, amidyl, ester, alkylcarbonyl, arylcarbonyl, alkylsulfinyl, carboxamido, alkylcarboxamido, arylcarboxamido, sulfonic acid, sulfonic ester, sulfonamide nitrate and nitro. Exemplary heterocyclic groups include pyrrolyl, furyl, thienyl, 3-pyrrolinyl,4,5,6-trihydro-2H-pyranyl, pyridinyl, 1 ,4-dihydropyridinyl, pyrazolyl, triazolyl, pyrimidinyl, pyridazinyl, oxazolyl, thiazolyl, thieno[2,3-d]pyrimidine, 4,5,6,7-tetrahydrobenzo[b]thiophene, imidazolyl, indolyl, thiophenyl, furanyl, tetrahydrofuranyl, tetrazolyl, pyrrolinyl, pyrrolindinyl, oxazolindinyl 1 ,3-dioxolanyl, imidazolinyl, imidazolindinyl, pyrazolinyl, pyrazolidinyl, isoxazolyl, isothiazolyl, 1 ,2,3-oxadiazolyl, 1 ,2,3- triazolyl, 1 ,3,4-thiadiazolyl, 2H-pyranyl, 4H-pyranyl, piperidinyl, 1 ,4-dioxanyl, morpholinyl, 1,4-dithianyl, thiomorpholinyl, pyrazinyl, piperazinyl, 1 ,3,5- triazinyl, 1 ,3,5-trithianyl, benzo(b)thiophenyl, benzimidazolyl, benzothiazolinyl, quinolinyl and 2,6-dioxabicyclo(3.3.0)octane.

"Heterocyclic compounds" refer to mono- and polycyclic compounds comprising at least one aryl or heterocyclic ring.

"Aryl" refers to a monocyclic, bicyclic, carbocyclic or heterocyclic ring system comprising one or two aromatic rings. Exemplary aryl groups include phenyl, pyridyl, napthyl, quinoyi, tetrahydronaphthyl, furanyl, indanyl, indenyl, indoyl. Aryl groups (including bicyclic aryl groups) can be unsubstituted or substituted with one, two or three substituents independently selected from alkyl, alkoxy, alkylthio, amino, alkylamino, dialkylamino, arylamino, diarylamino, alkylarylamino, halo, cyano, alkylsulfinyl, hydroxy, carboxyl, carboxylic ester, alkylcarboxylic acid, alkylcarboxylic ester, aryl, arylcarboxylic acid, arylcarboxylic ester, alkylcarbonyl, arylcarbonyl, amidyl, ester, carboxamido, alkylcarboxamido, carbomyl, sulfonic acid, sulfonic ester, sulfonamido and nitro. Exemplary substituted aryl groups include tetrafluorophenyl, pentafluorophenyl, sulfonamide, alkylsulfonyl and arylsulfonyl.

"Cycloalkenyl" refers to an unsaturated cyclic C3-C10 hydrocarbon (preferably a C 3 -C 8 hydrocarbon, more preferably a C3-C6 hydrocarbon), which can comprise one or more carbon-carbon double bonds. "Alkylaryl" refers to an alkyl group, as defined herein, to which is appended an aryl group, as defined herein. Exemplary alkylaryl groups include benzyl, phenylethyl, hydroxybenzyl, fluorobenzyl and fluorophenylethyl. "Arylalkyl" refers to an aryl radical, as defined herein, attached to an alkyl radical, as defined herein. Exemplary arylalkyl groups include benzyl, phenylethyl, 4-hydroxybenzyl, 3-fluorobenzyl and 2-fluorophenylethyl. "Arylalkenyl" refers to an aryl radical, as defined herein, attached to an alkenyl radical, as defined herein. Exemplary arylalkenyl groups include styryl and propenylphenyl. "Cycloalkylalkyl" refers to a cycloalkyl radical, as defined herein, attached to an alkyl radical, as defined herein.

"Cycloalkylalkoxy" refers to a cycloalkyl radical, as defined herein, attached to an alkoxy radical, as defined herein.

"Cycloalkylalkylthio" refers to a cycloalkyl radical, as defined herein, attached to an alkylthio radical, as defined herein.

"Heterocyclicalkyl" refers to a heterocyclic ring radical, as defined herein, attached to an alkyl radical, as defined herein.

"Arylheterocyclic ring" refers to a bi- or tricyclic ring comprised of an aryl ring, as defined herein, appended via two adjacent carbon atoms of the aryl ring to a heterocyclic ring, as defined herein. Exemplary arylheterocyclic rings include dihydroindole and 1 ,2, 3,4-tetra-hydroquinoline.

"Alkylheterocyclic ring" refers to a heterocyclic ring radical, as defined herein, attached to an alkyl radical, as defined herein. Exemplary alkylheterocyclic rings include 2-pyridylmethyl and 1-methylpiperidin-2-one-3- methyl.

"Alkoxy" refers to R50O-, wherein R50 is an alkyl group, an alkenyl group or an alkynyl group as defined herein (preferably a lower alkyl group or a haloalkyl group, as defined herein). Exemplary alkoxy groups include methoxy, ethoxy, t-butoxy, cyclopentyloxy, trifluoromethoxy, propenyloxy and propargyloxy. "Aryloxy" refers to R55O-, wherein R55 is an aryl group, as defined herein. Exemplary arylkoxy groups include phenoxy, napthyloxy, quinolyloxy, isoquinolizinyloxy. "Alkylthio" refers to R 50 S-, wherein R50 is an alkyl group, as defined herein.

"Lower alkylthio" refers to a lower alkyl group, as defined herein, appended to a thio group, as defined herein.

"Arylalkoxy" or "alkoxyaryl" refers to an alkoxy group, as defined herein, to which is appended an aryl group, as defined herein. Exemplary arylalkoxy groups include benzyloxy, phenylethoxy and chlorophenylethoxy. "Arylalklythio" refers to an alkylthio group, as defined herein, to which is appended an aryl group, as defined herein. Exemplary arylalklythio groups include benzylthio, phenylethylthio and chlorophenylethylthio.

"Arylalkylthioalkyl" refers to an arylalkylthio group, as defined herein, to which is appended an alkyl group, as defined herein. Exemplary arylalklythioalkyi groups include benzylthiomethyl, phenylethylthionnethyl and chlorophenylethylthioethy I .

"Alkylthioalkyl" refers to an alkylthio group, as defined herein, to which is appended an alkyl group, as defined herein. Exemplary alkylthioalkyl groups include allylthiomethyl, ethylthiomethyl and trifluoroethylthiomethyl.

"Alkoxyalkyl" refers to an alkoxy group, as defined herein, appended to an alkyl group, as defined herein. Exemplary alkoxyalkyl groups include methoxymethyl, methoxyethyl and isopropoxymethyl. "Alkoxyhaloalkyl" refers to an alkoxy group, as defined herein, appended to a haloalkyi group, as defined herein. Exemplary alkoxyhaloalkyl groups include 4- methoxy-2-chlorobutyl. "Cycloalkoxy" refers to R54O-, wherein R 54 is a cycioalkyi group or a bridged cycioalkyi group, as defined herein. Exemplary cycloalkoxy groups include cyclopropyloxy, cyclopentyloxy and cyclohexyloxy.

"Cycloalkylthio" refers to R54S-, wherein R 54 is a cycioalkyi group or a bridged cycioalkyi group, as defined herein. Exemplary cycloalkylthio groups include cyclopropylthio, cyclopentylthio and cyclohexylthio.

"Haloalkoxy" refers to an alkoxy group, as defined herein, in which one or more of the hydrogen atoms on the alkoxy group are substituted with halogens, as defined herein. Exemplary haloalkoxy groups include 1 ,1 ,1- trichloroethoxy and 2-bromobutoxy.

"Hydroxy" refers to -OH. "Oxy" refers to -O-.

"Oxo" refers to =O.

"Oxylate" refers to -O " R 77 + wherein R 77 is an organic or inorganic cation.

"Thiol" refers to -SH. hio" refers to -S-.

"Oxime" refers to wherein Rsi is a hydrogen, an alkyl group, an aryl group, an alkylsulfonyl group, an arylsulfonyl group, a carboxylic ester, an alkylcarbonyl group, an arylcarbonyl group, a carboxamido group, an alkoxyalkyl group or an alkoxyaryl group.

"Hydrazone" refers to wherein R' 8 i is independently selected from Rsi , and Rei is as defined herein.

"Hydrazino" refers to H 2 N-N(H)-.

Organic cation" refers to a positively charged organic ion. Exemplary organic cations include alkyl substituted ammonium cations.

"Inorganic cation" refers to a positively charged metal ion. Exemplary inorganic cations include Group I metal cations such as for example, sodium, potassium, magnesium and calcium.

"Hydroxyalkyl" refers to a hydroxy group, as defined herein, appended to an alkyl group, as defined herein.

"Nitrate" refers to -O-NO 2 i.e. oxidized nitrogen.

"Nitrite" refers to -O-NO i.e. oxidized nitrogen.

"Nitro" refers to the group -NO 2 and "nitrosated" refers to compounds that have been substituted therewith.

"Nitroso" refers to the group -NO and "nitrosylated" refers to compounds that have been substituted therewith.

"Nitrile" and "cyano" refer to -CN.

"Halogen" or "halo" refers to iodine (I), bromine (Br), chlorine (CI), and/or fluorine (F). "Imine" refers to -C(=N-Rs 1 )- wherein R 5 i is a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein.

"Amine" refers to any organic compound that contains at least one basic nitrogen atom.

"Amino" refers to -NH2, an alkylamino group, a dialkylamino group, an arylamino group, a diarylamino group, an alkylarylamino group or a heterocyclic ring, as defined herein.

"Alkylamino" refers to R 50 NH-, wherein R50 is an alkyl group, as defined herein. Exemplary alkylamino groups include methylamino, ethylamino, butylamino, and cyclohexylamino. "Arylamino" refers to R 55 NH-, wherein R 55 is an aryl group, as defined elsewhere herein.

"Dialkylamino" refers to R52R53N-, wherein R52 and R53 are each independently an alkyl group, as defined herein. Exemplary dialkylamino groups include dimethylamino, diethylamino and methyl propargylamino.

"Diarylamino" refers to R55R60N-, wherein R 55 and R60 are each independently an aryl group, as defined herein. "Alkylarylamino" or "arylalkylamino" refers to R52R55N-, wherein R 52 is an alkyl group, as defined herein, and R 55 is an aryl group, as defined herein.

"Alkylarylalkylamino " refers to R52R79N-, wherein R 52 is an alkyl group, as defined herein, and R79 IS an arylalkyl group, as defined herein.

"Alkylcycloalkylamino" refers to R52R80N-, wherein R 52 is an alkyl group, as defined herein, and Rao is a cycloalkyl group, as defined herein. "Aminoalkyi" refers to an amino group, an alkylamino group, a dialkylamino group, an arylamino group, a diarylamino group, an alkylarylamino group or a heterocyclic ring, as defined herein, to which is appended an alkyl group, as defined herein. Exemplary aminoalkyi groups include dimethylaminopropyl, diphenylaminocyclopentyl and methylaminomethyl.

"Aminoaryl" refers to an aryl group to which is appended an alkylamino group, an arylamino group or an arylalkylamino group. Exemplary aminoaryl groups include anilino, N-methylanilino and N-benzylanilino.

"Thio" refers to -S-.

"Sulfinyl" refers to -S(O)-.

"Methanthial" refers to -C(S)-.

"Thial" refers to =S. "Sulfonyl" refers to -S(0) 2 "

"Sulfonic acid" refers to -S(0) 2 OR 7 6, wherein R 76 is a hydrogen, an organic cation or an inorganic cation, as defined herein. "Alkylsulfonic acid" refers to a sulfonic acid group, as defined herein, appended to an alkyl group, as defined herein.

"Arylsulfonic acid" refers to a sulfonic acid group, as defined herein, appended to an aryl group, as defined herein.

"Sulfonic ester" refers to -S(0)20Rs8, wherein R 58 is an alkyl group, an aryl group, or an aryl heterocyclic ring, as defined herein. "Sulfonamide-" refers to -S(0)2-N(R 5 i)(R 5 7). wherein R 5 i and R 57 are each independently a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein, or R 5 i and R57 when taken together are a heterocyclic ring, a cycloalkyi group or a bridged cycloalkyi group, as defined herein.

"Alkylsulfonamido" refers to a sulfonamido group, as defined herein, appended to an alkyl group, as defined herein. "Arylsulfonamido" refers to a sulfonamido group, as defined herein, appended to an aryl group, as defined herein.

"Alkylthio" refers to R50S-, wherein R 50 is an alkyl group, as defined herein (preferably a lower alkyl group, as defined herein).

"Arylthio" refers to R55S-, wherein R55 is an aryl group, as defined herein.

"Arylalkylthio" refers to an aryl group, as defined herein, appended to an alkylthio group, as defined herein.

"Alkylsulfinyl" refers to Rso-S(O)-, wherein R50 is an alkyl group, as defined herein. "Alkylsulfonyl" refers to R50-S(O) 2 -, wherein R 50 is an alkyl group, as defined herein.

"Alkylsulfonyloxy" refers to R5o-S(0) 2 -0-, wherein R 50 is an alkyl group, as defined herein.

"Arylsulfinyl" refers to Rs5-S(0)-, wherein R55 is an aryl group, as defined herein. "Arylsulfonyl" refers to R55-S(0) 2 -, wherein R 55 is an aryl group, as defined herein.

"Arylsulfonyloxy" refers to R55-S(0)2-0-, wherein R55 is an aryl group, as defined herein.

"Amidyl" refers to R 5 iC(0)N(R 57 )- wherein R51 and R57 are each independently a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein.

"Ester" refers to RsiC(0)R82- wherein R51 is a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein and Rs2 is oxygen or sulfur. "Carbamoyl" refers to -0-C(0)N(R 5 i)(R57), wherein R51 and R57 are each independently a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein, or R51 and R57 taken together are a heterocyclic ring, a cycloalkyl group or a bridged cycloalkyl group, as defined herein.

"Carboxyl" refers to -C(0)OR 7 6. wherein R 7 6 is a hydrogen, an organic cation or an inorganic cation, as defined herein.

"Carbonyl" refers to -C(O)-.

"Alkylcarbonyl" refers to R52-C(0)-, wherein R 52 is an alkyl group, as defined herein.

"Arylcarbonyl" refers to R55-C(0)-, wherein R 55 is an aryl group, as defined herein.

"Arylalkylcarbonyl" refers to R 5 5-R52-C(0)-, wherein R 55 is an aryl group, as defined herein, and R52 is an alkyl group, as defined herein. "Alkylarylcarbonyl" refers to R52-Rs5-C(0)-, wherein R 55 is an aryl group, as defined herein, and R 52 is an alkyl group, as defined herein. "Heterocyclicalkylcarbonyl" refer to R 7 eC(0)- wherein R 7 s is a heterocyclicalkyl group, as defined herein.

"Carboxylic ester" refers to -C(0)ORs8. wherein R 58 is an alkyl group, an aryl group or an aryl heterocyclic ring, as defined herein.

"Alkylcarboxylic acid" and "alkylcarboxyl" refer to an alkyl group, as defined herein, appended to a carboxyl group, as defined herein.

"Alkylcarboxylic ester" refers to an alkyl group, as defined herein, appended to a carboxylic ester group, as defined herein.

"Alkyl ester" refers to an alkyl group, as defined herein, appended to an ester group, as defined herein. "Arylcarboxylic acid" refers to an aryl group, as defined herein, appended to a carboxyl group, as defined herein.

"Arylcarboxylic ester" and "arylcarboxyl" refer to an aryl group, as defined herein, appended to a carboxylic ester group, as defined herein.

"Aryl ester" refers to an aryl group, as defined herein, appended to an ester group, as defined herein.

"Carboxamido" refers to -C(0)N(R 5 )(R 57 ), wherein R 5 i and R 57 are each independently a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein, or R 5 i and R 57 when taken together are a heterocyclic ring, a cycloalkyi group or a bridged cycloalkyi group, as defined herein. "Alkylcarboxamido" refers to an alkyl group, as defined herein, appended to a carboxamido group, as defined herein.

"Arylcarboxamido" refers to an aryl group, as defined herein, appended to a carboxamido group, as defined herein.

"Urea" refers to -N(R 59 )-C(0)N(R 51 )(R57) wherein R 5 i , R 57 , and R 59 are each independently a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein, or R 51 and R 57 taken together are a heterocyclic ring, a cycloalkyl group or a bridged cycloalkyl group, as defined herein.

"Phosphoryl" refers to -P( 7o)(R7i)( 72). wherein R 7 o is a lone pair of electrons, thial or oxo, and R 1 and R 2 are each independently a covalent bond, a hydrogen, a lower alkyl, an alkoxy, an alkylamino, a hydroxy, an oxy or an aryl, as defined herein.

"Phosphoric acid" refers to -P(0)(OR 5 -i)OH wherein R 51 is a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein.

"Phosphinic acid" refers to -P(0)(R 5 )OH wherein R 51 is a hydrogen atom, an alkyl group, an aryl group or an arylheterocyclic ring, as defined herein.

"Silyl" refers to -Si(R 73 )(R 74 )(R75), wherein R 7 3, R74 and R 7 5 are each independently a covalent bond, a lower alkyl, an alkoxy, an aryl or an arylalkoxy, as defined herein. "Organic acid" refers to compound having at least one carbon atom and one or more functional groups capable of releasing a proton to a basic group. The organic acid preferably contains a carboxyl, a sulfonic acid or a phosphoric acid moeity. Exemplary organic acids include acetic acid, benzoic acid, citric acid, camphorsulfonic acid, methanesulfonic acid, taurocholic acid, chlordronic acid, glyphosphate and medronic acid.

"Inorganic acid" refers to a compound that does not contain at least one carbon atom and is capable of releasing a proton to a basic group. Exemplary inorganic acids include hydrochloric acid, sulfuric acid, nitric acid and phosphoric acid.

"Organic base" refers to a carbon containing compound having one or more functional groups capable of accepting a proton from an acid group. The organic base preferably contains an amine group. Exemplary organic bases include triethylamine, benzyldiethylamine, dimethylethyl amine, imidazole, pyridine and pipy dine. "Independently selected" groups are groups present in the same structure that need not all represent the same substitution. For example, where two substituents are represented as NORA and each R A is said to be independently selected from H, methyl, ethyl, and the like, this means that where one R A is methyl, the other R A may be methyl but could be H or ethyl (or any other recited substitution).

Some of the compounds for use in the methods of the present invention may contain one or more chiral centers and therefore may exist in enantiomeric and diastereomeric forms. The scope of the present invention is intended to cover use of all isomers per se, as well as mixtures of cis and trans isomers, mixtures of diastereomers and racemic mixtures of enantiomers (optical isomers) as well. Further, it is possible using well known techniques to separate the various forms, and some embodiments of the invention may feature purified or enriched species of a given enantiomer or diastereomer.

A "pharmacological composition" refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts thereof, with other chemical components, such as pharmaceutically acceptable carriers and/or excipients. The purpose of a pharmacological composition is to facilitate administration of a compound to an organism. The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. A physiologically acceptable carrier should not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.

A "solvate" is a complex formed by the combination of a solute (e.g., a metalloprotease inhibitor) and a solvent (e.g., water). See J. Honig et al., The Van Nostrand Chemist's Dictionary, p. 650 (1953).

The terms "optical isomer", "geometric isomer" (e.g., a cis and/or trans isomer), "stereoisomer", and "diastereomer" have the accepted meanings (see, e.g., Hawley's Condensed Chemical Dictionary, 11th Ed.). The illustration of specific protected forms and other derivatives of the compounds of the instant invention is not intended to be limiting. The application of other useful protecting groups, salt forms, prodrugs etc., is within the ability of the skilled artisan.

A "prodrug" is a form of a drug that must undergo chemical conversion by metabolic processes before becoming an active, or fully active, pharmacological agent. A prodrug is not active, or is less active, in its ingested or absorbed or otherwise administered form. For example, a prodrug may be broken down by bacteria in the digestive system into products, at least one of which will become active as a drug. Alternatively, it may be administered systemically, such as by intravenous injection, and subsequently be metabolized into one or more active molecules.

DETAILED DESCRIPTION OF THE INVENTION

In accordance with the present invention, it has been found that certain small molecule ligands are capable of reversibly binding non-covalently to the opsin protein and inhibiting the binding of 11-cis-retinal, to an opsin retinal binding pocket. Such interference with retinal binding reduces the formation of visual cycle products, such as all-trans-retinal, and thereby inhibits the production of compounds such as lipofuscin and A2E with resulting reduced risk and occurrence of toxicity that can result from accumulation of these substances. Such compounds, acting as pharmacologic chaperones, are also able to facilitate the proper folding and trafficking of mutant opsins associated with RP. Additionally, by inhibiting 1-cis-retinal binding and rhodopsin formation, the excessive stimulation and resulting activation of rhodopsin caused by exposure of the retina to bright light especially during retinal surgery reduces photocell death.

Certain synthetic retinoids (compounds structurally related to retinol (Vitamin A alcohol)) have been reported to bind to opsin. In the embodiments of the present invention, non-retinoid small molecules (compounds having a molecular weight less than about 000 daltons, less than 800, less than 600, less than 500, less than 400, or less than about 300 daltons) have been found to bind to opsin.

The invention features compositions and methods that are useful for reducing formation of visual cycle products and toxicity associated with the accumulation of such products in vivo, reducing the probability of apoptotic events associated with excessive rhodopsin activation as well as preventing rod cell death due to aberrant processing and trafficking of mutant opsin proteins associated with RP.

Mislocalization of photoreceptor cell visual pigment proteins (opsins) can occur in various ocular diseases, and also with normal aging. In such cases the accumulation of mislocalized opsin leads to the decline in viability of photoreceptor cells. With time this mislocalized opsin accumulation leads to rod and cone cell death, retinal degeneration, and loss of vision. In one aspect, the invention provides a method of correcting mislocalized opsin within a photoreceptor cell, comprising contacting a mislocalized opsin protein with an opsin-binding agent that binds reversibly and/or non-covalently to said mislocalized opsin protein, thereby promoting correct intracellular processing and transport of said opsin protein. Such opsin-binding agent is referred to as a "Productive Chaperone."

Such correction of mislocalization reduces photoreceptor cell stress, preventing photoreceptor cell decline in viability and death in various diseases of vision loss, and in normal age-related decline in dim-light and peripheral rod-mediated vision, central cone-mediated vision, and loss of night vision.

In another aspect of the invention, the opsin-binding agent promotes the degradation of the mislocalized opsin protein. This type of opsin-binding agent is referred to as a "Counterproductive", Shipwreck", or "Destructive Chaperone."

Enhancing the degradation of the mislocalized opsin by such an agent reduces the amount of mislocalized protein, thereby relieving photoreceptor cell stress, preventing decline in viability and death of photoreceptor cells in diseases of vision loss, as well as in normal age-related decline in dim-light and peripheral rod-mediated vision, central cone-mediated vision, and loss of night vision.

In embodiments of the foregoing, the ocular protein mislocalization disorder is one or more of wet or dry form of macular degeneration, retinitis pigmentosa, a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity, retinitis pigmentosa, normal vision loss related aging and normal loss of night vision related to aging.

Opsin, the GPCR (G-protein coupled receptor) responsible for vision, readily regenerates with 11-cis-retinal to form the visual pigment rhodopsin. The pigment is generated by formation of a protonated Schiff base between the aldehyde group of 11-cis-retinal and the ε-amino group of L-lysine in opsin (Matsumoto and Yoshizawa, Nature 1975 Dec 11 ;258(5535):523-6). Thus, the present invention provides compositions and methods of use of small molecule compounds that bind to wild type and mutant opsins and compete with, or other wise prevent, 11-cis-retinal from combining with opsin to form rhodopsin and thereby inhibit formation of 11-cis-retinal and other visual cycle products.

Binding to this site may be predicted by the efficiency upon which the ligand is able to displace and/or replace the waters in the various hydration sites in the 11-cis retinal binding pocket as defined by the water map technology. Hydration sites labeled with an "R" (Figure 1 shows hydration sites as circles or spheres) that are occupied by waters that are predicted to have hydrogen bonding interactions with the protein. Thus, ligands that displace these waters will ideally have functionality suitably oriented when the ligand binds to replace those hydrogen bonds that are broken in the process of the compound occupying the binding pocket.

In accordance with the present invention, ligand binding potency is enhanced by compounds that efficiently displace highly unstable waters from the opsin binding pocket. Occupation of the pocket by a pharmacologic chaperone creates interactions between the ligand and the protein that induces the proper folding and/or stabilization of the native 3-dimentional conformation of the protein that leads to it being properly processed and trafficked to its proper location in the cell membrane.

Alternatively, hydration sites labeled with a "D" (Figure 1) locate waters that are in hydrophobic environments and therefore it is optimal for the binding compound to displace all of these waters with nonpolar substituents that compliment the hydrophobic environment of the protein. Thus, displacing waters in hydrophobic environments while replacing the hydrogen bonds of waters in hydration sites predicted to have hydrogen bonding interactions with the protein with functionality on the ligand that can act as water mimetics when these waters are displaced leads to optimal potency and efficacy. Alternatively, displacing waters in hydration sites labeled with a "D" in Figure 1 and leaving those waters in hydration sites labeled with an "R: (shown in Figure 1) unperturbed such that their environment with the ligand bound does not adversely affect the intrinsic stability of these waters in the pocket in the absence of ligand occupation leads to potent and efficacious compounds. The hydration sites are predicted locations of waters in the absence of a ligand based on the hydration map. Binding of a ligand of the invention may follow one of four possible mechanisms: (i) displacing a water occupying a hydration site, (ii) replacing a hydrogen bond between protein and a water in a hydration site by a functionality of the ligand, (iii) binding of a ligand and leaving a water in the hydration site intact, and (iv) forming an extended hydrogen bonding network with the water in a hydration site while not displacing it.

In one aspect, the invention provides opsin binding ligands of Formula I and pharmaceutically acceptable salts, solvates and hydrates thereof,

wherein X--Y is:

1) C(R 4 )-C(0),

2) C(R 4 )-C(R 5 )(OR 1 ),

3) C=C(R 5 ),

4) C(R 4 )-C(H)(R 5 ),

5) C(R 4 )-C(D)(R 5 ),

6) C(R )-C(F)(R 5 )

7) C(R 4 )-0,

8) C(R 4 )-S(0) n , wherein n

9) C=N,

10) C(R 4 )-N(R 6 ),

11) C(R )-C(=N-OR 7 ), or

12) ) C(R 4 )-C(=N-CN)

R 1 and R 2 are independently:

1) hydrogen,

2) -CH 3 , or

3) -CH 2 CH 3 ; R 3 is:

1) hydrogen,

2) -CH 3l

3) -CH2CH3, or 4) deuteron;

R 4 is:

1) hydrogen;

2) -CH 3 ;

3) halogen; or

4) deuteron;

R 5 is:

1) hydrogen,

2) deuteron, 3) lower alkyl,

4) lower alkenyl,

5) lower alkynyl,

6) aryl,

7) nitrite, or 8) fluoro;

R 6 is:

1) -C(0)-R 8 ,

2) -S(0) 2 -R 10 , 3) lower alkyl,

4) lower alkenyl,

5) lower alkynyl, or

6) hydrogen; R 7 is:

1) hydrogen, or

2) lower alkyl; R° is:

1) hydrogen,

2) lower alkyl,

3) aryl,

4) -0-R 9 , or

5) -N(R 7 )(R 9 );

R 9 is:

1) hydrogen, 2) lower alkyl,

3) lower alkenyl,

4) lower alkynyl, or

5) aryl;

2)

3)

4)

5)

Ri. Rm- R n and R 0 are each independently:

1 ) C(R e ), or

2) nitrogen;

with the proviso that at least one of R|, R m , R n, or R 0 must be selected as nitrogen;

R a , and R , are each independently:

1) hydrogen;

2) -CH 3 ; or

with the proviso that if and/or R 2 are selected as hydrogen then R, and R b must be selected from methyl and ethyl;

Rc, and R d , are each independently:

1 ) hydrogen;

2) alkoxy;

3) lower alkyl;

4) lower alkenyl;

5) haloalkyl,

6) hydroxy,

7) fluoro, or 8) deuteron;

Ro is:

1) hydrogen,

2) lower alky I,

3) alkoxyalkyl, or

4) fluoro; and where in some embodiments R c and R 0 may be taken together as:

1) oxo,

2) =CH 2 , or

3) lower alkenyl;

R e , Rf, R g and R h are each independently:

1) hydrogen,

2) deuteron,

3) lower alkyl,

4) halogen,

5) nitro,

6) alkoxy,

7) nitrile,

8) carboxamido,

9) urea,

10) alkylcarbonyl,

11) arylcarbonyl,

12) carbamoyl,

13) amidyl, or

14) amino;

Z is:

1) oxygen, or

2) sulfur. In another aspect, the invention provides opsin binding ligands of Formula II and pharmaceutically acceptable salts thereof:

Formula II wherein R, and R j are each independently

1 ) hydrogen,

2) deuteron,

3) fluoro,

4) alkoxy,

5) hydroxy I, or

6) lower alkyl;

Ri and R j taken together are:

1) oxo,

2) =N-OR 7 , or

3) =N-CN;

R p and R q are each independently:

1) hydrogen,

2) methyl, or

3) ethyl;

B is:

1)

) cycloalkyl; ) hydrogen, ) lower alkyl, ) halogen, ) nitro, ) alkoxy, ) nitrile, 7) carboxamido,

8) urea,

9) alkylcarbonyl,

10) arylcarbonyl,

11) carbamoyl,

12) amidyl, or

13) amino; wherein n, R 1 , R 2 R 3 , R c , R d , R e , Rf. Rg, n, Ri, Rm. R n , > R 0 and Z are as defined herein for Formula I.

In preferred embodiments, the compound has the structure of Formula

I wherein X^Y is C(R )-C(0), C(R 4 )-C(H)OH, C=C(R 5 ), C(R )-C(=N-OR 7 ) or C(R 4 )-C(=N-CN) and wherein R and R 2 are hydrogen, R a and R b are independently methyl or ethyl, more preferably wherein both of R a and R b is methyl, R 3 is hydrogen or deuteron, most preferably wherein R 3 is deuteron and wherein R 4 and R 5 are each independently hydrogen or methyl, more preferably wherein at least one of R 4 and R 5 is hydrogen, most preferably wherein both R 4 and R 5 are hydrogen.

In preferred embodiments, the compound has the structure of Formula

II wherein R, and R j taken together are oxo, =N-OR 7 , or =N-CN, or wherein Ri is hydroxy and R j is hydrogen and wherein R 1 and R 2 are each independently methyl or ethyl, more preferably wherein both of R 1 and R 2 is methyl, and R 3 is hydrogen or methyl, most preferably wherein R 3 methyl.

In preferred examples of the invention, the compound has the structure of Formula I wherein A (i.e., the A ring) is an aryl group and one or more of R , R 2 is methyl, ethyl or hydrogen, more preferably methyl or hydrogen, and most preferably where each of R 1 and R 2 is a hydrogen, and R 3 is hydrogen, deuteron or methyl, more preferably hydrogen or deuteron and most preferably where R 3 is deuteron. In other specific embodiments, R a and R b are independently hydrogen, methyl or ethyl, preferably methyl or ethyl, and most preferably where both R a and R b are methyl. R c and R d are hydrogen, lower alkyi or alkoxy, more preferably hydrogen or lower alkyi, most preferably hydrogen or methyl. In preferred examples of the invention, the compound has the structure of Formula II wherein B (i.e., the B ring) is an aryl group, R 1 and R 2 are methyl or ethyl more preferably methyl and R 3 is hydrogen, methyl, or ethyl, more preferably a hydrogen or methyl more preferably methyl and most preferably where each of R , R 2 and R 3 is a methyl. In other specific embodiments, R p and R q are independently hydrogen or methyl, preferably hydrogen, R c and R d are hydrogen, lower alkyi or alkoxy, more preferably hydrogen or lower alkyi, most preferably hydrogen or methyl. In other specific embodiments, R, and Rj are each independently hydrogen or hydroxyl or taken together are oxo, =N- OR 7 or =N-CN, more preferably R, is hydrogen and R j is hydroxyl or R, and Rj taken together are oxo.

In specific embodiments the opsin binding compound of Formula I and Formula II are (wherein each compound number corresponds to the number of the example where it was prepared):

(±)-(4a ,9aS)-7-lsopropyl-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aH)-one (Compound 1);

(±)-(4aR,9aS)-6-Hydroxy-7-isopropyl-1 ,1 ,4a-tnmethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aH)-one (Compound 2);

(±)-(4aR,9aS)-6-Methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9a/-/)-one (Compound 3);

(+)-(4aR,9aS)-6-Methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9a/- )-one (Compound 3a);

(-)-(4aS,9aR)-6-Methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 3b);

(±)-(4aR,9aS)-6-Hydroxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 4); (±)-(4aR,9aS)-6-Ethoxy-1 ,1 ,4a-trimethyl-2 I 3,4,4a-tetrahydro-1/-/-fluoren- 9(9aH)-one (Compound 5);

(±)-(4af?,9aS)-1 ,1 ,4a-Trimethyl-6-propoxy-2,3,4,4a-tetrahydro-1/-/-fluoren- 9(9aH)-one (Compound 6);

(±)-(4aR,9aS)-6-(Allyloxy)-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 7);

(±)-(4aR,9aS)-1 ,1 ,4a-Trimethyl-6-(prop-2-ynyloxy)-2,3,4,4a-tetrahydro-1/-/- fluoren-9(9aH)-one (Compound 8);

(±)-(4af?,9aS)-6-(Benzyloxy)-1 ,1 ,4a-trimethyl-2,3 l 4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 9);

(iJ^aR^aSJ-l .l ^a-Trimethyl-g-oxo^, 3,4,4a, 9,9a-hexahydro-1/-/-fluoren-6-yl acetate (Compound 10);

(±)-(4a 9S,9aS)-6-Methoxy-1 ,1 ,4a-trimethyl-2 l 3,4,4a,9,9a-hexahydro-1H- fluoren-9-ol (Compound 11);

(±)-6-Methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluorene (Compound 12);

(±)-(4aR,9aS)-6-Methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1ry- fluorene (Compound 13);

(±)-(4af?,9aS)-6-(6-Chloropyridazin-3-yloxy)-1 ,1 ,4a-trimethyl-2, 3,4,4a- tetrahydro-1 7-fluoren-9(9a Y)-one (Compound 14);

P-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 17);

P-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 18);

(±)-(4af?,9aS)-1 ,1 ,4a,6-Tetramethyl-2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)- one (Compound 19);

(±)-(4a ,9aS)-1 ,1 ,4a,6-Tetramethyl-2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)- one oxime (Compound 20);

(±)-(4a ?,9aS)- ,1 ,4a,6-Tetramethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)- one O-methyl oxime (Compound 21);

(±)-(4aR,9f?,9aS)-1 ,1 ,4a,6-Tetramethyl-2, 3,4,4a, 9,9a-hexahydro-1H-fluoren-9- ol (Compound 22);

(±)-1 ,1 ,4a,6-Tetramethyl-2,3,4,4a-tetrahydro-1/-/-fluorene (Compound 23); (±)-(4aR,9aS)-1 ,1 ,4a,6-Tetramethyl-2,3,4,4a,9,9a-hexahyclro-1 /-/-fluorene (Compound 24);

1-(Methoxy(2,6,6-trimethylcyclohex-1-enyl)methyl)-4-methylbe nzene

(Compound 25);

l .l ^a.e.g-Pentamethyl^.a^^a-tetrahydro-I H-fluorene (Compound 26); (±)-(4aR,9S,9aS)-1 ,1 ,4a,6,9-Pentamethyl-2,3,4 l 4a,9,9a-hexahydro-1H- fluorene (Compound 27);

Phenyl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 28);

Phenyl(2,6,6-t methylcyclohex-1-enyl)methanone (Compound 29);

(±)-(4aR,9aS)-1 , 1 ,4a-Trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)-one (Compound .30);

(±)-(4aR,9R,9aS)-1 , 1 ,4a-Trimethyl-2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9-ol (Compound 31 );

(±)-1 , 1 ,4a-Trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 32);

(±)-(4aR,9aS)-1 ,1 ,4a-Trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)-one oxime (Compound 33);

( J^aR^aSJ-I ^a-Trimethyl^^^^a^^a-hexahydro-I H-fluorene

(Compound 34);

(±)-(4aR,9S,9aS)-1 ,1 ,4a,9-Tetramethyl-2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9- ol (Compound 35);

(±)-1 ,1 ,4a,9-Tetramethyl-2,3,4,4a-tetrahydro-1 /-/-fluorene (Compound 36); (±)-(4aR,9aS)-1 ,1 ,4a-Trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)-one O- methyl oxime (Compound 37);

(Methoxy(2,6,6-Trimethylcyclohex-1-enyl)methyl)benzene (Compound 38); (4-Chlorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 39); (4-Chlorophenyl)(2,6,6-trimethylcyclohex-1 -enyl)methanone (Compound 40); (±)-6-Chloro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 41 ); (±)-(4aR,9aR)-6-Chloro-1 ,1 ,4a-tnmethyl-2,3,4,4a,9,9a-hexahydro-1H-fluorene (Compound 42);

(±)-(4aR,9R,9aR)-6-Chloro-1 , ,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 43);

(±)-(4aR,9R,9aR)-6-Chloro-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 44); (±)-(4a ?,9 ?,9a ?)-6-Chloro-9-ethyl-1 , 1 ,4a-trimethyl-2,3 A4a,9,9a-hexahydro- 1 /-/-fluoren-9-ol (Compound 45);

(±)-(4-Ethylphenyl)(2,6,6-trimethylcyclohex-1 -enyl)methanol (Compound 46); (4-Ethylphenyl)(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 47); (±)-(4aR,9aS)-6-Ethyl-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 48);

(±)-(4a ,9R9aS)^-Ethyl-1 ,4a-trimethyl-2,3,4,4a,9,9a-hexahydr(>-1 H- fluoren-9-ol (Compound 49);

(±)-6-Ethyl-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 /-/-fluorene (Compound 50); (±)-6-Ethyl-1 ,1 ,4a,9-tetramethyl-2,3,4,4a-tetrahydro-1 /-/-fluorene (Compound 51);

(±)-(4aR,9S,9aS)-6-Ethyl- ,1 ,4a,9-tetramethyl-2,3 A4a,9,9a-hexahydro-1 H- fluorene (Compound 52);

(±)-(3,4-Dimethylphenyl)(2,6,6-trimethylcyclohex-1-enyl)met hanol (Compound 53);

(3,4-Dimethylphenyl)(2,6,6-trimethylcyclohex-1-enyl)methanon e (Compound 54);

(±)-(4a 9aS)-1 ,1 ,4a l 6 l 7-Pentamethyl-2,3 l 4,4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 55a);

(±)-(4a ?,9aS)-1 ,1 ,4a l 5,6-Pentamet yl-2 l 3 l 4 l 4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 55b);

(±)-(4-Fluorophenyl)(2,6,6-trimethylcyclohex-1 -enyl)methanol (Compound 56); (4-Fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 57); (±)-(4af?,9aS)-6-Fluoro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 58);

(±)-(4aft,9R,9aS)-6-Fluoro-1 , 1 ^a-trimethyl^^ a^.ga-hexahydro-l H- fluoren-9-ol (Compound 59);

(±)-(4a 9fi,9aS)-6-Chloro-1 , 1 ,4a-trimethyl-2, 3,4,4a, 9, 9a-hexahydro-1/-/- fluoren-9-ol (Compound 60);

(±)-(4af?,9aS)-6-Chloro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 61 );

(±)-(3-Chloro-4-methylphenyl)(2,6,6-trimethylcyclohex-1-eny l)methanol (Compound 62); (3-Chloro-4-methylphenyl)(2,6,6-trimethylcyclohex-1 -enyl)methanone

(Compound 63);

(±)-(4aR,9aS)-5-C loro-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 64a);

(±)-(4aR,9aS)-7-Chloro-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 64b);

(±)-(4aR,9R,9aS)-7-Chloro-1 ,1 ,4a,6-tetramethyl-2, 3,4,4a, 9, 9a-hexahydro-1 H- fluoren-9-ol (Compound 65);

(±)-(4aR,9R,9aS)-5-Chloro-1 ,1 ,4a,6-tetramethyl-2,3,4,4a,9,9a

hexahydro-1 /- -fluoren-9-ol (Compound 66);

(±)-(3-Fluoro-4-methoxyphenyl)(2,6,6-trimethylcyclohex-1-en yl)methanol (Compound 67);

(±)-(3-Fluoro-4-methoxyphenyl)(2,6,6-trimethylcyclohex-1-en yl)methanone (Compound 68);

(±)-(4a/?,9aS)-7-Fluoro-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1/-/ fluoren-9(9aH)-one (Compound 69);

(±)-(4aR,9/?,9aS)-7-Fluoro-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol (Compound 70);

(±)-(Thiophen-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 71 ); thiophen-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 72); (±H3bR aS)-3b -TnmetU/\-5^ a^ah 0TO-3bH-\ndeno[2^- b]thiophen-8(4H)-one (Compound 73);

(ti-iSbRJaS.eRVSb J-Trimethyl^.S.e^ a.e-hexahydro-SbH-indeno^.l- b]thiophen-8-ol (Compound 74);

(±)-Thiophen-3-yl(2,6,6-trimethylcyclohex-1-enyl)methano l (Compound 75); Thiophen-3-yl(2,6,6-trimethylcyclohex-1 -enyl)methanone (Compound 76); (±)-3b,7,7-Trimethyl-4,5,6,7-tetrahydro-3bH-indeno[2,1-b]th iophene

(Compound 77);

(±)-(3bR,7aS,8R)-3b,7,7-Thmethyl-4,5,6,7,7a,8-hexahydro-3bH -indeno[2,1- 6]thiophen-8-ol (Compound 78);

(±)-(4aS,8aR)-5, 5,8a-Trimethyl-4a, 5,6,7,8, 8a-hexahydro-4/-/-indeno[1 ,2- £>]thiophen-4-one (Compound 79); (±)-(4R^aS,8aR)-5,5,8a-Trimethyl-4a,5,6,7,8,8a-hexahyclro-4 H-incleno[1 ,2- b]thiophen-4-ol (Compound 80a);

(±)-(4S,4aS,8aR)-4a,5,5,8a-Tetramethyl-4a,5,6,7,8,8a-hexahy dro-4H- indeno[1 ,2-t»]thiophen-4-ol (Compound 80b);

Furan-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 81);

(±)-(4aS,8af?)-5,5,8a-Trimethyl-4a,5,6,7,8,8a-hexahydro- 4/-/-indeno[1 ,2- £>]furan-4-one (Compound 82);

(±)-(4R,4aS,8aR)-5,5,8a-Trimethyl-4a,5,6,7,8,8a-hexahydro-4 H-indeno[1 ,2- jb]furan-4-ol (Compound 83);

Furan-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 84);

Pyridin-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 85);

Pyridin-3-yl(2,6,6-trimet ylcyclohex-1-enyl)methanone (Compound 86);

1 ,1 ,4a,6-Tetramethyl-2,3,4,4a-tetrahydro-1 H-carbazole (Compound 87);

(±)-(4aS,9aS)-1 ,1 ,4a,6-Tetramethyl-2,3,4,4a,9,9a-hexahydro-1 H-carbazole (Compound 88);

(±)-(4aS,9aS)-1 ,1 ,4a,6,9-Pentamethyl-2, 3,4,4a, 9,9a-hexahydro-1 H-carbazole

(Compound 89);

1 ,1 ,4a-Trimethyl-2,3,4,4a-tetrahydro-1 H-carbazole (Compound 90);

(±)-(4aS,9aS)-1 ,1 ,4a-Trimethyl-2,3,4,4a,9 l 9a-hexahydro-1 H-carbazole (Compound 91);

(±)-(4aS,9aS)-1 ,1 , 4a, 9-Tetramethyl-2, 3,4,4a, 9, 9a-hexahydro-1 H-carbazole (Compound 92);

(±)-(4aS,9aS)-6- et oxy-1 -1 ,4a-trimethyl-2,3,4,4a,9,9a-hexa ydro-1 H- carbazole (Compound 93);

(±)-(4aS,9aS)-6-Methoxy-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro-1H- carbazole (Compound 94);

(3-Methoxyphenyl)(2,6,6-Trimethylcyclohex-1 -enyl)methanol (Compound 95); (3-Methoxyphenyl)(2,6,6-Trimethylcyclohex-1-enyl)methanone (Compound 96);

(±)-(4a ?,9aS)-7-Methoxy-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 97);

(±)-(4aR,9S,9aS)-7-Methoxy-1 , 1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro-1 H- fluoren-9-ol (Compound 98a); (±)-(4aR,9R,9aS)-7-Methoxy-1 , 1 ,4a-trimethyl-2, 3,4,4a, 9, 9a-hexahydro-1H- fluoren-9-ol (Compound 98b);

fluoren-9-ol (Compound 99);

(Perfluor0phenyl)(2,6,6-trimethylcyclo ex-1-enyl)met anol (Compound 100); (Perfluorophenyl)(2 > 6,6-trimethylcyclohex-1-enyl)methanone (Compound 101);

6-Fluoro- ,1 ,4a-trimethyl-2,3,4,4a-tetra ydro-1 H-carbazole (Compound 02); (4-Methoxy-3-methylphenyl)(2,6,6-trimethylcyclohex-1-enyl)me thanol

(Compound 103);

(4-Methoxy-3-methylphenyl)(2,6,6-trimethylcyclohex-1-enyl)me thanone (Compound 104);

(±)-(4aR,9aS)-6-Methoxy-1 ,1 ,4a,7-tetramethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 105);

(±)-5,5,8a-Trimethyl-6,7,8,8a-tetrahydro-5H-indeno[1 ,2-i>]thiophene

(Compound 106);

(4-(Trifluoromethyl)phenyl)(2,6,6-trimethylcyclohex-1-enyl)m ethanol

(Compound 107);

(4-(Trifluoromethyl)phenyl)(2,6,6-trimethylcyclohex-1-enyl)m ethanone

(Compound 108);

(±)-(4aR,9aS)-1 , 1 ,4a-Trimethyl-6-(trifluoromethyl)-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one (Compound 109);

(3-Fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 1 10); (3-Fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 1 1 1); (±)-(4aR,9aS)-7-Fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9a/-/)-one (Compound 1 12a);

(±)-(4af?,9aS)-5-Fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 1 12b);

(±)-(4af?,9 ,9aS)-7-Fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 1 13);

(3-Chloro-4-fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)met hanol

(Compound 1 14); (3-Chloro-4-fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)met hanone

(Compound 115);

(±)-(4af? 1 9aS)-7-Chloro-6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aH)-one (Compound 116a);

(±)-(4aR,9aS)-5-Chloro-6-fluoro-1 , ,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aH)-one (Compound 116b);

(±)-(4aR,9R,9aS)-7-Chloro-6-fluoro-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro- 1 H-fluoren-9-ol (Compound 117);

(±)-(4aR,9R,9aS)-5-Chloro-6-fluoro-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro- 1 H-fluoren-9-ol (Compound 18);

o-tolyl(2,6,6-Trimethylcyclohex-1-enyl)methanol (Compound 119);

o-tolyl(2,6,6-Trimethylcyclohex-1-enyl)methanone (Compound 120);

(±)-(4aR,9aS)-1 ,1 ,4a,8-Tetramethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 121);

(±)-(4aR,9R,9aS)-1 , 1 ,4a, 8-Tetramethyl-2, 3,4,4a, 9,9a-hexahydro-1H-fluoren-9- ol (Compound 122);

(±)-(4aR,9R,9aS)-6-Chloro-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro-1H- fluoren-9-ol (Compound 123);

(3,4-Difluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 124);

(3,4-Difluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanon e (Compound 125);

(±)-(4aR,9aS)-6,7-Difluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9a/-/)-one (Compound 126);

(3,4,5-Trifluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)me thanol (Compound

127) ;

(3,4,5-Trifluorophenyl)(2,6,6-tr ' imet ylcyclohex-1-enyl)methanone (Compound

128) ;

(±)-(4aR,9aS)-5,6,7-Trifluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluoren- 9(9aH)-one (Compound 129);

(4-(Trifluoromethoxy)phenyl)(2,6,6-trimethylcyclohex-1-enyl) methanol

(Compound 130); (4-(Trifluoromethoxy)phenyl)(2,6,6-trimethylcyclohex-1-enyl) methanone (Compound 131);

(±)-1 , 1 ^a-Trimethyl-e-itrifluoromethoxy^.S^^a-tetrahydro-l H-fluorene (Compound 132);

(±)-(4a ,9a )-1 A ,4a-Trimethyl-6-(trifluoromethoxy)-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one (Compound 133);

(±)-(4aR,9aS)-1 ,1 ,4a-Trimethyl-6-(trifluoromethoxy)-2 l 3,4,4a,9,9a-hexahydro- 1H-fluorene (Compound 34);

(±)-6-Fluoro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 134); (Compound 135);

(±)-(4aR,9aS)-6-Fluoro-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro-1 H-fluorene (Compound 136);

Cyclohexyl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 137);

Cyclohexyl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 138); Cyclopentyl(2,6,6-trimethylcyclohex-1-enyl)methanol (Compound 139);

Cyclopentyl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 140); (±)-(4aR,9R,9aS)-1 ,1 ,4a-Trimethyl-6-(trifluoromethoxy)-2, 3,4,4a, 9,9a- hexahydro-1 H-Fluoren-9-ol (Compound 141 );

(±)-(4aR,9aS)-1 ,1 ,4a-Trimethyl-6-(trifluoromethoxy)-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one (Compound 142);

(±)-(4aR,9S,9aS)-1 , 1 ,4a, 9-Tetramethyl-6-(trifluoromethoxy)-2, 3,4,4a, 9,9a- hexahydro-1 H-fluoren-9-ol (Compound 143);

(±)-(4aR,9R,9aR)-1 ,1 ,4a,9-Tetramethyl-6-(trifluoromethoxy)-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol (Compound 144);

(±)-(4aR,9R,9aR)- ,1 ,4a-Trimethyl-6-(trifluoromethoxy)-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol (Compound 145);

(±)-(4aS,9aS)-6-Fluoro-1 , 1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro-1 H- carbazole (Compound 146);

(±)-(4aR,9aR)-4,4,4a,9-Tetramethyl-4,4a,9,9a-tetrahydro-1 /-/-carbazol-2(3H)- one (Compound 147);

(±)-(4aR,9aR)-4,4,4a,9-Tetramethyl-2, 3,4,4a, 9, 9a-hexahydro-1 H-carbazole (Compound 148); (±)-(4aR,9aR)-4,4,4a,9-Tetramethyl-2,3,4 l 4a,9,9a-hexahydro-1 H-carbazol-2-ol (Compound 149);

(±)-(4aR,9aR)-4,4,4a,9-Tetramethyl-2-methylene-2 l 3,4,4a,9,9a-hexahydro- 1 H-carbazole (Compound 150);

(±)-(4aR,9R,9aS)-1 ,1 ^a-Trimethyl-e-itrifluoromethy ^^^^a^^a- hexahydro-1 H-fluoren-9-ol (Compound 151);

(±)-(4aR l 9aR)-2-Methoxy-4,4,4a,9-tetramethyl-2,3,4 l 4a,9,9a-hexahydro-1 H- carbazole (Compound 152);

(±)-(4aS,9aS)-6-Fluoro-4,4-dimethyl-2 l 3,4 > 4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 155);

(±)-(4aS,9aS)-6-Fluoro-4,4,4a-trimethyl-2,3,4,4a-tetrahydro -1 H-fluoren- 9(9aH)-one (Compound 158);

(±)-(4aR,9R,9aS)-1 , 1 ,4a-Trimethyl-6-(trifluoromethyl)-2,3,4,4a,9,9a- hexahydro-1 /-/-fluoren-9-ol (Compound 159);

(2,6,6-Trimethylcyclohex-1-enyl)methyl)benzene (Compound 160);

(±)-6-Fluoro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 161 );

fluoren-9-ol (Compound 162);

(±)-(4aR,9aR)-4,4,9-Trimethyl-4a-propyl-4,4a,9,9a-tetrah ydro-1 /-/-carbazol- 2(3H)-one (Compound 163);

(±)-((4aR,9aR)-4,4,4a,9-Tetramethyl-2,3,4,4a,9,9a-hexahydro -1 /-/-carbazol-2- yl)methanol (Compound 164);

(±)-(4aR,9aR)-2-(Methoxymethyl)-4,4,4a,9-tetramethyl-2, 3,4,4a, 9,9a- hexahydro- H-carbazole (Compound 165);

(±)-(4aS,9S,9aS)-6-Fluoro-4,4-dimethyl-2, 3,4,4a, 9, 9a-hexahydro-1 H-fluoren- 9-ol (Compound 166);

(±)-(4aR,9S,9aS)-6-Fluoro-1 , 1 ,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 167);

(±)-(4aR,9aR)-6-Fluoro-1 , 1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren- 9(9aH)-one (Compound 168);

(±)-(4aR,9R,9aR)-6-Fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 169); (±)-(4aR,9R,9aR)-6-FluoiO-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 170);

(±)-(4aR,9aR)-2,4 ) 4,4a,9-Pentamethyl-2,3,4,4a,9,9a-hexahydro-1 H-carbazol-

2-ol (Compound 171 );

(3,4-Dihydro-2H-pyran-6-yl)(2,6,6-trimethylcyclohex-1-eny l)methanol

(Compound 172);

(3,4-Dihydro-2/-/-pyran-6-yl)(2,6,6-trimethylcyclohex-1-enyl )methanone

(Compound 173);

(iJ^b eaSHb.e.e-Trimethyl-S^^b.S.ej.e.ea-octahydroindenop.l- b]pyran-9(2/-/)-one (Compound 174);

(±)-(4bR,8aS,9R)-4b > 8,8-T methyl-2,3,4,4b,5,6,7,8,8a,9- decahydroindeno[2,1-b]pyran-9-ol (Compound 175);

(±)-(4aR,9aR)-9-Ethyl-4,4 l 4a-trimethyl-4,4a,9,9a-tetrahydro-1 H-carbazol-

2(3H)-one (Compound 176);

6-Fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 181);

(±)-(4aS,9aS)-6-Fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1 /-/-fluoren-9(9aH)- one oxime (Compound 182);

6-Fluoro-4,4-dimethyl-2, 3,4,4a, 9, 9a-hexahydro-1 H-fluorene (Compound 186); (±)-(4aS,9aS)-6-Fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)- one O-methyl oxime (Compound 187);

(±)-(4aS,9aS)-4a-Deutero-6-fluoro-4,4-dimethyl-2,3,4,4a-tet rahydro-1 H- fluoren-9(9aH)-one (Compound 189);

(±)-(4aR,9aS)-4a-Ethyl-6-fluoro-1 ,1-dimethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluoren-9-ol (Compound 190);

4-(Hydroxy(2,6,6-trimethylcyclohex-1 -enyl)methyl)benzonitrile (Compound 191 );

4-(2,6,6-Trimethylcyclohex-1 -enecarbonyl)benzonitrile (Compound 192);

Pyrimidin-5-yl(2,6,6-trimethylcyclohex-1 -enyl)methanol (Compound 194); (3-Fluoro-4-(trifluoromethoxy)phenyl)(2,6,6-trimethylcyclohe x-1-enyl)methanol (Compound 195);

(±)-(4aS,9aS)-6-Fluoro-4,4,8-trimethyl-2,3,4,4a-tetrahydro- 1 H-fluoren-9(9a/-/)- one (Compound 196); (±)-(4aS I 9S l 9aS)-6-Fluoro-4 ( 4 I 8-trimethyl-2,3 l 4,4a l 9,9a-hexahydro-1H- fluoren-9-ol (Compound 197);

Pyrimidin-5-yl(2,6,6-trimethylcyclohex-1-enyl)methanone (Compound 199); (3-Fluoro-4-(trifluoromethoxy)phenyl)(2,6,6-trimethylcyclohe x-1- enyl)methanone (Compound 200);

(Tetrahydro-2H-pyran-4-yl)(2,6,6-trimethylcyclohex-1-enyl)me thanol

(Compound 201);

(±)-(4aft,9aS)-1 , 1 ,4a-Trimethyl-9-oxo-2, 3,4,4a, 9,9a-hexahydro-1 H-fluorene- 6-carbonitrile (Compound 202);

(±)-(4aR,9R,9aS)-9-Hydroxy-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a-hexahydro-1H- fluorene-6-carbonitrile (Compound 203);

(Tetrahydro-2H-pyran-4-yl)(2,6,6-trimethylcyclohex-1-enyl)me thanone

(Compound 204);

2-Fluoro-4-(hydroxy(2,6,6-trimethylcyclohex-1-enyl)methyl) benzonitrile (Compound 205);

2-Fluoro-4-(2,6,6-trimethylcyclohex-1 -enecarbonyl) benzonitrile (Compound 206);

(±)-(4aR,9aS)-7-Fluoro-1 ,1 ,4a-trimethyl-9-oxo-2,3,4,4a,9,9a-hexahydro-1H- fluorene-6-carbonitrile (Compound 207);

7-Fluoro-1 , 1 ,4a-trimethyl-6-(trifluoromethoxy)-2,3,4,4a-tetrahydro-1 H-fluorene (Compound 208);

(6,6-Dimethylcyclohex-1 -enyl)(4-fluorophenyl)methanol (Compound 209); (6,6-Dimethylcyclohex-1 -enyl)(4-fluorophenyl)methanone (Compound 210); (2-Deutero-6,6-dimethylcyclohex-1-enyl)(4-fluorophenyl) methanone (Compound 211);

(2-Deutero-6,6-dimethylcyclohex-1-enyl)(3,4-difluorophenyl) methanone (Compound 212);

(±)-(4a/?,9aS)-7-Fluoro-1 ,1 ,4a-trimethyl-6-(trifluoromethoxy)-2,3,4,4a- tetrahydro-1/-/-fluoren-9(9aH)-one (Compound 213);

(±)-(4aR,9R,9aS)-7-Fluoro-9-hydroxy-1 ,1 ,4a-thmethyl-2,3,4,4a,9,9a- hexahydro-1H-fluorene-6-carbonitrile (Compound 214);

4-((3,3-Dimethylcyclohex-1-enyl)(hydroxy)methyl)benzonitr ile (Compound 215); 4-(3,3-Dimethylcyclohex-1-enecarbonyl)benzonitrile (Compound 216);

4-((3,3-Dimethylcyclohex-1-enyl)(hydroxy)methyl)-2-fluoro benzonitrile

(Compound 217);

4-(3,3-Dimethylcyclohex-1-enecarbonyl)-2-fluorobenzonitrile (Compound 218);

4-Chlorophenyl)(3,3-dimethylcyclohex-1-enyl)methanol (Compound 219); (4-Chlorophenyl)(3,3-dimethylcyclohex-1-enyl)methanone (Compound 220); (±)-(4aR,9R,9aS)-7-Fluoro-1 , 1 ,4a-trimethyl-6-(trifluoromethoxy)-2, 3,4,4a, 9,9a- hexahydro-1 /-/-fluoren-9-ol (Compound 221);

(±)-(4aR,9aR)-6-Fluoro-1 ,1-dimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)- one (Compound 222);

(2-Deutero-6,6-dimethylcyclohex-1-enyl)(4-fluorophenyl)metha none

(Compound 223);

(±)-(4aS,9aR)-4a-Deutero-6-fluoro-1 ,1-dimethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one (Compound 224);

(2-Deutero-6,6-dimethylcyclohex-1-enyl)(3,4-difluorophenyl)m ethanone (Compound 225);

(±)-(4aS,9aS)-6-Chloro-4,4-dimethyl-2,3,4,4a-tetrahydro-1 /-/-fluoren-9(9aH)- one (Compound 226);

(±)-(4aS,9aR)-4a-Deutero-6,7-difluoro-1 , 1 -dimethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one (Compound 227);

(3,4-Difluorophenyl)(6,6-dimethylcyclohex-1-enyl)methanol (Compound 228); (±)-(4aS,9S,9aS)-6-Chloro-4,4-dimethyl-2,3,4,4a,9,9a-hexahy dro-1 H-fluoren- 9-ol (Compound 229);

(6,6-Dimethylcyclohex-1-enyl)(4-methoxyphenyl)methanol (Compound 230); (±)-(4aS,9aS)-6-Methoxy-4,4-dimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9a/-/)- one (Compound 233);

(±)-(4aS,9aS)-4,4,8-Trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aH)-one (Compound 236);

(±)-(4aS,9aS)-4,4-Dimethyl-9-oxo-2, 3,4,4a, 9,9a-hexahydro-1H-fluorene-6- carbonitrile (Compound 237);

(±)-(4aS,9aS)-6,7-Difluoro-4,4-dimethyl-2,3,4,4a-tetrahydro -1 H-fluoren- 9(9a/-/)-one (Compound 238); (±)-(4aS,9S,9aS)-67-Difluoro-4,4-dimethyl-2,3,4,4a,9,9a-hex ahydro-1H- fluoren-9-ol (Compound 239) including all pharmaceutically acceptable salts, hydrates or solvates thereof. All compound names were derived using ChemBioDraw Ultra 11.0.1.

The present invention does not include the following compounds (as represented by their indicated CAS registry numbers) as novel compositions of matter (but are claimed for use in the methods of the invention) and are referred to herein as the Excluded Compound Group: 1205544-36-0, 1205544-35-9, 205544-34-8, 1205544-33-7, 1205544-32-6, 1205544-30-4, 1205544-28-0, 1205544-26-8, 1205544-24-6, 1205544-22-4, 1205544-20-2, 1205544-18-8, 1205544-16-6, 1205544-14-4, 1205544-10-0, 1205544-09-7, 1205544-07-5, 1205544-05-3, 170307-10-4, 170307-09-1 , 1 70307-08-0, 1170307-07-9, 1148136-47-3, 1148136-43-9, 1148136-39-3, 1131696-63-3, 1131696-61-1 , 131696-42-8, 1021879-70-8, 1021879-69-5, 1021879-68-4, 1021879-67-3, 1021879-66-2, 1021879-65-1 , 1018443-44-1 , 915939-88-7, 915939-84-3, 911850-42-5, 911850-39-0, 911850-32-3, 911850-31-2, 911850-30-1 , 911850-29-8, 911850-28-7, 901132-21-6, 901132-04-5, 885676-94-8, 885676-93-7, 885676-89-1 , 885482-39-3, 885482-38-2, 885482-37-1 , 885482-35-9, 885482-34-8, 885482-33-7, 885482-32-6,

885482-31-5, 885482-30-4, 885482-29-1 , 885482-27-9, 885482-26-8, 885482-24-6, 885482-23-5, 885482-22-4, 885482-21-3, 885482-10-0, 883745-90-2, 883745-83-3, 866892-00-4, 866821-39-8, 866821-29-6, 858133-95-6, 696646-79-4, 696646-73-8, 622342-26-1 , 620167-65-9,

620167-64-8 620167-63-7, 620167-62-6, 620167-61-5, 620167-60-4, 620167-59-1 620167-58-0, 620167-57-9, 620167-56-8, 620167-55-7,

250367-96-5, 250367-95-4, 250367-94-3, 250367-93-2, 221462-91-5,

221462-90-4, 221462-88-0, 221462-87-9, 180253-17-2, 170384-72-2, 168920-76-1 , 168920-75-0,168920-74-9, 683800-20-6,172969-99-2,114201- 90-0 1170307-04-6, 1170307-03-5, 1148136-45-1 , 1 48136-41-7, 1148136- 37-1 , 1018443-41-8, 911850-41-4, 911850-40-3, 9 850-38-9, 91 850-37-8, 911850-25-4, 911850-24-3, 883745-87-7, 206862-16-0, 156416-84-1 , 131265-53-7, 122131-12-8, 60463-84-5, 53210-26-7, 151194-64-8, 114201- 45-5, 93024-59-0, 64884-70-4, 107547-21-7, 1233407-19-6, 1148136-48-4, 1148136-45-1 , 148136-41-7, 1148136-37-1 , 1148136-35-9, 1148136-27-9, 911850-47-0, 911850-44-7, 911850-41-4, 911850-40-3, 911850-38-9, 911850-37-8 , 911850-25-4, 911850-24-3, 257881-53-1 , 131265-53-7, 122131-12-8, 77838-00-7, 60463-84-5, 53210-26-7, 1243263-28-6, 1243263-

27- 5, 1240509-28-7, 1240509-27-6, 1240509-26-5, 1205544-36-0, 1205544- 35-9, 1205544-34-8, 1205544-33-7, 1205544-32-6, 1205544-30-4, 1205544-

28- 0, 1205544-26-8, 1205544-24-6, 1205544-22-4, 901132-31-8, 901132-19- 2, 620167-64-8, 408318-78-5 and 108842-22-4.

The Excluded Compound Group contains any and all of the compounds in the preceding list as identified by their indicated CAS (Chemical Abstracts Service) numbers.

Thus, the novel compounds and/or compositions of matter of the invention are compounds of Formula I and Formula II (including their indicated substitutent identities) but do not include compounds of the Excluded Compound Group.

However, the methods of the present invention employ any compounds of Formula I and Formula II (including their indicated substitutent identities) but do not exclude use of compounds of the Excluded Compound Group. Especially preferred examples of the compounds of the invention, and methods using said compounds, include compounds selected from one or more of the group consisting of compounds 3, 3a, 3b, 5, 7, 11 , 12, 13, 17, 18, 28, 29, 36, 37, 40 41 , 43 ,47, 48, 53, 54, 56, 57, 58, 61 , 62, 63, 64a, 64b, 67, 68, 69, 70, 71 , 73, 75, 79, 80, 84, 87, 88, 90, 93, 101 , 105, 109, 110, 112a, 112b, 113, 115, 116a, 116b, 123, 125, 126, 129, 131 , 132, 133, 137, 139, 142, 147, 149, 155, 166, 168, 169, 171 , 182, 187, 189, 190, 191 , 202, 205, 207, 213, 214, 217, 218 and 220 including all pharmaceutically acceptable salts, solvates and hydrates thereof. Another embodiment of the invention provides the opsin binding ligand metabolites of the opsin binding compounds. These metabolites, include but are not limited to, degradation products, hydrolysis products, gluconoride adducts and the like, of the opsin binding compounds and pharmaceutically acceptable salts thereof, of the opsin compounds.

Another embodiment of the invention provides processes for making the novel compounds of the invention and to the intermediates useful in such processes. The reactions are performed in solvents appropriate to the reagents and materials used are suitable for the transformations being effected. It is understood by one skilled in the art of organic synthesis that the functionality present in the molecule must be consistent with the chemical transformation proposed. This will, on occasion, necessitate judgment by the routineer as to the order of synthetic steps, protecting groups required, and deprotection conditions. Substituents on the starting materials may be incompatible with some of the reaction conditions required in some of the methods described, but alternative methods and substituents compatible with the reaction conditions will be readily apparent to one skilled in the art. The use of sulfur, nitrogen and oxygen protecting groups is well known for protecting thiol, amino and alcohol groups against undesirable reactions during a synthetic procedure and many such protecting groups are known and described by, for example, Greene and Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, New York (1999). Compounds of the invention that have one or more asymmetric carbon atoms may exist as the optically pure enantiomers, pure diastereomers, mixtures of enantiomers, mixtures of diastereomers, racemic mixtures of enantiomers, diastereomeric racemates or mixtures of diastereomeric racemates. It is to be understood that the invention anticipates and includes within its scope all such isomers and mixtures thereof.

The chemical reactions described herein are generally disclosed in terms of their broadest application to the preparation of the compounds of this invention. Occasionally, the reactions may not be applicable as described to each compound included within the disclosed scope. The compounds for which this occurs will be readily recognized by one skilled in the art. In all such cases, either the reactions can be successfully performed by conventional modifications known to one skilled in the art, e.g., by appropriate protection of interfering groups, by changing to alternative conventional reagents, by routine modification of reaction conditions, or other reactions disclosed herein or otherwise conventional, will be applicable to the preparation of the corresponding compounds of this invention. In all preparative methods, all starting materials are known or readily prepared from known starting materials.

Methods of the invention The present invention provides a method of using compounds of the

Formula I for reducing the formation of toxic visual cycle products, comprising contacting an opsin protein with small molecule ligands that reversibly bind to said opsin protein to inhibit 11-cis-retinal binding in said binding pocket, thereby reducing formation of toxic visual cycle products associated with wet or dry ARMD. and reducing photocell apoptosis associatiated with excessive rhodopsin activation as a result of bright light stimulation.

The present invention also provides a method of use of compounds of the Formula I for treating, preventing or reducing the risk of light toxicity in a mammal, comprising administering to a mammal, at risk of developing an ophthalmic condition that is related to the formation or accumulation of a visual cycle product or apoptotic photocell death.

The present invention also provides a method of use of compounds of the Formula I for treating, preventing or reducing the risk of light toxicity in a mammal, comprising administering to a mammal, at risk of developing an ophthalmic condition that is related to the formation or accumulation of a visual cycle product or apoptotic photocell death, an effective amount of a that small molecule ligand that reversibly binds (for example, at or near the retinal binding pocket) to an opsin protein present in the eye of said mammal, for example, to inhibit -cis-retinal binding in said binding pocket, thereby reducing light toxicity and photocell apoptosis.

The present invention also provides a method of use of compounds of the Formula I for treating, preventing or reducing the risk of RP in a mammal, comprising administering to a mammal, at risk of RP related to the improper folding and trafficking of mutant opsins, an effective amount of a that small molecule ligand that reversibly binds (for example, at or near the retinal binding pocket) to an opsin protein present in the eye of said mammal, for example, to inhibit 11-cis-retinal binding in said binding pocket, thereby reducing the vision loss caused by RP. In specific examples of such methods, the small molecule ligand is selective for binding to opsin and/or the small molecule ligand binds to said opsin in the retinal binding pocket of said opsin protein and/or the small molecule ligand binds to said opsin protein so as to inhibit covalent binding of 1 -cis-retinal to said opsin protein when said 11-cis-retinal is contacted with said opsin protein when said small molecule ligand is present and/or the mammal is a human being.

In one embodiment, light toxicity is related to an ophthalmic procedure, for example, ophthalmic surgery. Said agent may be administered prior to, during or after said surgery (or at any one or more of those times).

In specific embodiments of the methods of the invention, the native opsin protein is present in a cell, such as a rod cell, preferably, a mammalian and more preferably a human cell. In specific embodiments, the small molecule ligands of the invention inhibit binding of 11-cis-retinal in the binding pocket of opsin and slow the visual cycle thereby reducing the formation of all- trans-retinal, or a toxic visual cycle product formed from it, such as lipofuscin or N-retinylidene-N-retinylethanolamine (A2E). Alternatively, photocell apoptosis as a result of excessive rhodopsin activation is reduced or prevented by inhibition of rhodopsin formation. Additionally, improper folding and trafficking of mutant opsin proteins associated with RP is reduced. In methods of the invention, administering is preferably by topical administration (such as with an eye wash) or by systemic administration (including oral, intraocular injection or periocular injection). By way of preferred example, the ophthalmic condition to be treated is light toxicity, such as that resulting from ocular surgery, for example, retinal or cataract surgery.

Also encompassed is an ophthalmologic composition comprising an effective amount of compounds of the Formula I in a pharmaceutically acceptable carrier, wherein said agent reversibly binds non-covalently (for example, at or near the retinal binding pocket) to said opsin protein to inhibit 11-cis-retinal binding in said pocket, preferably where the small molecule ligand is selective for opsin protein.

The present invention further provides a screening method for identifying a small molecule ligand that reduces light toxicity in a mammalian eye, comprising:

(a) contacting a native opsin-protein with a test compound in the presence of 1 -cis-retinal and under conditions that promote the binding of the test compound and the 11-cis-retinal to the native opsin protein; and

(b) determining a reversible reduction in rate of formation of rhodopsin relative to the rate when said test compound is not present,

thereby identifying said test compound as a small molecule ligand that reduces light toxicity in a mammalian eye. In a preferred embodiment, said test compound is structurally related to a compound disclosed herein. In a typical competition assay of the invention, a compound is sought that will tie up the retinal binding pocket of the opsin protein. Thus, the assay seeks to identify a small molecule opsin binding compound (one that will not be tightly regulated by the retina as to amount entering rod cells) that competes with or prevents 11-cis-retinal or 9-cis-retinal from forming rhodopsin or isorhodopsin. Over time, this will slow the rate of formation of rhodopsin relative to the rate when 11-cis-retinal alone is present. In one embodiment, the assay is conducted in the presence of 11-cis-retinal, and the rate of formation of rhodopsin is measured as a way of determining competition for the retinal binding pocket, for example, by determining the rate of increase in the 500 nm peak characteristic for rhodopsin. No antibodies for rhodopsin are required for this assay. A useful compound will exhibit a rate of rhodopsin formation that is at least about 2 to 5 fold lower than that observed in the presence of 11-cis-retinal when said test compound is not present.

The compounds of the Formula I may be administered along with other agents, including a mineral supplement, an anti-inflammatory agent, such as a steroid, for example, a corticosteroid, and/or an anti-oxidant. Among the corticosteroids useful for such administration are those selected from the group consisting of cortisone, hydrocortisone, prednisone, prednisolone, methylprednisolone, triamcinolone, betamethasone, beclamethasone and dexamethasone. Useful anti-oxidants include vitamin A, vitamin C and vitamin E.

The methods of the invention also contemplate reducing light toxicity by using at least one additional agent (in addition to the compounds of the Formula I selected from the group consisting of a proteasomal inhibitor, an autophagy inhibitor, a lysosomal inhibitor, an inhibitor of protein transport from the ER to the Golgi, an Hsp90 chaperone inhibitor, a heat shock response activator, a glycosidase inhibitor, and a histone deacetylase inhibitor, wherein the small molecule opsin binding and the additional compound are administered simultaneously or within fourteen days of each other in amounts sufficient to treat the subject.

In a particular example of the methods of the invention, the compounds of the Formula I and/or Formula II the additional compound are administered within ten days of each other, within five days of each other, within twenty-four hours of each other and preferably are administered simultaneously. In one example, the small molecule opsin binding and the additional compound are administered directly to the eye. Such administration may be intraocular or intravitrial. In other examples, the small molecule opsin binding and the additional compound are each incorporated into a composition that provides for their long-term release, such as where the composition is part of a microsphere, nanosphere, nano emulsion or implant.

As described herein, the compounds of the Formula I and/or II useful in the methods of the invention are available for use alone or in combination with one or more additional compounds to treat or prevent conditions associated with excessive rhodopsin activation, such as light toxicity, for example, resulting from ocular surgical procedures. In one embodiment, compounds of the Formula I and/or II of the invention is administered without an additional active compound. In another embodiment, compounds of Formula I and/or II of the invention is used in combination and with another active compound (e.g., as discussed herein). In still another exemplary embodiment, compounds of the Formula I and/or II are administered in combination with the proteasomal inhibitor MG132, the autophagy inhibitor 3-methyladenine, a lysosomal inhibitor ammonium chloride, the ER-Golgi transport inhibitor brefeldin A, the Hsp90 chaperone inhibitor Geldamycin, the heat shock response activator Celastrol, the glycosidase inhibitor, and the histone deacetylase inhibitor Scriptaid, can be used to reduce formation of visual cycle products and cell apoptosis as a result of excessive rhodopsin activation.

As described herein, the compounds of the Formula I and/or II useful in the methods of the invention are available for use alone or in combination with one or more additional compounds to treat or prevent the aberrant processing and trafficking of mutant opsin proteins associated with rod cell death as a result of RP. In one embodiment, compounds of the Formula I and/or II of the invention is administered without an additional active compound. In another embodiment, compounds of the Formula I and/or II of the invention are used in combination and with another active compound (e.g., as discussed herein). In still another exemplary embodiment, compounds of the Formula I and/or II are administered in combination with the proteasomal inhibitor MG132, the autophagy inhibitor 3-methyladenine, a lysosomal inhibitor ammonium chloride, the ER-Golgi transport inhibitor brefeldin A, the Hsp90 chaperone inhibitor Geldamycin, the heat shock response activator Celastrol, the glycosidase inhibitor, and the histone deacetylase inhibitor Scriptaid, can be used to reduce or prevent the rod cell death and resulting blindness associated with RP.

As described herein, the compounds of the Formula I and/or II useful in the methods of the invention are available for use alone or in combination with one or more additional compounds to treat or prevent conditions associated with production and accumulation of toxic visual cycle products derived from all-trans-retinal, such as lipofucin and A2E, for example, the blindness associated with wet or dry ARMD. In one embodiment, compounds of the Formula I and/or II of the invention is administered without an additional active compound. In another embodiment, compounds of the Formula I and/or II of the invention is used in combination and with another active compound (e.g., as discussed herein). In still another exemplary embodiment, compounds of the Formula I and/or II are administered in combination with the proteasomal inhibitor MG132, the autophagy inhibitor 3-methyladenine, a lysosomal inhibitor ammonium chloride, the ER-Golgi transport inhibitor brefeldin A, the Hsp90 chaperone inhibitor Geldamycin, the heat shock response activator Celastrol, the glycosidase inhibitor, and the histone deacetylase inhibitor Scriptaid, can be used to reduce formation of toxic visual cycle product metabolites and photo cell death as a result of dry ARMD.

In specific embodiments of the methods of the invention, the mis-folded opsin protein comprises a mutation in its amino acid sequence, for example, one of the mutations T17M, P347S or P23H, preferably P23H.

Preferably, in any of the methods of the invention, the opsin-binding agent binds to opsin in its retinal binding pocket.

In one aspect, the present invention provides a method of inhibiting the formation or accumulation of a visual cycle product, comprising contacting an opsin protein with a compound that reduces hydration of said opsin protein, preferably wherein said compound competes with one or more water molecules for binding to opsin. In specific embodiments of such methods, the compound binds chemically to the opsin protein, for example, through hydrogen bonding.

In specific examples of the methods of the invention, a compound useful therein may bind to opsin at any hydration site found within the retinal binding pocket of the opsin molecule so long as said binding excludes wholly, or in part, the binding of one or more water molecules in said binding pocket. Preferably the compound used in such method binds so as to occupy the left side of the binding pocket as shown in Figure 1 and displace waters in hydration sites 5-20 (D labeled circles on left side of Figure 1), more preferably binds so that waters in hydration sites 5-20 are displaced (circles with D in Figure 1), and waters at hydration sites 3 or 4 (R labeled circles within 1 -cis retinal molecular surface as shown in Figure 1 are displaced and replaced with functionality on the ligand that mimics the hydrogen bonding interactions that these waters are predicted to have with residues on the protein. A specific example of these methods contemplates binding of a compound by chemical interaction with Cys 87 or Glu1 3 of the opsin protein. In separate embodiments thereof, said interaction is with Cys187 or said interaction is with Glu1 3 or is with both sites. A preferred mode of said interaction is hydrogen bonding.

In other specific examples, said interaction is with a carbonyl group on the opsin protein. In specific embodiments thereof, said carbonyl is on Cys187 or Glu 13 of said opsin protein. Separate embodiments include where the carbonyl is on Cys187 of the opsin protein or where the carbonyl is on Glu113 of the opsin protein. In one embodiment, the carbonyl is in the gamma- carboxyl group of Glu113 of the opsin protein. A preferred embodiment is where the interaction is through an amine, carboxamido or urea group on the compound.

While use of any of the compounds disclosed herein as a means of reducing hydration in the opsin binding pocket should be considered a preferred embodiment of such method, the reduction of formation of a visual cycle product by reducing the formation of rhodopsin is a general method of the invention for reducing such visual cycle product formation, especially production of lipofuscin and/or A2E, and for treating an ophthalmic disease by reducing said hydration is a general aim of the invention and is not necessarily limited in scope only to the use of chemicals disclosed herein but may include use of other known or yet to be known chemical compounds so long as they function in the methods of the invention and reduce hydration (i.e., binding of water) in the retinal binding pocket of opsin.

It should be noted that the compounds disclosed herein for use in the methods of the invention may not function to reduce hydration in the retinal binding pocket of opsin but may still function in one or more of the methods of the invention. For example, a compound of Formula I or II may bind to an allosteric site on the protein thereby excluding retinal from the retinal binding site without necessarily decreasing hydration yet still reduce formation of a visual cycle product, such as lipofuscin and/or A2E, by virtue of its excluding retinal from the binding pocket, thus non-covalently reducing the activity of the visual cycle.

In embodiments of any of the compositions and methods of the invention, the opsin-binding agent (e.g., a non-retinoid binding agent) is selective for binding to opsin. Such selectivity is not to be taken as requiring exclusivity that said agent may bind to other proteins as well as to opsin but its binding to opsin will be at least selective, whereby the binding constant (or dissociation constant) for binding to opsin will be lower than the average value for binding to other proteins that also bind retinoids, such as retinal analogs. Preferably, opsin binding agents are non-retinoid opsin-binding agents that bind non-covalently to opsin. Preferably, the opsin binding agent binds at or near the opsin retinal binding pocket, where the native ligahd, 1-cis-retinal, normally binds. Without wishing to be bound by theory, in one embodiment the binding pocket accommodates retinal or an agent of the invention, but not both. Accordingly, when an agent of the invention is bound at or near the retinal binding pocket, other retinoids, such as 11-cis-retinal, are unable to bind to opsin. Binding of an agent of the invention inside the retinal binding pocket of a mis-folded opsin molecule serves to direct formation of the native or wild-type conformation of the opsin molecule or to stabilize a correctly folded opsin protein, thereby facilitating insertion of the now correctly-folded opsin into the membrane of a rod cell. Again, without wishing to be bound by theory, said insertion may help to maintain the wild-type conformation of opsin and the opsin-binding agent is free to diffuse out of the binding pocket, whereupon the pocket is available for binding to retinal to form light-sensitive rhodopsin. Other methods of the invention provide a means to restore photoreceptor function in a mammalian eye containing a mis-folded opsin protein that causes reduced photoreceptor function, comprising contacting said mis-folded opsin protein with an opsin-binding agent (e.g., a non-retinoid) that reversibly binds (e.g., that binds non-covalently) at or near the retinal binding pocket. In other embodiments, binding of the opsin-binding agent to the mis-folded opsin protein competes with 11-cis-retinal for binding in said binding pocket. Desirably, binding of the opsin-binding agent restores the native conformation of said mis-folded opsin protein. In preferred embodiments, the mammalian eye is a human eye. In additional embodiments, said contacting occurs by administering said opsin- binding agent (e.g., non-retinoid) to a mammal afflicted with an ophthalmic condition, such as a condition characterized by reduced photoreceptor function. In various embodiments, the condition is the wet or dry form of macular degeneration, diabetic RP, a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity (e.g., due to retinal surgery), or retinitis pigmentosa. The administration may be topical administration or by systemic administration, the latter including oral administration, intraocular injection or periocular injection. Topical administration can include, for example, eye drops containing an effective amount of an agent of the invention in a suitable pharmaceutical carrier.

In another embodiment, the present invention also provides a method of stabilizing a mutant opsin protein, comprising contacting said mutant opsin protein with a non-retinoid opsin-binding agent that reversibly binds non- covalently (for example, at or in the retinal binding pocket) to said mutant opsin protein to prevent retinoid binding in said binding pocket, thereby stabilizing said mutant opsin protein.

The present invention also provides a method of ameliorating loss of photoreceptor function in a mammalian eye, comprising administering an effective amount of an opsin-binding agent, such as a non-retinoid, to a mammal afflicted with a mutant opsin protein that has reduced affinity for 11- cis-retinal, whereby the opsin binding agent reversibly binds (e.g., non- covalently) to the retinal binding pocket of said mutant opsin, thereby ameliorating loss of photoreceptor function in said mammalian eye. In one embodiment, the contacting occurs by administering said opsin-binding agent to a mammal afflicted with said reduced photoreceptor function, wherein said administering may be by topical administration or by systemic administration, the latter including oral, intraocular injection or periocular injection, and the former including the use of eye drops containing an agent of the invention. Such loss of photoreceptor function may be a partial loss or a complete loss, and where a partial loss it may be to any degree between 1% loss and 99% loss. In addition, such loss may be due to the presence of a mutation that causes mis-folding of the opsin, such as where the mutation is the P23H mutation. In another embodiment, the opsin binding agent is administered to ameliorate an ophthalmic condition related to the mislocalization of an opsin protein. In one embodiment, the invention provides for the treatment of a subject having the dry form of age-related macular degeneration, where at least a portion of the opsin present in an ocular photoreceptor cell (e.g., a rod or cone cell) is mislocalized. The mislocalized protein fails to be inserted into the membrane of a photoreceptor cell, where its function is required for vision. Administration of the opsin binding agent to a subject having a mislocalized opsin protein rescues, at least in part, opsin localization. Accordingly, the invention is useful to prevent or treat an ophthalmic condition related to opsin mislocalization or to ameliorate a symptom thereof.

The present invention provides a method for treating and/or preventing an ophthalmic condition or a symptom thereof, including but not limited to, wet or dry form of macular degeneration, retinitis pigmentosa, a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity (e.g., due to retinal surgery), or retinitis pigmentosa in a subject, such as a human patient, comprising administering to a subject afflicted with, or at risk of developing, one of the aforementioned conditions or another ophthalmic condition related to the expression of a misfolded or mislocalized opsin protein using a therapeutically effective amount of an opsin-binding agent, e.g., an agent that shows positive activity when tested in any one or more of the screening assays of the invention.

Such a method may also comprise administering to said subject at least one additional agent selected from the group consisting of a proteasomal inhibitor, an autophagy inhibitor, a lysosomal inhibitor, an inhibitor of protein transport from the ER to the Golgi, an Hsp90 chaperone inhibitor, a heat shock response activator, a glycosidase inhibitor, and a histone deacetylase inhibitor, wherein the opsin-binding compound and the additional compound are administered simultaneously or within fourteen days of each other in amounts sufficient to treat the subject.

Here again the patient may comprise a mutation that affects protein folding where said mutation(s) causes mis-folding, e.g., in an opsin protein, and may be any of the mutations recited elsewhere herein, such as a P23H mutation. In other embodiments, the patient has an ophthalmic condition that is related to the mislocalization of an opsin protein. The mislocalized opsin fails to insert into the membrane of a photoreceptor cell (e.g., a rod or cone cell). In general, this failure in localization would effect only a portion of the opsin present in an ocular cell of a patient.

In particular examples of the methods of the invention, the opsin- binding compound and the additional compound are administered within ten days of each other, more preferably within five days of each other, even more preferably within twenty-four hours of each other and most preferably are administered simultaneously. In one example, the opsin-binding compound and the additional compound are administered directly to the eye. Such administration may be intra-ocular. In other examples, the opsin-binding compound and the additional compound are each incorporated into a composition that provides for their long-term release, such as where the composition is part of a microsphere, nanosphere, or nano emulsion. In one example, the composition is administered via a drug-delivery device that effects long-term release. Such methods also contemplate administering a vitamin A supplement along with an agent of the invention.

As described herein, the opsin-binding agents useful in the methods of the invention are available for use alone or in combination with one or more additional compounds to treat or prevent conditions associated with the wet or dry form of macular degeneration, retinitis pigmentosa, a retinal or macular dystrophy, Stargardt's disease, Sorsby's dystrophy, autosomal dominant drusen, Best's dystrophy, peripherin mutation associate with macular dystrophy, dominant form of Stargart's disease, North Carolina macular dystrophy, light toxicity (e.g., due to retinal surgery), retinitis pigmentosa or another ophthalmic condition related to the expression of a misfolded or mislocalized opsin protein. In one embodiment, an opsin-binding compound of the invention (e.g., a non-retinoid or a retinoid that fails to covalently bind to opsin) is administered to a subject identified as having or at risk of developing such a condition. Optionally, the opsin binding agent is administered together with another therapeutic agent. In another embodiment, a non-retinoid opsin- binding compound of the invention is used in combination with a synthetic retinoid (e.g., as disclosed in U.S. Patent Publication No. 2004-0242704), and optionally with another active compound (e.g., as discussed herein). In still another exemplary embodiment, an opsin-binding compound is administered in combination with the proteasomal inhibitor MG132, the autophagy inhibitor 3-methyladenine, a lysosomal inhibitor, such as ammonium chloride, the ER- Golgi transport inhibitor brefeldin A, the Hsp90 chaperone inhibitor Geldamycin, the heat shock response activator Celastrol, the glycosidase inhibitor, and/or the histone deacetylase inhibitor Scriptaid, or any other agent that can stabilize a mutant P23H opsin protein in a biochemically functional conformation that allows it to associate with 1 1-cis-retinal to form rhodopsin. In specific embodiments, an opsin-binding compound is a non- polymeric (e.g., a small molecule, such as those disclosed herein for use in the methods of the invention) compound having a molecular weight less than about 1000 daltons, less than 800, less than 600, less than 500, less than 400, or less than about 300 daltons. In certain embodiments, a compound of the invention increases the amount (e.g., from or in a cell) of a stably-folded and/or complexed mutant protein by at least 10%, 15%, 20%, 25%, 50%, 75%, or 00% compared to an untreated control cell or protein.

Proteasomal inhibitors

The 26S proteasome is a multicatalytic protease that cleaves ubiquinated proteins into short peptides. MG-132 is one proteasomal inhibitor that may be used. MG- 132 is particularly useful for the treatment of light toxicity and other ocular diseases related to the accumulation of visual cycle products (e.g., all-trans-retinal, A2E, lipofuscin), protein aggregation or protein misfolding. Other proteasomal inhibitors useful in combination with of the invention in the methods of the invention include lactocystin (LC), clasto- lactocystin-beta-lactone, PSI (N-carbobenzoyl-lle-Glu-(OtBu)-Ala-Leu-CHO), MG-132 (N-carbobenzoyl-Leu-Leu-Leu-CHO), MG-115 (N-carbobenzoyl-Leu- Leu-Nva-CHO), MG-101 (N-Acetyl-Leu-Leu-norLeu-CHO), ALLM (N-Acetyl- Leu-Leu-Met-CHO), N-carbobenzoyl-Gly-Pro-Phe-leu-CHO, N-carbobenzoyl- Gly-Pro-Ala-Phe-CHO, N-carbobenzoyl-Leu-Leu-Phe-CHO, and salts or analogs thereof. Other proteasomal inhibitors and their uses are described in U.S. Patent No. 6,492,333.

Autophagy inhibitors

Autophagy is an evolutionary conserved mechanism for the degradation of cellular components in the cytoplasm, and serves as a cell survival mechanism in starving cells. During autophagy pieces of cytoplasm become encapsulated by cellular membranes, forming autophagic vacuoles that eventually fuse with lysosomes to have their contents degraded. Autophagy inhibitors may be used in combination with an opsin-binding or opsin-stabilizing compound of the invention. Autophagy inhibitors useful in combination with a of the invention in the methods of the invention include, but are not limited to, 3-methyladenine, 3-methyl adenosine, adenosine, okadaic acid, N 6 -mercaptopurine riboside (N 6 -MPR), an aminothiolated adenosine analog, 5-amino-4-imidazole carboxamide riboside (AICAR), bafilomycin A1 , and salts or analogs thereof.

Lysosomal inhibitors

The lysosome is a major site of cellular protein degradation. Degradation of proteins entering the cell by receptor-mediated endocytosis or by pinocytosis, and of plasma membrane proteins takes place in lysosomes. Lysosomal inhibitors, such as ammonium chloride, leupeptin, trans- epoxysaccinyl-L-leucylamide-(4-guanidino) butane, L-methionine methyl ester, ammonium chloride, methylam/ne, chloroquine, and salts or analogs thereof, are useful in combination with an opsin-binding or opsin-stabilizing compound of the invention.

HSP90 chaperone inhibitors

Heat shock protein 90 (Hsp90) is responsible for chaperoning proteins involved in cell signaling, proliferation and survival, and is essential for the conformational stability and function of a number of proteins. HSP-90 inhibitors are useful in combination with an opsin-binding or opsin-stabilizing compound in the methods of the invention. HSP-90 inhibitors include benzoquinone ansamycin antibiotics, such as geldanamycin and 17- allylamino-17-demethoxygeldanamycin (I7-AAG), which specifically bind to Hsp90, alter its function, and promote the proteolytic degradation of substrate proteins. Other HSP-90 inhibitors include, but are not limited to, radicicol, novobiocin, and any Hsp9O inhibitor that binds to the Hsp90 ATP/ADP pocket.

Heat shock response activators

Celastrol, a quinone methide triterpene, activates the human heat shock response. In combination with an opsin-binding or opsin-stabilizing compound in methods of the invention, celastrol and other heat shock response activators are useful for the treatment of PCD. Heat shock response activators include, but are not limited to, celastrol, celastrol methyl ester, dihydrocelastrol diacetate, celastrol butyl ester, dihydrocelastrol, and salts or analogs thereof. Histone deacetylase inhibitors

Regulation of gene expression is mediated by several mechanisms, including the post-translational modifications of histones by dynamic acetylation and deacetylation. The enzymes responsible for reversible acetylationl/deacetylation processes are histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. Histone deacetylase inhibitors include Scriptaid, APHA Compound 8, Apicidin, sodium butyrate, (-)- Depudecin, Sirtinol, trichostatin A, and salts or analogs thereof. Such inhibitors may be used in combination with compounds of the invention in the methods disclosed herein.

Glycosidase inhibitors Glycosidase inhibitors are one class of compounds that are useful in the methods of the invention, when administered in combination with an opsin-binding or opsin-stabilizing compound of the invention. Castanospermine, a polyhydroxy alkaloid isolated from plant sources, inhibits enzymatic glycoside hydrolysis. Castanospermine and its derivatives are particularly useful for the treatment of light toxicity or of an ocular Protein Conformation Disorder, such as RP. Also useful in the methods of the invention are other glycosidase inhibitors, including australine hydrochloride, 6-Acetamido-6-deoxy-castanosperrnine, which is a powerful inhibitor of hexosaminidases, Deoxyfuconojirimycin hydrochloride (DFJ7), Deoxynojirimycin (DNJ), which inhibits glucosidase I and II, Deoxygalactonojirimycin hydrochloride (DGJ), winch inhibits a-D- galactosidase, Deoxymannojirimycin hydrochloride (DM1 ), 2R.5R- Bis(hydroxymethyl)-3R,4R-dihydroxypyrrolidine (DMDP), also known as 2,5- dideoxy-2,5-imino-D-mannitol, 1 ,4-Dideoxy-1 ,4-imino-D-mannitol hydrochloride, (3R,4R,5R,6R)-3,4,5,6-Tetrahydroxyazepane Hydrochloride, which inhibits β-Ν-acetylglucosaminidase, 1 ,5-Dideoxy-1 ,5-imino-xylitol, which inhibits β-glucosidase, and Kifunensine, an inhibitor of mannosidase 1. Also useful in combination with an opsin-binding or opsin-stabilizing compound are N-butyldeoxynojirimycin (EDNJ), N-nonyl DNJ (NDND, N-hexyl DNJ (I5TDNJ), N-methyldeoxynojirimycin (MDNJ), and other glycosidase inhibitors known in the art. Glycosidase inhibitors are available commercially, for example, from Industrial Research Limited (Wellington, New Zealand) and methods of using them are described, for example, in U.S. Patent Nos. 4,894,388, 5,043,273, 5,103,008, 5,844,102, and 6,831 ,176; and in U.S. Patent Publication Nos. 20020006909.

Pharmaceutical Compositions

The present invention features pharmaceutical preparations comprising compounds together with pharmaceutically acceptable carriers, where the compounds provide for the inhibition of visual cycle products, such as all- trans-retinal or other products formed from 1 1-cis-retinal. Such preparations have both therapeutic and prophylactic applications. In one embodiment, a pharmaceutical composition includes an opsin-binding or stabilizing compound (e.g., a compound identified using the methods of Example 1 ) or a pharmaceutically acceptable salt thereof; optionally in combination with at least one additional compound that is a proteasomal inhibitor, an autophagy inhibitor, a lysosomal inhibitor, an inhibitor of protein transport from the ER to the Golgi, an Hsp90 chaperone inhibitor, a heat shock response activator, a glycosidase inhibitor, or a histone deacetylase inhibitor. The opsin-binding or opsin-stabilizing compound is preferably not a natural or synthetic retinoid. The opsin-binding or opsin-stabilizing compound and the additional compound are formulated together or separately. Compounds of the invention may be administered as part of a pharmaceutical composition. The non-oral compositions should be sterile and contain a therapeutically effective amount of the opsin-binding or opsin-stabilizing compound in a unit of weight or volume suitable for administration to a subject. The compositions and combinations of the invention can be part of a pharmaceutical pack, where each of the compounds is present in individual dosage amounts. The phrase "pharmaceutically acceptable" refers to those compounds of the present invention, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

Non-oral pharmaceutical compositions of the invention to be used for prophylactic or therapeutic administration should be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 μητι membranes), by gamma irradiation, or any other suitable means known to those skilled in the art. Therapeutic opsin-binding or opsin-stabilizing compound compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. These compositions ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. The compounds may be combined, optionally, with a pharmaceutically acceptable excipient.

The components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction that would substantially impair the desired pharmaceutical efficacy.

Compounds of the present invention can be contained in a pharmaceutically acceptable excipient. The excipient preferably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetate, lactate, tartrate, and other organic acids or their salts; tris- hydroxymethylaminomethane (TRIS), bicarbonate, carbonate, and other organic bases and their salts; antioxidants, such as ascorbic acid; low molecular weight (for example, less than about ten residues) polypeptides, e.g., polyarginine, polylysine, polyglutamate and polyaspartate; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers, such as polyvinylpyrrolidone (PVP), polypropylene glycols (PPGs), and polyethylene glycols (PEGs); amino acids, such as glycine, glutamic acid, aspartic acid, histidine, lysine, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, sucrose, dextrins or sulfated carbohydrate derivatives, such as heparin, chondroitin sulfate or dextran sulfate; polyvalent metal ions, such as divalent metal ions including calcium ions, magnesium ions and manganese ions; chelating agents, such as ethylenediamine tetraacetic acid (EDTA); sugar alcohols, such as mannitol or sorbitol; counterions, such as sodium or ammonium; and/or nonionic surfactants, such as polysorbates or poloxamers. Other additives may be included, such as stabilizers, anti-microbials, inert gases, fluid and nutrient replenishers (i.e., Ringer's dextrose), electrolyte replenishes, which can be present in conventional amounts.

The compositions, as described above, can be administered in effective amounts. The effective amount will depend upon the mode or administration, the particular condition being treated and the desired outcome. It may also depend upon the stage of the condition, the age and physical condition of the subject, the nature of concurrent therapy, if any, and like factors well known to the medical practitioner. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result.

With respect to a subject suffering from, or at risk of developing, light toxicity, such as that due to ocular surgery, an effective amount is an amount sufficient to reduce the rate or extent of formation and accumulation of visual cycle products, such as all-trans-retinal, or lipofuscin, or A2E as well as preventing photocell apoptosis as a result of excessive rhodopsin activation. Here, the compounds of the present invention would be from about 0.01 mg/kg per day to about 1000 mg/kg per day. It is expected that doses ranging from about 50 to about 2000 mg/kg will be suitable. Lower doses will result from certain forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of a composition of the present invention.

A variety of administration routes are available. The methods of the invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. In one preferred embodiment, a composition of the invention is administered intraocularly. Other modes of administration include oral, rectal, topical, intraocular, buccal, intravaginal, intracisternal, intracerebroventricular, intratracheal, nasal, transdermal, within/on implants, or parenteral routes. Compositions comprising a composition of the invention can be added to a physiological fluid, such as to the intravitreal humor. For CNS administration, a variety of techniques are available for promoting transfer of the therapeutic across the blood brain barrier including disruption by surgery or injection, drugs which transiently open adhesion contact between the CNS vasculature endothelial cells, and compounds that facilitate translocation through such cells. Oral administration can be preferred for prophylactic treatment because of the convenience to the patient as well as the dosing schedule.

Pharmaceutical compositions of the invention can optionally further contain one or more additional proteins as desired, including plasma proteins, proteases, and other biological material, so long as it does not cause adverse effects upon administration to a subject. Suitable proteins or biological material may be obtained from human or mammalian plasma by any of the purification methods known and available to those skilled in the art; from supernatants, extracts, or lysates of recombinant tissue culture, viruses, yeast, bacteria, or the like that contain a gene that expresses a human or mammalian plasma protein which has been introduced according to standard recombinant DNA techniques; or from the fluids (e.g., blood, milk, lymph, urine or the like) or transgenic animals that contain a gene that expresses a human plasma protein which has been introduced according to standard transgenic techniques.

Pharmaceutical compositions of the invention can comprise one or more pH buffering compounds to maintain the pH of the formulation at a predetermined level that reflects physiological pH, such as in the range of about 5.0 to about 8.0 (e.g., 6.0, 6.5, 6.8, 6.9, 7.0, 7.1 , 7.2, 7.3, 7.4, 7.5, 7.6, 7.8). The pH buffering compound used in the aqueous liquid formulation can be an amino acid or mixture of amino acids, such as histidine or a mixture of amino acids such as histidine and glycine. Alternatively, the pH buffering compound is preferably an agent which maintains the pH of the formulation at a predetermined level, such as in the range of about 5.0 to about 8.0, and which does not chelate calcium ions. Illustrative examples of such pH buffering compounds include, but are not limited to, imidazole and acetate ions. The pH buffering compound may be present in any amount suitable to maintain the pH of the formulation at a predetermined level.

Pharmaceutical compositions of the invention can also contain one or more osmotic modulating agents, i.e., a compound that modulates the osmotic properties (e.g., tonicity, osmolality and/or osmotic pressure) of the formulation to a level that is acceptable to the blood stream and blood cells of recipient individuals. The osmotic modulating agent can be an agent that does not chelate calcium ions. The osmotic modulating agent can be any compound known or available to those skilled in the art that modulates the osmotic properties of the formulation. One skilled in the art may empirically determine the suitability of a given osmotic modulating agent for use in the inventive formulation. Illustrative examples of suitable types of osmotic modulating agents include, but are not limited to: salts, such as sodium chloride and sodium acetate; sugars, such as sucrose, dextrose, and mannitol; amino acids, such as glycine; and mixtures of one or more of these agents and/or types of agents. The osmotic modulating agent(s) maybe present in any concentration sufficient to modulate the osmotic properties of the formulation. Compositions comprising an opsin-binding or opsin-stabilizing compound of the present invention can contain multivalent metal ions, such as calcium ions, magnesium ions and/or manganese ions. Any multivalent metal ion that helps stabilize the composition and that will not adversely affect recipient individuals may be used. The skilled artisan, based on these two criteria, can determine suitable metal ions empirically and suitable sources of such metal ions are known, and include inorganic and organic salts.

Pharmaceutical compositions of the invention can also be a nonaqueous liquid formulation. Any suitable non-aqueous liquid may be employed, provided that it provides stability to the active agents (a) contained therein. Preferably, the non-aqueous liquid is a hydrophilic liquid. Illustrative examples of suitable non-aqueous liquids include: glycerol; dimethyl sulfoxide (DMSO); polydimethylsiloxane (PMS); ethylene glycols, such as ethylene glycol, diethylene glycol, triethylene glycol, polyethylene glycol ("PEG") 200, PEG 300, and PEG 400; and propylene glycols, such as dipropylene glycol, tripropylene glycol, polypropylene glycol ("PPG") 425, PPG 725, PPG 1000, PEG 2000, PEG 3000 and PEG 4000.

Pharmaceutical compositions of the invention can also be a mixed aqueous/non-aqueous liquid formulation. Any suitable non-aqueous liquid formulation, such as those described above, can be employed along with any aqueous liquid formulation, such as those described above, provided that the mixed aqueous/non-aqueous liquid formulation provides stability to the compound contained therein. Preferably, the non- aqueous liquid in such a formulation is a hydrophilic liquid. Illustrative examples of suitable nonaqueous liquids include: glycerol; DMSO; EMS; ethylene glycols, such as PEG 200, PEG 300, and PEG 400; and propylene glycols, such as PPG 425, PPG 725, PEG 1000, PEG 2000, PEG 3000 and PEG 4000. Suitable stable formulations can permit storage of the active agents in a frozen or an unfrozen liquid state. Stable liquid formulations can be stored at a temperature of at least -70 °C, but can also be stored at higher temperatures of at least 0 °C, or between about 0 °C and about 42 °C, depending on the properties of the composition. It is generally known to the skilled artisan that proteins and polypeptides are sensitive to changes in pH, temperature, and a multiplicity of other factors that may affect therapeutic efficacy.

In certain embodiments a desirable route of administration can be by pulmonary aerosol. Techniques for preparing aerosol delivery systems containing polypeptides are well known to those of skill in the art. Generally, such systems should utilize components that will not significantly impair the biological properties of the antibodies, such as the paratope binding capacity (see, for example, Sciarra and Cutie, "Aerosols," in Remington's Pharmaceutical Sciences 18th edition, 1990, pp 1694-1712; incorporated by reference). Those of skill in the art can readily modify the various parameters and conditions for producing polypeptide aerosols without resorting to undue experimentation. Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of compositions of the invention, increasing convenience to the subject and the physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as polylactides (U.S. Pat. No. 3,773,919; European Patent No. 58,481 ), poly(lactide-glycolide), copolyoxalates polycaprolactones, polyesteramides, polyorthoesters, poiyhydroxybutyric acids, such as poly-D-(- )-3-hydroxybutyric acid (European Patent No. 133,988), copolymers of L- glutamic acid and gamma-ethyl-L-glutamate (Sidman, KR. et at, Biopolymers 22: 547-556), poly (2-hydroxyethyl methacrylate) or ethylene vinyl acetate (Langer, et al., J. Biomed. Mater. Res. 15:267-277; Langer, B.. Chem. Tech. 12:98-105), and polyanhydrides. Other examples of sustained-release compositions include semi ¬ permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules. Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-, di- and tri-glycerides; hydrogel release systems such as biologically-derived bioresorbable hydrogel (i.e., chitin hydrogels or chitosan hydrogels); sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially filled implants; and the like. Specific examples include, but are not limited to: (a) aerosional systems in which the agent is contained in a form within a matrix such as those described in 13.5. Patent Nos. 4,452,775, 4,667,014, 4,748,034 and 5,239,660 and (b) diffusional systems in which an active component permeates at a controlled rate from a polymer such as described in U.S. Patent Nos. 3,832,253, and 3,854,480.

Another type of delivery system that can be used with the methods and compositions of the invention is a colloidal dispersion system. Colloidal dispersion systems include lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Liposomes are artificial membrane vessels, which are useful as a delivery vector in vivo or in vitro. Large unilamellar vessels (LUV), which range in size from 0.2 - 4.0 μητι, can encapsulate large macromolecules within the aqueous interior and be delivered to cells in a biologically active form (Fraley, R., and Papahadjopoulos, D., Trends Biochem. Sci. 6: 77-80).

Liposomes can be targeted to a particular tissue by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein. Liposomes are commercially available from Gibco BRL, for example, as LIPOFECTIN™ and LIPOFECTACE™, which are formed of cationic lipids such as N-[1-(2, 3 dioleyloxy)-propyl]-N,N,N- trimethylammonium chloride (DOTMA) and dimethyl dioctadecylammonium bromide (DDAB). Methods for making liposomes are well known in the art and have been described in many publications, for example, in DE 3,218,121 ; Epstein et al., Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); K. Hwang et al., Proc. Natl, Acad. Sci. (USA) 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641; Japanese Pat. Appl. 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324. Liposomes also have been reviewed by Gregoriadis, G., Trends Biotechnol., 3: 235-241.

Another type of vehicle is a biocompatible microparticle or implant that is suitable for implantation into the mammalian recipient. Exemplary bioerodible implants that are useful in accordance with this method are described in PCT International application no. PCTIUS/03307 (Publication No- WO 95/24929, entitled "Polymeric Gene Delivery System"). PCT/US/0307 describes biocompatible, preferably biodegradable polymeric matrices for containing an exogenous gene under the control of an appropriate promoter. The polymeric matrices can be used to achieve sustained release of the exogenous gene or gene product in the subject.

The polymeric matrix preferably is in the form of a microparticle such as a microsphere (wherein an agent is dispersed throughout a solid polymeric matrix) or a microcapsule (wherein an agent is stored in the core of a polymeric shell). Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Patent 5,075,109. Other forms of the polymeric matrix for containing an agent include films, coatings, gels, implants, and stents. The size and composition of the polymeric matrix device is selected to result in favorable release kinetics in the tissue into which the matrix is introduced. The size of the polymeric matrix further is selected according to the method of delivery that is to be used. Preferably, when an aerosol route is used the polymeric matrix and composition are encompassed in a surfactant vehicle. The polymeric matrix composition can be selected to have both favorable degradation rates and also to be formed of a material, which is a bioadhesive, to further increase the effectiveness of transfer. The matrix composition also can be selected not to degrade, but rather to release by diffusion over an extended period of time. The delivery system can also be a biocompatible microsphere that is suitable for local, site-specific delivery. Such microspheres are disclosed in Chickering, D.B., et al., Biotechnot. Bioeng, 52. 96-101 ; Mathiowitz, B., et at., Nature 386: 410-414.

Both non-biodegradable and biodegradable polymeric matrices can be used to deliver the compositions of the invention to the subject. Such polymers may be natural or synthetic polymers. The polymer is selected based on the period of time over which release is desired, generally in the order of a few hours to a year or longer. Typically, release over a period ranging from between a few hours and three to twelve months is most desirable. The polymer optionally is in the form of a hydrogel that can absorb up to about 90% of its weight in water and further, optionally is cross-linked with multivalent ions or other polymers.

Exemplary synthetic polymers which can be used to form the biodegradable delivery system include: polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluoses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulfate sodium salt, poly(methyl methacrylate), poly(ethyl methacrylate), poly(butyl methacrylate), poly(isobutyl methacrylate), poly(hexyl methacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, polypropylene, poly(ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), polyvinyl alcohols), polyvinyl acetate), polyvinyl chloride), polystyrene, poly(viny Ipyrrolidone), and polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, poly(butic acid), poly(valeric acid), and poly(lactide- cocaprolactone), and natural polymers such as alginate and other polysaccharides including dextran and cellulose, collagen, chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), albumin and other hydrophilic proteins, zein and other prolamines and hydrophobic proteins, copolymers and mixtures thereof. In general, these materials degrade either by enzymatic hydrolysis or exposure to water in vivo, by surface or bulk erosion. Methods of Ocular Delivery

The compositions of the invention are particularly suitable for treating ocular diseases or conditions, such as light toxicity, in particular light toxicity related to an ocular surgical procedure.

In one approach, the compositions of the invention are administered through an ocular device suitable for direct implantation into the vitreous of the eye. The compositions of the invention may be provided in sustained release compositions, such as those described in, for example, U.S. Pat. Nos. 5,672,659 and 5,595,760. Such devices are found to provide sustained controlled release of various compositions to treat the eye without risk of detrimental local and systemic side effects. An object of the present ocular method of delivery is to maximize the amount of drug contained in an intraocular device or implant while minimizing its size in order to prolong the duration of the implant. See, e.g., U.S. Patents 5,378,475; 6,375,972, and 6,756,058 and U.S. Publications 20050096290 and 200501269448. Such implants may be biodegradable and/or biocompatible implants, or may be non-biodegradable implants. Biodegradable ocular implants are described, for example, in U.S.

Patent Publication No. 20050048099. The implants may be permeable or impermeable to the active agent, and may be inserted into a chamber of the eye, such as the anterior or posterior chambers or may be implanted in the sclera, transchoroidal space, or an avascularized region exterior to the vitreous. Alternatively, a contact lens that acts as a depot for compositions of the invention may also be used for drug delivery. In a preferred embodiment, the implant may be positioned over an avascular region, such as on the sclera, so as to allow for transcleral diffusion of the drug to the desired site of treatment, e.g. the intraocular space and macula of the eye. Furthermore, the site of transcleral diffusion is preferably in proximity to the macula. Examples of implants for delivery of a composition of the invention include, but are not limited to, the devices described in U.S. Pat. Nos. 3,416,530; 3,828,777; 4,014,335; 4,300,557; 4,327,725; 4,853,224; 4,946,450; 4,997,652; 5,147,647; 164,188; 5,178,635; 5,300,114; 5,322,691 ; 5,403,901 ; 5,443,505; 5,466,466; 5,476,511 ; 5,516,522; 5,632,984; 5,679,666; 5,710,165; 5,725,493; 5,743,274; 5,766,242; 5,766,619; 5,770,592; 5,773,019; 5,824,072; 5,824,073; 5,830,173; 5,836,935; 5,869,079, 5,902,598; 5,904,144; 5,916,584; 6,001 ,386; 6,074,661 ; 6,110,485; 6,126,687; 6,146.366; 6,251 ,090; and 6,299,895, and in WO 01/30323 and WO 01/28474, all of which are incorporated herein by reference.

Examples include, but are not limited to the following: a sustained release drug delivery system comprising an inner reservoir comprising an effective amount of an agent effective in obtaining a desired local or systemic physiological or pharmacological effect, an inner tube impermeable to the passage of the agent, the inner tube having first and second ends and covering at least a portion of the inner reservoir, the inner tube sized and formed of a material so that the inner tube is capable of supporting its own weight, an impermeable member positioned at the inner tube first end, the impermeable member preventing passage of the agent out of the reservoir through the inner tube first end, and a permeable member positioned at the inner tube second end, the permeable member allowing diffusion of the agent out of the reservoir through the inner tube second end; a method for administering a compound of the invention to a segment of an eye, the method comprising the step of implanting a sustained release device to deliver the compound of the invention to the vitreous of the eye or an implantable, sustained release device for administering a compound of the invention to a segment of an eye; a sustained release drug delivery device comprising: a) a drug core comprising a therapeutically effective amount of at least one first agent effective in obtaining a diagnostic effect or effective in obtaining a desired local or systemic physiological or pharmacological effect; b) at least one unitary cup essentially impermeable to the passage of the agent that surrounds and defines an internal compartment to accept the drug core, the unitary cup comprising an open top end with at least one recessed groove around at least some portion of the open top end of the unitary cup; c) a permeable plug which is permeable to the passage of the agent, the permeable plug is positioned at the open top end of the unitary cup wherein the groove interacts with the permeable plug holding it in position and closing the open top end, the permeable plug allowing passage of the agent out of the drug core, though the permeable plug, and out the open top end of the unitary cup; and d) at least one second agent effective in obtaining a diagnostic effect or effective in obtaining a desired local or systemic physiological or pharmacological effect; or a sustained release drug delivery device comprising: an inner core comprising an effective amount of an agent having a desired solubility and a polymer coating layer, the polymer layer being permeable to the agent, wherein the polymer coating layer completely covers the inner core. Other approaches for ocular delivery include the use of liposomes to target a compound of the present invention to the eye, and preferably to retinal pigment epithelial cells and/or Bruch's membrane. For example, the compound maybe complexed with liposomes in the manner described above, and this compound/liposome complex injected into patients with an ophthalmic condition, such as light toxicity, using intravenous injection to direct the compound to the desired ocular tissue or cell. Directly injecting the liposome complex into the proximity of the retinal pigment epithelial cells or Bruch's membrane can also provide for targeting of the complex with some forms of ocular PCD. In a specific embodiment, the compound is administered via intra-ocular sustained delivery (such as VITRASERT or ENVISION. In a specific embodiment, the compound is delivered by posterior subtenons injection. In another specific embodiment, microemulsion particles containing the compositions of the invention are delivered to ocular tissue to take up lipid from Bruchs membrane, retinal pigment epithelial cells, or both.

Nanoparticles are a colloidal carrier system that has been shown to improve the efficacy of the encapsulated drug by prolonging the serum half- life. Polyalkylcyanoacrylates (PACAs) nanoparticles are a polymer colloidal drug delivery system that is in clinical development, as described by Stella et al, J. Pharm. Sci., 2000. 89: p. 1452-1464; Brigger et al., Tnt. J. Pharm., 2001. 214: p. 37-42; Calvo et al., Pharm. Res., 2001. 18: p. 1157-1166; and Li et al., Biol. Pharm. Bull., 2001. 24: p. 662-665. Biodegradable poly (hydroxyl acids), such as the copolymers of poly (lactic acid) (PLA) and poly (lactic-co- glycolide) (PLGA) are being extensively used in biomedical applications and have received FDA approval for certain clinical applications. In addition, PEG- PLGA nanoparticles have many desirable carrier features including (i) that the agent to be encapsulated comprises a reasonably high weight fraction (loading) of the total carrier system; (ii) that the amount of agent used in the first step of the encapsulation process is incorporated into the final carrier (entrapment efficiency) at a reasonably high level; (iii) that the carrier have the ability to be freeze-dried and reconstituted in solution without aggregation; (iv) that the carrier be biodegradable; (v) that the carrier system be of small size; and (vi) that the carrier enhance the particles persistence.

Nanoparticles are synthesized using virtually any biodegradable shell known in the art. In one embodiment, a polymer, such as poly (lactic-acid) (PLA) or poly (lactic-co-glycolic acid) (PLGA) is used. Such polymers are biocompatible and biodegradable, and are subject to modifications that desirably increase the photochemical efficacy and circulation lifetime of the nanoparticle. In one embodiment, the polymer is modified with a terminal carboxylic acid group (COOH) that increases the negative charge of the particle and thus limits the interaction with negatively charge nucleic acid aptamers. Nanoparticles are also modified with polyethylene glycol (PEG), which also increases the half-life and stability of the particles in circulation. Alternatively, the COOH group is converted to an N-hydroxysuccinimide (NHS) ester for covalent conjugation to amine-modified aptamers.

Biocompatible polymers useful in the composition and methods of the invention include, but are not limited to, polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, poly(viny Ipyrrolidone), polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulfate sodium salt poly-methyl methacrylate), poly(ethyl methacrylate), poly(butyl methacrylate), poly(isobutyl methacrylate\ poly(hexyl methacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), polypheny I methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, polypropylene poly(ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), polyvinyl alcohols), polyvinyl acetate, polyvinyl chloride polystyrene, polyvinyl pyrrolidone), polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacry ( ates), poly (butyl methacrylate), poly(isobutyl methacrylate), poly(hexyl methacrylate) poly(isodecyl methaerylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylatee), poly(isobutyl acrylate), poly(octadecyl acrylate) and combinations of any of these, In one embodiment, the nanoparticles of the invention include PEG-PLGA polymers. Compositions of the invention may also be delivered topically. For topical delivery, the compositions are provided in any pharmaceutically acceptable excipient that is approved for ocular delivery. Preferably, the composition is delivered in drop form to the surface of the eye. For some application, the delivery of the composition relies on the diffusion of the compounds through the cornea to the interior of the eye.

Those of skill in the art will recognize that treatment regimens for using the compounds of the present invention to treat light toxicity or other ophthalmic conditions (e.g., RP) can be straightforwardly determined. This is not a question of experimentation, but rather one of optimization, which is routinely conducted in the medical arts. In vivo studies in nude mice often provide a starting point from which to begin to optimize the dosage and delivery regimes. The frequency of injection will initially be once a week, as has been done in some mice studies. However, this frequency might be optimally adjusted from one day to every two weeks to monthly, depending upon the results obtained front the initial clinical trials and the needs of a particular patient. Human dosage amounts can initially be determined by extrapolating from the amount of compound used in mice, as a skilled artisan recognizes it is routine in the art to modify the dosage for humans compared to animal models. For certain embodiments it is envisioned that the dosage may vary from between about 1 mg compound/Kg body weight to about 5000 mg compound/Kg body weight; or from about 5 mg/Kg body weight to about 4000 mg/Kg body weight or from about 10mg/Kg body weight to about 3000 mg/Kg body weight; or from about 50mg/Kg body weight to about 2000 mg/Kg body weight; or from about 100 mg/Kg body weight to about 1000 mg/Kg body weight; or from about 150 mg/Kg body weight to about 500 mg/Kg body weight. In other embodiments this dose maybe about 1 , 5, 10, 25, 50,75, 100, 150, 10 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1 100, 1 150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1600, 1700, 1800, 1900, 2000, 2500, 3000,. 3500, 4000, 4500, 5000 mg/Kg body weight, in other embodiments, it is envisaged that lower does may be used, such doses may be in the range of about 5 mg compound/Kg body to about 20 mg compound/Kg body. In other embodiments the doses may be about 8, 10, 12, 14, 16 15 or 18 mg/Kg body weight. Of course, this dosage amount may be adjusted upward or downward, as is routinely done in such treatment protocols, depending on the results of the initial clinical trials and the needs of a particular patient.

Screening Assays

Useful compounds of the invention are compounds of the formula (I) that reversibly bind to a native or mutated opsin protein, such as in or near the 11-cis-retinal binding pocket. The non bleachable or slowly bleachable pigment rhodopsins formed from these small molecule opsin bindings will prevent light toxicity related to, for example, the accumulation of visual cycle products as -well as apoptotic photocell death resulting from excessive rhodopsin stimulation. Such binding will commonly inhibit, if not prevent, binding of retinoids, especially 11-cis-retinal, to the binding pocket and thereby reduce formation of visual cycle products, such as all-trans-retinal. Any number of methods are available for carrying out screening assays to identify such compounds. In one approach, an opsin protein is contacted with a candidate compound or test compound that is a non-retinoid in the presence of 11-cis-retinal or retinoid analog and the rate or yield of formation of chromophore is determined. If desired, the binding of the non-retinoid to opsin is characterized. Preferably, the non-retinoid binding to opsin is non- covalent and reversible. Thus, inhibition of rhodopsin formation by a non- retinoid indicates identification of a successful test compound. An increase in the amount of rhodopsin is assayed, for example, by measuring the protein's absorption at a characteristic wavelength (e.g., 498 nm for rhodopsin) or by measuring an increase in the biological activity of the protein using any standard method (e.g., enzymatic activity association with a ligand). Useful compounds inhibit binding of 11-cis-retinal (and formation of rhodopsin) by at least about 10%, 15%, or 20%, or preferably by 25%, 50%, or 75%, or most preferably by up to 90% or even 100%.

Alternatively, the efficacy of compounds useful in the methods of the invention may be determined by exposure of a mammalian eye to a high intensity light source prior to, during, or following administration of a test compound, followed by determination of the amount of visual cycle products (e.g., all-trans retinal, A2E, or lipofuscin) formed as a result of exposure to the high intensity light source, wherein a compound of the invention will have reduced the amount of visual cycle products related to the exposure.

In sum, preferred test compounds identified by the screening methods of the invention are non-retinoids, are selective for opsin and bind in a reversible, non-covalent manner to opsin protein. In addition, their administration to transgenic animals otherwise producing increased lipofuscin results in a reduced rate of production or a reduced accumulation of lipofuscin in the eye of said animal. Compounds identified according to the methods of the invention are useful for the treatment of light toxicity or other ophthalmic condition in a subject, such as a human patient. Combination Therapies

Compositions of the invention useful for the prevention of light toxicity, as well as AMD and retinitis pigmentosa, can optionally be combined with additional therapies as heretofore described.

EXAMPLES

The following non-limiting examples further describe and enable one of ordinary skill in the art to make use of the invention.

Example 1 : (±)-(4afi,9aS)-7-isopropyl-6-methoxy-1 ,1 ,4a-trimethyl- 2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)- ne The title compound, obtained as a colorless oil (463 mg, 51 %), was prepared from 2,6,6-trimethylcyclohex-2-enecarboxylic acid (500 mg, 3.00 mol) and 2-isopropylanisole (1.35 g, 9.00 mmol) according to the procedure of [Tang, S.; Xu, Y.; He, J.; He, Y.; Zheng, J.; Pan, X.; She, X. Org. Lett. 2008, 10, 855-1858]. R f = 0.50 (10:90 ethyl acetate/hexanes); 1 H-NMR (400 MHz, CDC ) δ 7.53 (s, 1 H), 6.76 (s, 1 H), 3.92 (s, 3H), 3.33-3.20 (m, 1 H), 2.13 (s, 1 H), 2.06-1.98 (m, 1 H), 1.66 (ddd, J = 21 .0, 13.0, 6.0 Hz, 2H), 1.53-1.42 (m, 1 H), 1.40-1.34 (m, 2H), 1.28 (s, 3H), 1.23 (s, 3H), 1.20 (d, J = 7.0 Hz, 6H), 0.69 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 301 (M + H + ).

Example 2: (±)-(4a/?,9aS)-6-hydroxy-7-isopropyl-1 ,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1W-fluoren-9(9aW)-one

The title compound, obtained as an off-white solid (134 mg, 70%), was prepared from the product of Example 1 according to the procedure of [Tang, S.; Xu, Y.; He, J.; He, Y.; Zheng, J.; Pan, X.; She, X. Org. Lett. 2008, 10, 1855-1858]. Mp = 206-209 °C; R f = 0.20 (10:90 ethyl acetate/hexanes); 1 H- NMR (400 MHz, CDCI 3 ) δ 10.44 (br s, 1H), 7.32 (s, 1 H), 6.82 (s, 1H), 3.17 (td, J = 13.5, 7.0 Hz, 1 H), 2.02 (s, 1 H), 1.87-1.73 (m, 1 H), 1.69-1.52 (m, 2H), 1.46- 1.26 (m, 3H), 1.21 (s, 3H), 1.16 (m, 9H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 287 (M + H + ).

Example 3: (±)-(4aR,9aS)-6-methoxy-1,1,4a-trimethyl-2,3,4,4a-tetrahydr o- 1 A/-fluoren-9(9aH)-one

The title compound, obtained as a colorless oil (27.0 mg, 25%), was prepared from 2,6,6-trimethylcyclohex-2-enecarboxylic acid (38.0 mg, 0.530 mmol) and anisole (171 mg, 1.58 mmol) according to the procedure of [Tang, S.; Xu, Y.; He, J.; He, Y.; Zheng, J.; Pan, X.; She, X. Org. Lett. 2008, 10, 1855-1858]. R, = 0.30 (10:90 ethyl acetate/hexanes); H NMR (400 MHz, CDCI 3 ) δ 7.62 (d, J = 8.0 Hz, 1 H), 6.85 (m, 2H), 3.88 (s, 3H), 2.15 (s, 1 H), 2.06 (td, J = 15.0, 6.0 Hz, 1 H), 1.73-1.53 (m, 2H), 1.50-1.31 (m, 3H), 1.26 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 259 (MH + ). Enantiomers of the title compounds were resolved via supercritical fluid chromatography on a AS-H (2 x 15 cm) 911 101 column (8% methanol (+0.1%) DEA/CO2, 00 bar eluent; 50 mL/min flow rate).

Example 3a. (+)-(4a/?,9aS)-6-methoxy-1,1,4a-trimethyl-2,3,4,4a- tetrahydro-1 W-fluoren-9(9a H)-one

[a] D 23 = +9.8° (c = 0.61 , MeOH); R f = 0.30 (10:90 ethyl acetate/hexanes); 1 H NMR (400 MHz, CDCI3) δ 7.62 (d, J = 8.0 Hz, 1 H), 6.85 (m, 2H), 3.88 (s, 3H), 2.15 (s, 1 H), 2.06 (td, J = 15.0, 6.0 Hz, 1 H), 1.73-1.53 (m, 2H), 1.50-1.31 (m, 3H), 1.26 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 259 (M + H + ).

Example 3b. (-)-(4aS,9a ?)-6-methoxy-1,1,4a-trimethyl-2,3,4,4a-tetrahydro- 1H-fluoren-9(9aW)-one

[a] D 23 = -9.9° (c = 0.69, MeOH); R f = 0.30 (10:90 ethyl acetate/hexanes); 1 H NMR (400 MHz, CDCI 3 ) δ 7.62 (d, J = 8.0 Hz, 1 H), 6.85 (m, 2H), 3.88 (s, 3H), 2.15 (s, 1 H), 2.06 (td, J = 15.0, 6.0 Hz, 1 H), 1.73-1.53 (m, 2H), 1.50-1.31 (m, 3H), 1.26 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 259 (M + H + ).

Example 4: (±)-(4a ?,9aS)-6-hydroxy-1,1,4a-trimethyl-2,3,4,4a-tetrahydro- 1H-fluoren-9(9aW)-one

The title compound, obtained as a yellow solid (68.0 mg, 41 %), was prepared from the product of Example 3 according to the procedure of [Tang, S.; Xu, Y.; He, J.; He, Y.; Zheng, J.; Pan, X.; She, X. Org. Lett. 2008, 10, 1855-1858]. Mp = 169-170 °C; R, = 0.10 (10:90 ethyl acetate/hexanes); 1 H- NMR (400 MHz, DMSO) δ 10.45 (br s, 1 H), 7.42 (d, J = 8.5 Hz, 1 H), 6.82 (s, 1 H), 6.76 (d, J = 8.5 Hz, 1 H), 2.05 (s, 1 H), 1 .96-1.82 (m, 1 H), 1.67-1.54 (m, 2H), 1.44-1.27 (m, 3H), 1.20 (s, 3H), 1.14 (s, 3H), 0.60 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 245 (M + H + ). Example 5: (±)-(4aR,9aS)-6-ethoxy-1 ,1 ^a-trimethyl^.a^a-tetrahydro- 1 H-fluoren-9(9aH)-one

To a stirred solution of the product of Example 4 (38.0 mg, 0.150 mmol) and potassium carbonate (43.0 mg, 0.310 mmol) in acetonitrile (1.5 ml_) was added iodoethane (27.0 mg, 0.170 mmol). The reaction was heated to 40°C and stirred for 18 hours and then concentrated to from the solvent. The crude residue was purified by preparative plate thin layer chromatography (eluent: 10:90 ethyl acetate/hexanes) to afford a clear oil (33.0 mg, Yield: 73%). Ri = 0.60 (10:90 ethyl acetate: hexanes); H-NMR (400 MHz, CDCI 3 ) δ 7.61 (d, J = 9.0 Hz, 1 H), 6.86-6.81 (m, 2H), 4.11 (q, J = 7.0 Hz, 2H), 2.15 (s, 1 H), 2.10-2.01 (m, 1 H), 1.69-1.56 (m, 2H), 1.44 (dd, J = 13.0, 6.0 Hz, 4H), 1.35 (td, J = 7.0, 4.0 Hz, 2H), 1.25 (s, 3H), 1.21 (s, 3H), 0.63 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 273 (M + H + ).

Example 6: (±)-(4aR,9aS)-1 ,1,4a-trimethyl-6-propoxy-2,3,4,4a-tetrahydro- 1 W-fluoren-9(9aH)-one

The title compound, obtained as a clear oil (54.0 mg, 95%), was prepared from the product of Example 4 according to the procedure of Example 5 except 1-bromopropane was substituted for iodoethane. H f = 0.40 (10:90 ethyl acetate/ hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.61 (d, J = 8.0 Hz, 1 H), 6.84 (d, J = 8.0 Hz, 2H), 4.00 (t, J = 6.5 Hz, 2H), 2.15 (s, 1 H), 2.1 1- 2.02 (m, 1 H), 1.90-1.79 (m, 2H), 1.69-1.56 (m, 2H), 1.44 (m, 1 H), 1.36 (m, 2H), 1.26 (s, 3H), 1.22 (s, 3H), 1.06 (t, J = 7.5 Hz, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 287 (M.+ H + ).

Example 7: (±)-(4a ?,9aS)-6-(allyloxy)-1,1,4a-trimethy|.2,3,4,4a-tetrahydro- 1 H-fluoren-9(9aW)-one

The title compound, obtained as a clear oil (53.9 mg, 94%), was prepared from the product of Example 4 according to the procedure of Example 5 except 3-bromoprop-1-ene was substituted for iodoethane. R f = 0.40 (10:90 ethyl acetate/hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.62 (d, J = 9.0 Hz, 1 H), 6.87 (m, 2H), 6.06 (ddd, J = 16.0, 1 1.0, 5.5 Hz, 1 H), 5.45 (d, J = 16.0 Hz, 1 H), 5.33 (d, J = 1 1.0 Hz, 1 H), 4.62 (d, J = 5.5 Hz, 2H), 2.16 (s, 1 H), 2.09-2.01 (m, 1 H), 1.69-1.56 (m, 2H), 1.50-1.32 (m, 3H), 1.26 (s, 3H), 1.22 (s, 3H), 0.65 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 285 (M + H + ).

Example 8: (+)-(4a/?,9aS)-1 ,1 ,4a-trimethyl-6-(prop-2-ynyloxy)-2,3,4,4a- tetrahydro-1 AV-fluoren-9(9a W)-one

The title compound, obtained as a clear oil (49.0 mg, 88%), was prepared from the product of Example 4 according to the procedure of Example 5 except 3-bromoprop-1-yne was substituted for iodoethane. R f = 0.30 (10:90 ethyl acetate/hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.64 (d, J = 9.0 Hz, 1 H), 6.92 (m, 2H), 4.78 (d, J = 2.5 Hz, 2H), 2.57 (d, J = 2.5 Hz, 1 H), 2.17 (s, 1 H), 2.10-2.01 (m, 1 H), 1.71-1.57 (m, 2H), 1.50-1.34 (m, 3H), 1.27 (s, 3H), 1.22 (s, 3H), 0.65 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 283 (M + H + ).

Example 9: (±)-(4a ?,9aS)-6-(benzyloxy)-1 ,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1 H-fluoren-9(9aW)-one

The title compound, obtained as an off-white solid (17.0 mg, Yield: 30%), was prepared from the product of Example 4 according to the procedure of Example 5 except benzyl bromide was substituted for iodoethane. R f - 0.45 (25:75 ethyl acetate/hexanes); 1 H-NMR (400 MHz, CDCI3) δ 7.63 (d, J = 9.0 Hz, 1 H), 7.48-7.32 (m, 5H), 6.94 (m, 2H), 5.13 (s, 2H), 2.16 (s, 1 H), 2.12-1.99 (m, 1 H), 1.71-1.56 (m, 2H), 1.51-1.32 (m, 3H), 1.26 (s, 3H), 1.22 (s, 3H), 0.65 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 335 (M + H + ).

Example 10: (±)-(4aR,9aS)-1 ,1 ,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-6-yl acetate

The title compound, obtained as a pale yellow oil (25.0 mg, Yield:

68%), was prepared from the product of Example 4 according to the procedure of Example 5 except acetylchloride was substituted for iodoethane, and pyridine was substituted for potassium carbonate. Rf = 0.60 (10:90 ethyl acetate/ hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.68 (d, J = 8.0 Hz, 1 H), 7. 5 (d, J = 2.0 Hz, 1 H), 7.06 (d, J = 8.0 Hz, 1H), 2.33 (s, 3H), 2.20 (s, 1H), 2.06- 1.98 (m, 1 H), 1.71-1.58 (m, 2H), 1.50-1.33 (m, 3H), 1.28 (s, 3H), 1.21 (s, 3H), 0.66 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 287 (M + H + ).

Example 11 : (+)-(4aR,9S,9aS)-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 W-fluoren-9-ol

The product of Example 3 (480 mg, 1.86 mmol) was added to a stirred slurry of lithium aluminum hydride (150 mg, 3.70 mmol) in anhydrous tetrahydrofuran (15 mL) cooled to 0°C under argon. The reaction was heated to reflux for 18 hours.

The reaction was cooled to 0°C, diluted with tetrahydrofuran (30 mL) and quenched by sequentially adding water (0. 5 mL), 15% sodium hydroxide solution (0.15 mL) and water (0.45 mL). Magnesium sulfate was then added to the reaction and the slurry was vigorously stirred for 30 minutes. The reaction was filtered, concentrated and the crude purified by flash column chromatography (eluent: ethyl acetate/hexanes 0:100 -> 10:90) to afford the title compound as a white solid (248 mg, Yield: 51%). R f = 0.40 (10/90 ethyl acetate/ hexanes); H-NMR (400 MHz, CDCI 3 ) δ 7.29 (d, J = 8.0 Hz, 1 H), 6.75 (m, 2H), 5.04 (t, J = 5.5 Hz, 1 H), 3.82 (s, 3H), 1.83-1.63 (m, 4H), 1.60-1.54 (m, 1 H), 1.48-1.44 (m, 1 H), 1.42 (s, 3H), 1 .31-1.28 (m, 1 H), 1.29 (s, 3H), 1.16 (s, 3H), 1.06 (d, J = 5.5 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 12: (±)-6-Methoxy-1,1,4a-trimethyl-2,3,4,4a-tetrahydro-1W- fluorene

The title compound, obtained a clear oil (41.1 mg, Yield: 45%), was prepared from the product of Example 11 according to the procedure of [Tang, S.; Xu, Y.; He, J.; He, Y.; Zheng, J.; Pan, X.; She, X. Org. Lett. 2008, 10, 1855-1858], R f = 0.30 (20:80 dichloromethane/hexanes); 1 H-NMR (400 MHz, CDC ) δ 7.17 (d, J = 8.0 Hz, 1 H), 6.84 (d, J = 2.0 Hz, 1 H), 6.75 (dd, J = 8.0, 2.5 Hz, 1 H), 6.31 (s, 1 H), 3.82 (s, 3H), 2.11 (d, J = 13.0 Hz, 1 H), 1.96 (tq, J = 13.0, 3.5 Hz, 1 H), 1.69-1.59 (m, 2H), 1.37 (s, 3H), 1.29 (s, 3H), 1.24 (s, 3H), 1.01 (dt, J = 13.0, 3.5 Hz, 1 H), 1.12 (dt, J = 13.0, 3.5 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 242 (M + H + ).

Example 13: (+)-(4a ?,9aS)-6-methoxy-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a- hexahydro-1 H-i luorene The product of Example 1 1 (40.0 mg, 0.170 mmol) was added to a stirred slurry of 10% palladium on carbon (~100 mg) in methanol. The reaction flask was capped, evacuated and then charge with a balloon of hydrogen gas. The reaction was stirred at room temperature for 18 hours.

The reaction solution was filtered through Celite and concentrated. The crude residue was purified by preparative plate thin layer chromatography (eluent: 10:90 dichloromethane/hexanes) to afford a clear oil (27.0 mg, Yield: 66%). R, = 0.40 (20:80 dichloromethane/hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.10 (d, J = 7.5 Hz, 1 H), 6.67 (m, 2H), 3.80 (s, 3H), 2.72 (d, J = 8.0 Hz, 2H), 1.87 (t, J = 9.0 Hz, 1 H), 1.61 (t, J = 12.0 Hz, 1 H), 1.49-1.35 (m, 6H), 1.26 (m, 2H), 1.12 (s, 3H), 0.93 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 245 (M + H + ).

Example 14: (±)-(4aR,9aS)-6-(6-chloropyridazin-3-yloxy)- ,1 ,4a-trimethyl- 2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)-one The product of Example 4 (108 mg, 0.440 mmol) was added to a stirred slurry of sodium hydride (19.0 mg, 0.490 mmol, 60% dispersion) in /V,/V-dimethylformamide (0.67 mL). 3,6-Dichloropyridazine (66.0 mg, 0.440 mmol) was added to the reaction mixture, and the reaction was stirred at 60 °C for 18 hours and then heated to 90 °C for 1 hour. The reaction was quenched by pouring into a flask containing ice chips, and the resulting aqueous solution was extracted with ethyl acetate (3 x 30 ml_). The combined organic phases were washed with brine (50 ml_) then dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by preparative plate thin layer chromatography (eluent: dichloromethane/ hexanes = 5:95) to afford a white solid (31.0 mg, Yield: 20%). R f = 0.80 (1 :1 ethyl acetate/ hexanes); 1 H-NMR (400 MHz, CDCI 3 ) δ 7.73 (d, J = 8.5 Hz, 1 H), 7.55 (d, J = 9.0 Hz, 1 H), 7.23 (d, J = 9.0 Hz, 2H), 7.17 (s, 2H), 2.22 (s, 1 H), 2.07-1.98 (m, 1 H), 1.71-1.59 (m, 2H), 1.54 (s, 2H), 1.39 (m, 2H), 1.30 (s, 3H), 1.23 (s, 3H), 0.70 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 357 (M + H + ).

Example 15: 1,1-Dimethyl-2,3,4,9-tetrahydro-1 tf-carbazole

2,2-Dimethylcyclohexanone (1.00 g, 7.90 mmol) was combined, neat, with phenylhydrazine (0.857 g, 7.90 mmol) and heated to 120 °C in an oil bath. Upon completed evolution of steam (~15 minutes) the resulting hydrazone was treated with a 1 :1 (v/v) mixture of acetic acid and concentrated hydrochloric acid (8.0 ml_). The mixture was left at 120 °C until complete evaporation of the liquid phase leaving solid residue (~3 hours).

The residue was dissolved in water (50 ml_) and extracted with diethyl ether (3 x 30 ml_). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give a crude brown solid. The title compound was purified by flash column chromatography (eluent: methanol/ dichloromethane = 5:95) and isolated as a yellow solid (400 mg, Yield: 25%). Mp = 99-104 °C; R f = 0.50 (10:90 ethyl acetate/hexanes); 1 H-NMR (400 MHz, CDCb) δ 7.72 (s, 1 H), 7.47 (d, J = 7.5 Hz, 1 H), 7.30 (d, J = 8.0 Hz, 1 H), 7.1 1 (td, J = 20.0, 7.5 Hz, 2H), 2.70 (t, J = 6.0 Hz, 2H), 1.96-1.86 (m, 2H), 1.75 (dd, J = 7.5, 4.5 Hz, 2H), 1.33 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 200 (M + H + ). Example 16: 1 ,1 ,9-Trimethyl-2,3,4,9-tetrahydro-1H-carbazole

The product of Example 15 (100 mg, 0.500 mmol) was added to a stirred slurry of sodium hydride (40.0 mg, 1.00 mmol, 60% dispersion) in N,N- dimethylformamide (1.5 mL) at 0 °C and stirred for 30 minutes, lodomethane (213 mg, 3.00 mmol) was added to the reaction mixture, and the reaction was stirred at room temperature for another 30 minutes.

The reaction was quenched with water (50 mL), and extracted with chloroform (3 x 10 mL). The combined organic phases were dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by flash column chromatography ( 00% hexanes) to afford a white solid (80.0 mg, Yield: 75%). Mp = 81-83 °C; R, = 0.20 (hexanes); 1 H-NMR (400 MHz, CDGI3) δ 7.48 (d, J = 7.5 Hz, 1 H), 7.27 (s, 1 H), 7.18 (t, J = 7.5 Hz, 1 H), 7.08 (t, J = 6.5 Hz, 1 H), 3.82 (s, 3H), 2.72 (s, 2H), 1.86 (d, J = 4.5 Hz, 2H), 1.76 (d, J = 6.5 Hz, 2H), 1.44 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 214 (M + H + ).

Example 17: P-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a solution of p-tolylmagnesium bromide (1 M in tetrahydrofuran, 33 mL, 33 mmol) in diethyl ether (32 mL) at 0 °C was added a solution of 2,6,6- trimethylcyclohex-1-enecarbaldehyde (2.0 g, 13.14 mmol) in diethyl ether (16 mL) drop wise over 5 min. The reaction mixture was allowed to stir at room temperature for 1.5 hours. Then the reaction mixture was poured into water (100 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a colorless oil (2.9 g, Yield: 91 %). 1 H NMR (400 MHz, CDC ) δ 7.30 (d, J = 8.0 Hz, 2H), 7.12 (d, J = 8.4 Hz, 2H), 5.39 (d, J = 4.8 Hz, 1 H), 2.34 (s, 3H), 1.99 (t, J = 6.0 Hz, 2H), 1.81 (d, J = 4.8 Hz, 1 H), 1.70 - 1.61 (m, 2H), 1.56-1.52 (m, 2H), 1.40 (s, 3H), 1.19 (s, 3H), 1.04 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 227 (M - H 2 0 + H + ). Example 18: P-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 17 (100 mg, 0.41 mmol) in dichloromethane (5 mL) was added manganese dioxide (356 mg, 4.1 mmol). The reaction mixture was stirred at room temperature overnight. The mixture was then filtered and the filtrate was concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography to give the title compound as a colorless oil (72 mg, Yield: 73%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.85 (d, J = 8.4 Hz, 2H), 7.27 (d, J = 8.0 Hz, 2H), 2.43 (s, 3H), 2.10 (t, J = 6.4 Hz, 2H), 1.85 - 1.73 (m, 2H), 1.63 - 1 .53 (m, 2H), 1 .46 (s, 3H), 1.05 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 19: (±)-(4aR,9aS)-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one

A solution of the product of Example 18 (100 mg, 0.44 mmol) in methanesulfonic acid (2 mL) was stirred at 50 °C for 1.5 hours. Water (15 mL) was added to the reaction mixture and it was extracted with ethyl acetate (3 x10 mL). The organic layer was washed with brine (5 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid. (84 mg, Yield: 84%). Mp = 81-84 °C; 1 H NMR (400 MHz, CDCI 3 ) δ 7.56 (d, J = 7.6 Hz, 1 H), 7.20 (s, 1 H), 7.14 (d, J = 7.6 Hz, 1 H), 2.44 (s, 3H), 2.16 (s, 1 H), 2.1 1 - 2.07 (m, 1 H), 1.67 - 1.63 (m, 2H), 1.45 - 1 1.33 (m, 3H), 1.25 (s, 3H), 1.21 (s, 3H), 0.63 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 20: (±)-(4a ,9aS)-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aW)-one oxime

To a solution of the product of Example 19 (100 mg, 0.413 mmol) in pyridine (3 mL) was added hydroxylamine hydrochloride (143 mg, 2.06 mmol). The mixture was refluxed for 1.5 hours after which another 143 mg of hydroxylamine hydrochloride was added and the reaction was refluxed for an additional 3 hours. The reaction mixture was concentrated under reduced pressure and the obtained residue was purified by preparative thin layer chromatography to give the title compound as a white solid (87 mg, Yield: 82%). Mp = 144-146 °C; H NMR (400 MHz, CDCI 3 ) δ 8.13 (d, J = 8 Hz, 0.93H, major product), 7.42 (d, J = 8 Hz, 0.07H, minor product), 7.07 (d, J = 8 Hz, 0.93H, major product), 7.04 (s, 1 H), 7.02 (d, J = 8 Hz, 0.07H, minor product), 2.39 (s, 0.93H, major product), 2.38 (s, 0.07H, minor product), 2.32 (d, J = 16 Hz, 1 H), 2.30 (s, 1 H), 1.64 -1.50 (m, 2H), 1.38 -1.25 (m, 3H), 1.20(s, 3H), 1.07 (s, 3H), 0.41 (s, 0.21 H, minor product), 0.33 (s, 2.79H, major product) ppm; Mass spectrum (ESI +ve) m/z 258 (M + H + ). Example 21 : (±)-(4a ?,9aS)-1 ,1,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one O-methyl oxime

To a solution of the product of Example 20 (40 mg, 0.136 mmol) in tetrahydrofuran (1 mL) at 0 °C was added sodium hydride (5 mg, 0.187 mmol) and the mixture was stirred at room temperature for 2 hours. Then a solution of methyl iodide (40 mg, 0.272 mmol) in tetrahydrofuran (1 mL) was added to the reaction mixture and it was stirred at room temperature for 2 hours. Water (2 mL) was added to quench the reaction. The mixture was concentrated and the residue was partitioned between ethyl acetate (30 mL) and water (3 mL). The organic layer was washed with brine (2 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a white solid. (15 mg, Yield: 36%). Mp = 92-94 °C; major product: R f = 0.4 in (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.99 (d, J = 7.6 Hz, 0.83H, major product), 7.43 (d, J = 7.6 Hz, 0.13H, minor product), 7.03 (d, J - 8.0 Hz, 1 H), 7.00 (s, 1 H), 3.99 (s, 2.53H, major product), 3.94 (s, 0.44H, minor product), 2.97 (s, 0.13H, minor product), 2.37 (s, 3H), 2.30-2.34 (m, 1 H), 2.27 (s, 0.83H, major product), 1.42-1.48 (m, 2H), 1.29-1.40 (m, 1 H), 1.24-1.30 (m, 2H), 1.10 (s, 3H), 1.04 (s, 3H), 0.38 (s, 0.44H, minor product), 0.30 (s, 2.52H, major product) ppm; Mass spectrum (ESI +ve) m/z 272 (M + H + ). Example 22: (±)-(4aR,9R,9aS)-1 ,1 ,4a,6-tetramethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 19 (250 mg, 1.032 mmol) in methanol (6 mL) was added sodium borohydride (1 17 mg, 3.1 mmol) portion wise in an ice bath under argon. The mixture was stirred overnight at room temperature. The reaction mixture was concentrated and the residue was partitioned between water and ethyl acetate (2 x 20 mL). The combined organic layers were washed with brine (10 mL), dried over magnesium sulfate and concentrated. The obtained residue was purified by silica gel chromatography to give the title compound as white solid (114 mg, Yield: 45%). Mp = 99-101 °C; R f = 0.3 (50:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDC ) δ 7.28 (d, J = 7.6 Hz, 1 H), 7.06 (d, J = 7.6 Hz, 1 H), 6.94 (s, 1 H), 5.00 (dd, Ji = 17.2 Hz, J 2 = 8.8 Hz, 1 H), 2.36 (s, 3H), 1.63 (t, J = 7.6 Hz, 3H), 1.58 (s, 1 H), 1.54 (s, 3H), 1 .39 (t, J = 8.7 Hz, 2H), 1.19 (s, 3H), 1.16 (s, 3H), 1.05 - 0.92 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 227 (M - H 2 0 + H + ).

Example 23: (±)-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1H-fluorene

To a solution of the product of Example 22 (80 mg, 0.33 mmol) in anhydrous dichloromethane (1 mL) at 0 °C was added thionyl chloride (50 yL) drop wise. The mixture was stirred for 2 hours at room temperature. Then the reaction mixture was diluted in dichloromethane (15 mL) and washed with water (5 mL), saturated sodium bicarbonate, water (5 mL) and brine (5 mL), dried over magnesium sulfate and concentrated. The obtained residue was purified by silica gel chromatography to afford the title compound as a colorless oil (64 mg, Yield: 86%). R f = 0.95 (100/1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.16 (d, J = 7.6 Hz, 1 H), 7.06 (s, 1 H), 7.01 (d, J = 7.5 Hz, 1 H), 6.33 (s, 1 H), 2.38 (s, 3H), 2.17 - 2.10 (m, 1 H), 2.04 - 1.90 (m, 1 H), 1.68 - 1.58 (m, 2H), .37 (s, 3H), 1.30 (s, 3H), 1.25 (s, 3H), 1.1 1 (td, J = 13.2, 3.6 Hz, 1 H), 0.99 (td, J = 13.2, 3.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 227 (M + H + ). Example 24: (±)-(4aR,9aS)-1 ,1 ,4a,6-tetramethyl-2,3,4,4a,9,9a-hexahydro- 1H-fluorene

To a solution of the product of Example 23 (34 mg, 0.15 mmol) in methanol (1 ml_) and tetrahydrofuran (2 ml_) was added palladium on carbon (5 mg). The mixture was stirred under a hydrogen atmosphere overnight. Then the reaction mixture was filtered under reduced pressure and the filtrate was concentrated and purified by silica gel chromatography to afford the title compound as colorless oil (33 mg, Yield: 96%). R f = 0.95 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) 6 7.1 1 (d, J = 1.2 Hz, 1 H), 6.96 (d, J = 8.4 Hz, 1 H), 6.93 (s, 1 H), 2.74 (d, J = 10.0 Hz, 2H), 2.36 (s, 3H), 1.84 (t, J = 9.6 Hz, 1 H), 1.60 (t, J = 13.2 Hz, 1 H), 1.46 (s, 3H), 1.49-1.38 (m, 2H), 1.32-1.22 (m, 3H), 1.1 1 (s, 3H), 0.90 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 229 (M + H + ).

Example 25: 1 -(methoxy(2,6,6-trimethy lcyclohex-1 -enyl)methyl)-4- methylbenzene

To a stirred solution of the product of Example 17 (50 mg, 0.204 mmol) in dichloromethane (7 ml_) at 0 °C was added proton sponge (480 mg, 2.24 mmol), followed by trimethyloxonium tetrafluoroborate (274 mg, 1.843 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate (10 ml_). The organic phase was separated and the aqueous phase was extracted with dichloromethane (30 mL x 2). The combined organic phase was washed with 5% aqueous hydrochloric acid (3 x 5 mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by silica gel column chromatography to afford the title compound as colorless oil (20 mg, Yield: 38%). R f = 0.7 (50:1 petroleum ether/ethyl acetate);. 1 H NMR (400 MHz, CDCI 3 ) 5 7.29 (d, J = 8.0 Hz, 2H), 7.14 (d, J = 8.0 Hz, 2H), 4.94 (s, 1 H), 3.44 (s, 3H), 2.35 (s, 3H), 2.09 (t, J = 5.6 Hz, 2H), 1.75 - 1.63 (m, 2H), 1.57 - 1.49 (m, 2H), 1.14 (s, 3H), 0.91 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 281 (M + Na + ). Example 26: 1 ,1 ,4a,6,9-pentamethyl-2,3,4,4a-tetrahydro-1 H-fluorene

To a solution of the product of Example 19 (100 mg, 0.413 mmol) in tetrahydrofuran (3 mL) was added 3 M methyl magnesium iodide (0.7 mL) under argon with stirring. Then the mixture was heated to reflux for 5 hours. Water (10 mL) was added to quench the reaction. The mixture was concentrated to remove the tetrahydrofuran and the residue was partitioned between ethyl acetate (20 mL) and water (3 mL). The organic layer was washed with brine (2 mL), dried (sodium sulfate) and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a colorless oil (14 mg, Yield: 14%). R f = 0.95 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.14 (d, J - 7.6 Hz, 1 H), 7.06 (d, J = 7.6 Hz, 1 H), 7.05 (s, 1 H), 2.39 (s, , 3H), 2.23 (s, 3H), 2.12 (dd, J = 7.4, 6.0 Hz, 1 H), 1.95 (ddd, J = 23.2, 13.1 , 4.1 Hz, 1 H), 1 .68 - 1.63 (m, 1 H), 1.59 (d, J = 14.8 Hz, 1 H), 1.46 (s, 3H), 1 .35 (s, 3H), 1.30 (s, 3H), 1.29 (s, 1 H), 1.13 (td, J = 12.7, 4.2 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 241 (M + H + ).

Example 27: (±)-(4aR,9S,9aS)-1 ,1 ,4a,6,9-pentamethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluorene

To a solution of the product of Example 26 (40 mg, 0.166 mmol) in methanol (2 mL) and tetrahydrofuran (2 mL) was added Pd/C (5 mg). The mixture was stirred under a hydrogen atmosphere overnight. Then the reaction mixture was filtered under reduced pressure and the filtrate was concentrated and purified by silica gel chromatography to give the title compound as colorless oil (20 mg, Yield: 50%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.04 (d, J = 7.6 Hz, 1 H), 6.96 (d, J = 7.6 Hz, 1 H), 6.93 (s, 1 H), 3.35-3.25 (m, 1 H), 2.34 (s, 3H), 1.89 (d, J = 8.0 Hz, 1 H), 1.78-1.70 (m, 1 H), 1.58-1.45 (m, 3H), 1.40 (s, 3H), 1.38 (d, J = 7.2 Hz, 3H), 1 .28-1.18 (m, 2H), 1.13 (s, 3H), 1 .06 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 28: phenyl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a solution of phenylmagnesium bromide (3 M in ether, 8.21 mL, 24.64 mmol) in diethyl ether (24 mL) at 0 °C was added a solution of 2,6,6- trimethylcyclohex-1-enecarbaldehyde (1.5 g, 9.85 mmol) in diethyl ether (12 mL) drop wise over 5 min. The reaction mixture was allowed to stir at room temperature for 1.5 hours. Then the reaction mixture was poured into water (80 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (3 x 80 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a colorless oil (2.2 g, Yield: 97%). H NMR (400 MHz, CDCI 3 ) δ 7.43 (d, J = 8.0 Hz, 2H), 7.32 (dd, J = 8.4, 7.2 Hz, 2H), 7.21 (t, J = 7.2 Hz, 1 H), 5.42 (d, J = 4.8 Hz, 1 H), 1.99 (t, J = 6.0 Hz, 2H), 1.83 (d, J = 4.8 Hz, 1 H), 1.71 - 1.60 (m, 2H), 1.57 - 1.49 (m, 2H), 1.38 (s, 3H), 1.20 (s, 3H), .07 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 2 3 (M - H 2 0 + H + ).

Example 29: phenyl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 28 (0.8 g, 3.47 mmol) in dichloromethane (50 mL) was added manganese dioxide (3.0 g, 34.7 mmol). The reaction mixture was stirred at room temperature overnight. Then the mixture was filtered and the filtrate was concentrated under reduced pressure. The obtained residue was purified by chromatography to give the title compound as a colorless oil (0.64 g, Yield: 81 %). 1 H NMR (400 MHz, CDCI 3 ) δ 7.94 (d, J = 7.6 Hz, 2H), 7.54 (t, J = 8.4 Hz, 1 H), 7.47-7.43 (m, 2H), 2.08 (t, J = 6.4 Hz, 2H), 1.79-1.75 (m, 2H), 1.58 - 1.52 (m, 2H), 1.44 (s, 3H), 1.05 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 229 (M + H + ). Example 30: (±)-(4a ,9aS)-1 ,1,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aH)-one

A solution of the product of Example 29 (100 mg, 0.44 mmol) in methanesulfonic acid (2 mL) was stirred at 50 °C for 1.5 hours. The reaction mixture was quenched with water (15 mL) and organics were extracted with ethyl acetate (3 x 10 mL). The organic layer was washed with brine (5 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a yellow oil (70 mg, Yield: 70%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.68 (d, J = 8.0 Hz, 1 H), 7.57 (dd, J = 7.2, 8.0 Hz, 1 H), 7.42 (d, J = 7.2 Hz, 1 H), 7.36-7.31 (m, 1 H), 2.18 (s, 1 H), 2.12 - 2.08 (m, 1 H), 1.71 - 1.59 (m, 2H), 1.45 -1.34 (m, 3H), 1.27 (s, 3H), 1.21 (s, 3H), 0.63 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 229 (M + H + ).

Example 31 : (+)-(4aR,9R,9aS)-1 ,1 ^a-trimethyl-Z.a. a.S^a-hexahydro- 1H-fluoren-9-ol

To a solution of the product of Example 30 (250 mg, 1.1 mmol) in methanol (6.0 mL) at 0 °C was added sodium borohydride (150.0 mg, 3.96 mmol). The resulting mixture was allowed to gradually warm to room temperature and stirred overnight. The reaction mixture was concentrated under reduced pressure. Water (20 mL) was added and the mixture was extracted with ethyl acetate (30 mL x 2). The combined organic phase was washed with brine (30 mL), dried over sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: hexane/ethyl acetate = 60:1) to afford the title compound as a white solid (160 mg, Yield 63%). Mp = 107-108 °C; R f = 0.6 (60/1 hexane/ethyl acetate); 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.31-7.29 (m, 1 H), 7.21-7.12 (m, 3H), 5.05 (d, J = 7.6 Hz, 1 H), 4.83 (t, J = 7.6 Hz, 1 H), 1.63- 1.48 (m, 3H), 1.43 (s, 3H), 1.37-1.27 (m, 3H), 1.12 (s, 3H), 1.11 (s, 3H), 0.91 (td, J = 13.6, 3.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 213 (M - H 2 0 +

H + ).

Example 32: (±)-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene

To a solution of the product of Example 31 (95 mg, 0.41 mmol) in dichloromethane (2.0 mL) at 0 °C was added thionyl chloride (60 μΐ) drop wise at 0 °C. The reaction mixture was allowed to room temperature and stirred for 3 hours. Dichloromethane (10 mL) was added and the organic phase was washed with saturated aqueous sodium bicarbonate (20 mL x 2) and brine (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: hexane) to afford the title compound as a colorless oil (75 mg, Yield: 87%). R f = 0.8 (hexane); 1 H NMR (400 MHz, DMSO) δ: 7.30-7.22 (m, 2 H), 7.21-7.18 (m, 1 H), 7.15-7.12 (m, 1 H), 6.39 (s, 1 H), 2.19-2.15 (m, 1 H), 2.01-1.93 (m, 1 H), 1.70-1.58 (m, 2 H), 1.39 (s, 3H), 1.32 (s, 3H), 1.27 (s, 3H), 1.12 (dt, J = 13.6, 4.0 Hz, 1 H), 1.01 (dt, J = 13.6, 4.0 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 213 (M + H + ).

Example 33: (±)-(4aR,9aS)-1 ,1 ^a-trimethyl-a^^^a-tetrahydro-l H- fluoren-9(9aH)-one oxime

To a solution of the product of Example 30 (100 mg, 0.44 mmol) in pyridine (3.0 mL) was added hydroxylamine hydrochloride (285 mg, 4.4 mmol) and the resulting mixture was refluxed for 6 hours. The pyridine was evaporated under reduced pressure and the residue was purified by preparative thin layer chromatography (eluent: hexane/ethyl acetate = 20:1) to afford the title compound as a white solid (42 mg, Yield: 40%). Mp = 135-137 °C; 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.95 (s, 1H), 8.17 (d, J = 8.0 Hz, 1H), 7.38-7.29 (m, 2H), 7.26-7.22 (m, 1 H), 2.36-2.32 (m, 1 H), 2.22 (s, 1 H), 1.53- 1.42 (m, 2H), 1.34-1.14 (m, 4H), 1.04 (s, 3H), 1.02 (s, 3H), 0.22 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 244 (M + H + ).

Example 34: (±)-(4aR,9aS)-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1 H- fluorene

To a solution of the product of Example 32 (65 mg, 0.31 mmol) in methanol (3.0 mL) was added palladium on carbon (10.0 mg) and the mixture was stirred at room temperature overnight under an atmosphere of hydrogen. The catalyst was filtered off and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: hexane) to afford the title compound as a colorless oil (60 mg, Yield: 90%) R f = 0.9 (hexane); 1 H NMR (400 MHz, CDCI 3 ) δ: 7.21-7.08 (m, 4 H), 2.78 (d, J = 9.6 Hz, 2 H), 1.87 (t, J = 9.6 Hz, 1 H), 1.65-1.61 (m, 1 H), 1.48- 1.35 (m, 3H), 1.44 (s, 3 H), 1.31-1.20 (m, 2 H), 1.12 (s, 3 H), 0.94 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 215 (M + H + ). Example 35: (±)-(4aR,9S,9aS)-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 30 (25 mg, 0.11 mmol) in tetrahydrofuran (2.0 mL) was added methyl magnesium iodide (0.18 mL, 0.55 mmol) at 0 °C and the resulting mixture was stirred for 5 hours. The solution was poured into ice water (5.0 mL), and to the solution was added ethyl acetate (10 mL). The organic phase was separated washed with saturated aqueous ammonium chloride (5.0 mL), water (10 mL) and brine (20 mL). The organic phase was dried over anhydrous sodium sulfate and the volatiles evaporated under reduced pressure. The residue was purified by preparative thin layer chromatography (eluent: hexane/ethyl acetate = 15:1 ) to give the title compound .as a white solid (8 mg, Yield: 30%). Mp = 98-99 °C; R f = 0.6 (50/1 hexane/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.32 (t, J = 8.0 Hz, 1 H), 7.29-7.21 (m, 2H), 7.17 (d, J = 8.0 Hz, 1 H), 1.78-1.65 (m, 5H), 1.56 (s, 1 H), 1.55-1.49 (m, 1 H), 1.47 (s, 3H), 1.37 (s, 3H), 1.33-1.21 (m, 3H), 1.18 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 227 (M - H 2 0 + H + ).

Example 36: (±)-1 ,1 ,4a,9-tetramethyl-2,3,4,4a-tetrahydro-1 H-fluorene

To a solution of the product of Example 35 (24 mg, 0.10 mmol) in dichloromethane (2.0 mL) at 0 °C was added thionyl chloride (40 μΐ) and the resulting solution was stirred at room temperature for 2 hours. Saturated aqueous sodium bicarbonate (10 mL) was added and the reaction mixture was extracted with dichloromethane (10 mL x 2). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: hexane) to afford the title compound as a colorless oil (15 mg, Yield: 70%). R f = 0.9 (hexane); 1 H NMR (400 MHz, CDC ) δ: 7.26-7.14 (m, 4 H), 2.23 (s, 3 H), 2.16-2.13 (m, 1 H), 1.99-1.91 (m, 1 H), 1.67-1.58 (m, 2 H), 1.47 (s, 3 H), 1.36 (s, 3 H ), 1.32 (s, 3 H), 1.30-1.25 (m, 1 H), 1.13 (dt, J = 12.8, 4.0 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 215 (M + H + ).

I l l Example 37: (±)-(4aR,9aS)-1 ,1 ,4a-trimethy l-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one O-methyl oxime

To a 0 °C solution of the product of Example 33 (42 mg, 0.17 mmol) in tetrahydrofuran (2.0 mL) was added sodium hydride (8.2 mg, 0.2 mmol). The reaction mixture was allowed to room temperature and stirred for 2 hours after which time the solution was cooled back down to 0 °C and methyl iodide (48.3 mg, 0.34 mmol) was added. The solution was allowed to warm to room temperature and was stirred for an additional 2 hours. Water (5 mL) was added and the organics extracted with ethyl acetate (10 mL x 2). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent: hexane/ethyl acetate = 60: 1 ) to afford the title compound as a colorless oil (18 mg, Yield: 42%). 1 H NMR (400 MHz, DMSO, major product) δ: 8.13 (d, J = 8.0 Hz, 1 H), 7.36-7.32 (m, 1 H), 7.26-7.17 (m, 2 H), 4.01 (s, 3 H), 2.35-2.30 (m, 2 H), 1.56-1.48 (m, 2 H), 1.38-1.20 (m, 4H), 1.12 (s, 3 H). 1.09 (s, 3 H), 0.29 (s, 3 H) ) ppm; Mass spectrum (ESI +ve) m/z 258 (M + H + ).

Example 38: (methoxy(2,6,6-trimethy lcyclohex-1 -enyl)methyl)benzene To a stirred solution of the product of Example 28 (20 mg, 0.09 mmol) in dichloromethane (3.0 mL) at 0 °C was added proton sponge (212.2 mg, 0.99 mmol), followed by trimethyloxonium tetrafluoroborate (120 mg, 0.81 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate (5 mL). The phases were separated and the aqueous phase was extracted with dichloromethane (30 mL x 2). The combined organic phase was washed with saturated ammonium chloride (20 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: hexane/ethyl acetate = 60:1 ) to yield the title compound as a colorless oil. (17 mg, Yield: 77%). R f = 0.6 (20:1 hexane/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ: 7.32 (d, J = 7.2 Hz, 2H), 7.25 (d, J = 7.6 Hz, 2H), 7.20-7.16 (m, 1 H), 4.90 (s, 1 H), 3.38 (s, 3 H), 2.01 (t, J = 6.4 Hz, 1 H), 1.63-1.58 (m, 2 H), 1.50 (s, 3 H ), 1.47-1.44 (m, 2 H), 1.06 (s, 3 H), 0.85 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 267 (M + Na + ).

Example 39: (4-chlorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol To a solution of (4-chlorophenyl)magnesium bromide (33 mL, 32.75 mmol) in diethyl ether (32 mL) at -78 °C was added 2,6,6-trimethylcyclohex-1- enecarbaldehyde (2.0 g, 13.1 mmol) in diethyl ether (16 mL) drop wise. The mixture was stirred at that temperature for 2 hours. The reaction mixture was quenched with water (10 mL) and the organics extracted with ethyl acetate (3 x 50 mL). The combined organic phase was washed with water (50 mL) and brine (30 mL), dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue was purified by silica gel chromatography (eluent: petroleum ether/ethyl acetate = 100:1) to afford the title compound as a colorless oil (3.3 g, Yield: 95%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.34 (d, J = 8.8 Hz, 2H), 7.26 (d, J = 8.8 Hz, 2H), 5.34 (d, J = 4.8 Hz, 1 H), 2.42 (t, J = 6.4 Hz, 2H), 2.25 (d, J = 4.8 Hz, 1 H), 1.62-1.61 (m, 2H), 1 .55-1.51 (m, 2H), 1.34 (s, 3H), 1.17 (s, 3H), 1.04 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 247 (M - H 2 0 + H + ). Example 40: (4-chlorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanone

A mixture of the product of Example 39 (1.0 g, 3.8 mmol) and manganese dioxide (3.3 g, 38 mmol) in dichloromethane (50 mL) was stirred at room temperature for 48 hours. The reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (eluent: petroleum ether/ethyl acetate = 00:1) to afford the title compound as a yellow oil. (700 mg, Yield: 70%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.87 (d, J = 8.0 Hz, 2H), 7.42 (d, J = 8.0 Hz, 2H), 2.07 (t, J = 6.4 Hz, 2H), 1.82 - 1.70 (m, 2H), 1.55-1.53 (m, 2H), 1.42 (s, 3H), 1.02 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 263 (M + H + ).

Example 41 : (±)-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-fluorene

To a mixture of the product of Example 39 (100 mg, 0.38 mmol) in dichloromethane (6 mL) at 0 °C was added stannic chloride (148 mg, 0.57 mmol). The reaction was stirred at 0 °C for 5 minutes, and then the mixture was stirred at room temperature for 2 hours. The reaction mixture was cooled to 0 °C, and water (1 mL) was added to quench the reaction and the organic volatiles were evaporated in vacuo. The organics were extracted from the aqueous residue with diethyl ether (20 mL x 3) and the combined organic layer was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), water (20 mL) and then brine (20 mL). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford the title compound as a yellow oil (88 mg, Yield: 95%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.20-7.17 (m, 3H), 6.32 (s, 1 H), 2.14 - 1.93 (m, 2H), 1.67 - 1.61 (m, 2H), 1.36 (s, 3H), 1.33 (s, 3H), 1 .30 (s, 3H), 1.1 1-0.98 (m, 2H) ) ppm; Mass spectrum (ESI +ve) m/z 246 (M + H + ).

Example 42: (±)-(4aR,9a )-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1H-fluorene

Example 42a: (5aR)-8-chloro-2,2,5a-trimethyl-2,3,4,5,5a,9b-hexahydro- fluoreno[8a,9-b]oxirene

To a mixture of the product of Example 41 (72 mg, 0.29 mmol) and sodium bicarbonate (49 mg, 0.59 mmol) in dichloromethane (6 mL) at 0 °C was added meta-chloroperoxybenzoic acid (76 mg, 0.44 mmol). The reaction was stirred at 0 °C for 30 minutes, allowed to warm to room temperature and stirred for an additional 2 hours. The mixture was cooled to 0 °C and then saturated aqueous sodium bicarbonate (2 mL) was added to quench the reaction. The volatiles were evaporated in vacuo and the residue was taken up in diethyl ether (60 mL). The organic phase was washed with water ( 0 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2) and brine (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to afford the title compound that was directly used in the next step. Example 42b: (+)-(4a/?,9aR)-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluorene

To the product of Example 42a in dichloromethane (6 mL) at 0 °C was added boron trifluoride etherate (0.1 mL, 0.788 mmol). The reaction mixture was stirred at 0 °C for 30 minutes, allowed to warm to room temperature and then stirred for an additional 2 hours. The mixture was cooled to 0 °C and water (2 mL) was added to quench the reaction. The reaction was concentrated under reduced pressure and the residue was taken up in diethyl ether (60 mL). The organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by preparative thin layer chromatography and then subsequently by prep-HPLC to afford the title compound as a yellow oil. (26 mg, Yield: 34%). 1 H N R (400 MHz, CDCI 3 ) δ 7.23-7.22 (m, 2H), 7.16 (s, 1 H), 3.07 (s, 1 H), 1.70 (m, 2H), 1.40 (m, 5H), 1.23 (m, 4H), 1.06 (m, 4H) ppm; Mass spectrum (ESI +ve) m/z 262 (M + H + ).

Example 43: (±)-(4aR,9R,9aR)-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a mixture of the product of Example 42b (50 mg, 0.19 mmol) in methanol (2 mL) at 0 °C was added sodium borohydride (21.6 mg, 0.57 mmol). The reaction mixture stirred at 0 °C for 30 minutes, allowed to warm to room temperature, and stirred overnight. The mixture was cooled to 0 °C, water (20 mL) was added to quench the reaction, and then volatiles were evaporated in vacuo. The residue was taken up in ethyl acetate (60 mL) and the organic phase was washed with water (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue was purified by preparative thin layer chromatography to afford the title compound as a colorless oil (40 mg, Yield: 80%) R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, DMSO) δ 7.23-7.13 (m, 3H), 5.31 (d, J = 6.0 Hz, 1 H), 4.04 (m, 1 H), 2.75 (d, J = 8.0 Hz, 1 H), 2.05 (t, J = 12.8 Hz, 1 H), 1.84 (m, 1 H), 1.39-1.09 (m, 9H), 0.85 (s, 3H), 0.60 (t, J = 12.8 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 248 (M - H 2 0 + H + ). Example 44: (±)-(4a ?,9R,9aR)-6-chloro-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a mixture of the product of Example 42 (50 mg, 0.19 mmol) in tetrahydrofuran (3 mL) at 0 °C was added 3M methyl magnesium iodide (0.32 mL, 0.95 mmol). The reaction was stirred at room temperature overnight. The solvent was evaporated under reduced pressure and 20 mL of saturated aqueous ammonium chloride was added. The organics were extracted with ethyl acetate (20 mL x 3) and the combined organic phase was washed with water (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and then the volatiles were evaporated in vacuo to afford the title compound as a yellow oil (50 mg, Yield: 96%). R f = .0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.09 (d, J = 8.0 Hz, 1 H), 7.05 (dd, J = 8.0, 1.6 Hz, 1 H), 6.96 (d, J = 1.6 Hz, 1 H), 2.55 (s, 1 H), 1.98-1.94 (m, 2H), 1.55-1.50 (m, 1 H), 1 .44-1.40 (m, 2H), 1.24 (s, 3H), 1.19 (s, 3H), 1.14 (s, 3H), 0.93 (s, 3H), 0.76-0.72 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 262 (M - H 2 0 + H + ).

Example 45: (±)-(4aR,9 ?,9a ?)-6-chloro-9-ethyl-1 ,1 ,4a-trimethyl- 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol

To a mixture of the product of Example 42 (50 mg, 0.19 mmol) in tetrahydrofuran (3 mL) at 0 °C was added 3M ethyl magnesium iodide (0.32 mL, 0.95 mmol). The reaction was warmed to room temperature and stirred overnight. The volatiles were evaporated under reduced pressure and saturated aqueous ammonium chloride (20 mL) was added. The organics were extracted with ethyl acetate (20 mL x 3) and then the combined organic phase was washed with water (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue was purified by preparative thin layer chromatography to afford the title compound as a white solid (15 mg, Yield: 27%). Mp = 75.0-75.7 °C; R f = 0.7 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, DMSO+D 2 0) δ 7.16 (d, J = 8.0 Hz, 1 H), 7.07 (d, J = 8.0 Hz, 1 H), 7.00 (d, J = 2.4 Hz, 1 H), 2.64 (s, 1 H), 2.13 (t, J = 13.6 Hz, 1 H), 2.01 (t, J = 13.6 Hz, 1 H), 1.44-1.38 (m, 1 H), 1.28- 1.05 (m, 9H), 0.94-0.87 (m, 4H), 0.78-0.74 (m, 3H), 0.59-0.55 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 276 (M - H 2 0 + H + ).

Example 46: (±)-(4-ethylphenyl)(2,6,6-trimethy lcyclohex-1 -enyl)methanol To a solution of (4-ethylphenyl) magnesium bromide (0.5 M in tetrahydrofuran, 44 mL, 22.0 mmol) in diethyl ether (20 mL) at 0 °C was added a solution of 2,6,6-trimethylcyclohex-l-enecarbaldehyde (1.67 g, 11.0 mmol) diethyl ether (16 mL) drop wise over 5 minutes under argon. The reaction mixture was stirred for 2 hours at room temperature. The reaction mixture was quenched with saturated aqueous ammonium chloride (10 mL) and water (50 mL) and then extracted with ethyl acetate (2 x 60 mL). The combined organic layer was washed with brine (50 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1 :70) to give the title compound as a colorless oil (2.62 g, Yield: 92%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.33 (d, J = 8.0 Hz, 2H), 7.15 (d, J = 7.6 Hz, 2H), 5.40 (d, J = 4.0 Hz, 1 H), 2.QA (q, J = 7.6 Hz, 2H), 1.99 (t, J = 6.4 Hz, 2H), 1.82 (d, J = 4.0 Hz, 1 H), 1.71 - 1.47 (m, 5H), 1.41 (s, 3H), 1.23 (t, J = 7.6 Hz, 3H), 1.19 (s, 3H), 1.05 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 241 (M - H 2 0 + H + ).

Example 47: (4-ethy Ipheny l)(2,6,6-trimethylcyclohex-1 -enyl)methanone

To a solution of the product of Example 46 (1.07 g, 4.14 mmol) in dichloromethane (20 mL) was added manganese dioxide (3.6 g, 41.4 mmol). The mixture was stirred at room temperature for 42 hours. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1 :100) to afford the title compound as a colorless oil (0.74 g, Yield: 70%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.85 (d, J = 8.4 Hz, 2H), 7.26 (d, J = 8.0 Hz, 2H), 2.70 (q, J = 7.6 Hz, 2H), 2.07 (t, J = 6.4 Hz, 2H), 1.79-1.73 (m, 2H), .55 - 1.52 (m, 2H), 1.43 (s, 3H), 1 .26 (t, J = 7.6 Hz, 3H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 257 (M + H + ). Example 48: (±)-(4aR,9aS)-6-ethyl-1 ,1 ,4a-trimethy l-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one

A solution of the product of Example 47 (700 mg, 2.73 mmol) in methanesulfonic acid (5 mL) was stirred at 50 °C for 4h. Water (30 mL) was added to the reaction mixture and then it was extracted with ethyl acetate (2 x 40 mL). The combined organic phase was washed with brine (40 mL), dried over anhydrous magnesium sulfate, filtered and the volatiles were evaporated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether = 1 :200) to give the title compound as a yellow oil (530 mg, Yield: 76%). 1 H NMR (400 MHz, CDC ) δ 7.59 (d, J = 8.0 Hz, 1 H), 7.26 (s, 1 H), 7.17 (d, J = 8.0 Hz, 1 H), 2.73 (q, J = 7.6 Hz, 2H), 2.17 (s, 1 H), 2.10-2.04 (m, 1 H), 1.68-1.59 (m, 2H), 1.48- 1.41 (m, 1 H), 1.38-1.35 (m, 2H), 1.30-1.26 (m, 6H), 1.22 (s, 3H), 0.65 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 257 (M + H + ).

Example 49: (+)-(4aR,9/?,9aS)-6-ethyl-1 ,1 ,4a-trlmethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 48 (370 mg, 1.44 mmol) in methanol (8 mL) at 0 °C was added sodium borohydride (164 mg, 4.33 mmol) portion wise. The mixture was stirred overnight at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was diluted with water. The reaction mixture was extracted with ethyl acetate (2 x 30 mL) and the combined organic phase was washed with brine (20 mL), dried over anhydrous magnesium sulfate, filtered and then concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent: ethyl acetate/petroleum ether = 1 :125) to afford the title compound as a white solid (205 mg, Yield: 55%). Mp = 75.0-77 °C; R f = 0.4 (20:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI 3 ) δ 7.32 (d, J = 7.6 Hz, 1 H), 7.10 (d, J = 7.6 Hz, 1 H), 6.96 (s, 1 H), 5.01 (t, J = 8.0 Hz, 1 H), 2.66 (q, J = 7.6 Hz, 2H), 1.70-1.30 (m, 7H), 1.49 (s, 3H), 1.24 (t, J = 7.6 Hz, 3H), 1.20 (s, 3H), 1.16 (s, 3H), 1.04-0.99 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 241 (M - H 2 0 + H + ). Example 50: (±)-6-ethyl-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluorene

To a solution of the product of Example 49 (160 mg, 0.619 mmol) in dichloromethane (2 mL) at 0 °C was added thionyl chloride (100 μί). The mixture was stirred for 2 hours at 0 °C. The reaction mixture was diluted with dichloromethane (30 mL) and washed with water ( 5 mL), saturated aqueous sodium bicarbonate (15 mL), then brine (15 mL), dried over anhydrous magnesium sulfate, filtered and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether) to give the title compound as a colorless oil (130 mg, Yield: 87%). R f = 0.9 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.19 (d, J = 8.0 Hz, 2H), 7.07 (s, 1 H), 7.03 (d, = 8.0 Hz, 2H), 6.33 (s, 1 H), 2.67 (q, J = 7.6 Hz, 2H), 2.20-1 .80 (m, 2H), 1.65-1.58 (m, 2H), 1.38 (s, 3H), 1.29 (s, 3H), 1.25 (t, J = 8.0 Hz, 2H), 1.247 (s, 3H), 1.15-0.95 (m, 2H) ppm; Mass spectrum (ESI +ve) m/z 241 (M + H + ).

Example 51 : (±)-6-ethyl-1 ,1 ,4a,9-tetramethy l-2,3,4,4a-tetrahydro-1 H- fluorene

To a solution of the product of Example 48 (128 mg, 0.5 mmol) in tetrahydrofuran (10 mL) at -78 °C was added methyl magnesium iodide (3.0 M, 0.5 mL) and the reaction was stirred at -78 °C for 2 hours after which it was allowed to warmed to room temperature and stirred overnight. The reaction was then heated to 50 °C stirred an additional 24 hours. The reaction was cooled to room temperature and quenched by the addition of saturated aqueous ammonium chloride. The organics were extracted with dichloromethane and then the organic phase was washed with brine, dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a colorless oil (42 mg, Yield: 33 %). R f = 0.68 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.14 (d, J = 7.6 Hz, 1 H), 7.08-7.05 (m, 2H), 2.67 (q, J = 7.6 Hz, 2H), 2.19 (s, 3 H), 2.13-2.08 (m, 1 H), 1.95-1.86 (m, 1 H), 1.64-1.54 (m, 2H), 1.43 (s, 3 H), 1 .33 (s, 3 H),1.28 (s, 3H), 1.24(m, 4 H), 1.1 1 (dt, J = 12.4, 4.4 Hz, 1 H) ppm. 3 C NMR (100 MHz, CDCI 3 ) δ 154.4, 153.1 , 142.8, 140.4, 127.2, 125.5, 119.9, 1 18.0, 50.8, 43.3, 36.6, 35.9, 33.0, 29.0, 26.6, 25.9, 19.3, 16.1 , 12.6. ppm; Mass spectrum (ESI +ve) m/z 255 (M + H + ).

Example 52: (±)-(4a ?,9S,9aS)-6-ethyl-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a- exahydro-1H-fluorene

To a solution of the product of Example 51 (40 mg, 0.16 mmol) in methanol (2 mL) was added Pd/C (8.0 mg) and the mixture was stirred under 1 atmosphere of hydrogen for 2 days. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give the title compound as a colorless oil (36 mg, Yield: 90 %). 1 H NMR (400 MHz, CDCI 3 ) δ 7.08 (d, J = 7.6 Hz, 1 H), 7.00 (dd, J = 7.6, 1.6 Hz, 1 H), 6.96 (s, 1 H), 3.33- 3.29 (m, 1 H), 2.65 (q, J = 7.2 Hz, 2H), 1.91 (d, J = 7.6 Hz, 1 H), 1.78-1.61 (m, 1 H), 1.59-1.49 (m, 4H), 1.42 (s, 3 H), 1.39 (d, J = 7.2 Hz, 3 H), 1.25 (m, 4 H), 1.15 (s, 3H), 1.07(s, 3 H) ppm; 13 C NMR (100 MHz, CDCI 3 ) δ 152.5, 145.5, 142.2, 125.8, 123.2, 121.5, 59.4, 45.2, 41.5, 37.0, 36.4, 33.1 , 32.1 , 28.9, 28.6, 28.2, 21.7, 19.7,15.8. ppm Mass spectrum (ESI +ve) m/z 257 (M + H + ).

Example 53: (±)-(3,4-dimethylphenyl)(2,6,6-trimethylcyclohex-1 - enyl)methanol

To a solution of (3,4-dimethyl)magnesium chloride (17 mL, 8.21 mmol) in tetrahydrofuran (8 mL) was added a solution of 2,6,6-trimethylcyclohex-1- enecarbaldehyde (500 mg, 3.28 mmol) in tetrahydrofuran (4 mL) drop-wise at -78 °C. The mixture was stirred at -78 °C for 2 hours. The reaction mixture was quenched with water (10 mL), extracted with ethyl acetate (3 x 20 mL), and the combined organic phase was washed with water (20 mL), brine (30 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was purified by silica gel column chromatography (petroleum ether/:ethyl acetate = 50:1 ) to give the title compound as a white solid (500 mg, Yield: 59%). Mp = 76.5-77.8 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, DMSO) δ 7.14 (s, 1 H), 7.00 (m, H), 5.20 (d, J = 4.8 Hz, 1 H), 5.08 (d, J = 4.8 Hz, 1 H), 2.18 (s, 3H), 2.16 (s, 3H), 1.91 (d, J = 6.0 Hz, 2H), 1.59-1.57 (m, 2H), 1.46-1.4 (m, 2H), 1.32 (s, 3H), 1.06 (s, 3H), 1.00 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 241 (M - H 2 0 + H + ). Example 54: (3,4-dimethylphenyl)(2,6,6-trimethylcyclohex-1- enyl)methanone

A mixture of the product of Example 53 (150 mg, 0.58 mmol) and manganese dioxide (505 mg, 5.8 mmol) in dichloromethane (6 ml_) was stirred at room temperature for 3 days. The reaction mixture was filtered. The filtrate was concentrated in vacuo and the residue purified by silica gel chromatography (petroleum ether/ethyl acetate 50:1 ) to afford the title compound the as a colorless oil (120 mg, Yield: 80%) R f = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.73 (s, 1 H), 7.63 (d, J = 7.6 Hz, 1 H), 7.19 (d, J = 8.0 Hz, 1 H), 2.31 (s, 6H), 2.07 (t, J = 6.4 Hz, 2H), 1.78- 1.75 (m, 2H), 1.56-1.52 (m, 2H), 1.43 (s, 3H), 1.02 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 257 (M + H + ).

Examples 55a and 55b: (+)-(4a ,9aS)-1 ,1 ,4a,6,7-pentamethyl-2,3,4,4a- tetrahydro-1W-fluoren-9(9aH)-one (55a) and (±)-(4aR,9aS)-1 ,1 ,4a,5,6- pentamethyl-2,3 ,4,4a-tetrahydro-1 H-fluoren-9(9aW)-one (55b)

A stirred solution of the product of Example 54 (120 mg) in methansulfonic acid (2 mL) was stirred at 50 °C for 4 hours. The mixture was added to ethyl acetate (100 mL), washed with saturated aqueous sodium bicarbonate (100 mL x *3) and brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a mixture of the title compounds as a pale yellow solid (71 mg). Purification by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 100:1) gave two compounds: the title compound 55b (20 mg Yield: 17%); = 0.38 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.46 (d, J = 8.0 Hz, 1 H), 7.15 (d, J = 8.0 Hz, 1 H), 2.38 (s, 3H), 2.35 (s, 3H), 2.17-2.1 1 (m, 1 H), 2.10 (s, 1 H), 2.04-1.97 (m, 1 H), 1.75-1.61 (m, 2H), 1.51-1.43 (m, 2H), 1.40 (s, 3H), 1.25 (s, 3H), 0.71 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 257 (M + H + ); and the title compound 55a as a white solid (34 mg, Yield: 28%). Mp = 100-101 °C; R f = 0.4 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 7.44 (s, 1 H), 7.17 (s, 1 H), 2.35 (s, 3H), 2.29 (s, 3H), 2.14 (s, 1 H), 2.10-2.04 (m, 1 H), 1.68-1.60 (m, 2H), 1.49-1.40 (m, 1 H), 1.37-1.33 (m, 2H), 1.24 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 257 (M + H + ).

Example 56: (+)-(4-fluorophenyl)(2,6,6-trimethylcyclohex-1- eny1)methanol

To a stirred solution of (4-fluorophenyl)magnesium bromide (0.8 M in tetrahydrofuran, 12.5 mL, 10.0 mmol) cooled to -78 °C was added a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (609 mg, 4.0 mmol) in tetrahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for additional 2 hours. The mixture was cooled to 0 °C, quenched by slow addition of saturated aqueous ammonium chloride (30 mL) followed by the addition of water (20 mL). The reaction mixture was then extracted with ethyl acetate (50 mL x 4) and the combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by column chromatography (eluent: petroleum ether/ethyl acetate = 100:1->50:1) afforded the title compound as a colorless oil (882 rhg; Yield: 89%); R f = 0.3 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.39-7.36 (m, 2H), 7.01 - 6.97 (m, 2H), 5.37 (d, J = 4.8 Hz, 1 H), 1.98 (t, J = 6.2 Hz, 2H), .82 (d, J = 5.2 Hz, 1 H), 1.66-1.61 (m, 2H), 1.56-1.51 (m, 2H), 1.38 (s, 3H), 1.19 (s, 3H), 1.05 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ).

Example 57: (4-fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanone

To a stirred solution of the product of Example 56 (320 mg, 1.29 mmol) in dichloromethane (10 mL) was added manganese dioxide (871 mg, 10.00 mmol). The reaction mixture was stirred at room temperature for 60 hours.

The mixture was filtered and concentrated under reduced pressure.

Purification of the residue by column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) gave the title compound as a colorless oil (306 mg; Yield: 96%). R f = 0.8 (10:1 petroleum ether/ethyl acetate); H NMR (400

MHz, CDCI 3 ) δ 7.97-7.94 (m, 2H), 7.14-7.09 (m, 2H), 2.07 (t, J = 6.6 Hz, 2H),

1.80-1.74 (m, 2H), 1.56-1.53 (m, 2H), 1 .43 (s, 3H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 58: (±)-(4a 9aS)-6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro- 1 H-fluoren-9(9aH)-one

A stirred solution of the product of Example 57 (282 mg, 1.15 mmol) in methansulfonic acid (4 mL) was stirred at 65 °C for 3 hours. To the mixture was added ethyl acetate (100 mL) and the organic phase was washed with saturated aqueous sodium bicarbonate (100 mL x 3), then brine (30 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by column chromatography (eluent: petroleum ether/ethyl acetate = 200:1 ) and then preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) afforded the title compound as a pale yellow solid (50 mg, Yield: 18%). Mp = 76-78 °C; R f = 0.4 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.69-7.66 (m, 1 H), 7.08- 7.00 (m, 2H), 2.20 (s, 1 H), 2.04-2.02 (m, 1 H), 1.69-1.60 (m, 2H), 1 .42-1.36 (m, 3H), 1.27 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ).

Example 59: (±)-(4aR,9R,9aS)-6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 58 (32 mg, 0.13 mmol) in methanol (2 mL) was added sodium borohydride (42 mg, 1 .1 mmol). The reaction was stirred at room temperature for 46 hours. Water (20 mL) was added and the mixture was extracted with ethyl acetate (15 mL x 4). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1->100:1) gave material (16 mg) which was further purified by column chromatography (eluent: petroleum ether/ethyl acetate = 10:1) to give the title compound as a white solid (12 mg, Yield: 37%). Mp = 91-94 °C; R, = 0.2 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 O) δ 7.32 (dd, J = 8.0, 5.6 Hz, 1 H), 6.93-.688 (m, 1 H), 6.81-6.78 (m, 1 H), 4.98 (d, J = 8.8 Hz, 1 H), 1.68 (d, J = 8.8 Hz, 1 H), 1.65-1.54 (m, 2H), 1.46 (s, 3H), 1.42-1.35 (m, 3H), 1.18 (s, 3H), 1.15 (s, 3H), 1.02-0.94 (m, H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ). Example 60: (±)-(4aR,9 ?,9aS)-6-chloro-1 ,1 ,4a-trimethyl-2,3 ,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To the product of Example 41 (50 mg, 0.21 mmol) in tetrahydrofuran at -78 °C under argon was added 1 M borane in tetrahydrofuran (1.7 mL, 1.7 mmol) and the reaction mixture was stirred at for 10 minutes after which time it was allowed to warm to room temperature and stirred for an additional 20 hours. The reaction was cooled to 0 °C, ethanol was added to quench the reaction, and to the reaction mixture was added a solution of aqueous sodium hydroxide (136.2 mg in 2.4 mL water) and then 30% hydrogen peroxide (1.26 mL) were added with stirring. After stirring for 2 hours at room temperature, the mixture was poured into saturated aqueous ammonium chloride and then extracted with diethyl ether (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a white solid (30 mg, Yield: 54%). Mp = 107 °C; H NMR (400 MHz, CDCI 3 +D 2 O) δ 7.30 (d, J = 8.0 Hz, 1 H), 7.19 (dd, J = 8.0, 2.0 Hz, 1 H), 7.09 (d, J = 1.6 Hz, 1 H), 4.98 (d, J = 8.8 Hz, 1 H), 1.67-1.54 (m, 3H), 1.46-1.34 (m, 6H), 1.18 (s, 3H), 1.15 (s, 3H), 1.02-0.96 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M - H 2 0 + H + ).

Example 61 : (±)-(4aft,9aS)-6-chloro-1 ,1 ,4a-trimethy l-2,3,4,4a-tetrahydro- 1W-fluoren-9(9aW)-one

To a mixture of the product of Example 60 (50 mg, 0.19 mmol) and sodium bicarbonate (16 mg, 0.38 mmol) in dichloromethane (5 mL) at 0 °C was added Dess-Martin periodinane (160.2 mg, 0.38 mmol). The reaction was stirred at 0 °C for 30 minutes after which time it was stirred at room temperature for 4 hours. Hydrochloric acid (2 mL, 5%) was added to quench the reaction which was then extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), then dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel chromatography to afford the title compound as a white solid (30 mg, Yield: 60%). Mp = 1 1 1-1 12 °C; 1 H NMR (400 MHz, CDCI 3 )□ δ 7.60 (d, J = 8.0 Hz, 1 H), 7.39 (d, J = 2.0 Hz, 1 H), 7.31 (d, J = 8.0, 2.0 Hz, 1 H), 2.19 (s, 1 H), 2.12-2.05 (m, 1 H), 1.68-1.61 (m, 2H), 1.38-1.36 (m, 3H), 1.25 (s, 3H), 1.20 (s, 3H), 0.62 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 263 (M + H + ).

Example 62: (±)-(3-chloro-4-methylphenyl)(2,6,6-trimethylcyclohex-1 - enyl)methanol

To a stirred solution of (3-chloro-4-methylphenyl)magnesium bromide

(0.5 M in tetrahydrofuran, 20 mL, 10.0 mmol) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (609 mg, 4.0 mmol) in dry tetrahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for additional 2 hours. The mixture was cooled to 0 °C, and saturated aqueous ammonium chloride (30 mL) was added followed by the addition of water (20 mL). The reaction mixture was extracted with ethyl acetate (50 mL x 4) and the combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) afforded the title compound as a white solid (595 mg, Yield: 53%). Mp = 83-84 °C; R f = 0.4 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ7.42 (s, 1 H), 7.19-7.14 (m, 2H), 5.34 (d, J = 5.2 Hz, 1 H), 2.36 (s, 3H), 1.99 (t, J = 6.2 Hz, 2H), 1.80 (d, J = 5.2 Hz, 1 H), 1.68-1.62 (m, 2H), 1.55-1.52 (m, 2H), 1.39 (s, 3H), 1.18 (s, 3H), 1.06 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 261 (M - H 2 0 + H + ).

Example 63: (3-chloro-4-methylphenyl)(2,6,6-trimethylcyclohex-1 -enyl) methanone

To a stirred solution of the product of Example 62 (275mg, 0.98mmol) in dichloromethane (15 mL) was added manganese dioxide (852mg, 9.8mmol). The reaction mixture was stirred at room temperature for 60 hours. The reaction mixture was filtered and then concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1 ) afforded the title compound as a pale yellow oil (188.4mg, Yield: 68.5%). R f = 0.4 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.90 (d, J = 1.2 Hz, 1 H), 7.71 (dd, J = 8.0, 1.2 Hz, 1 H), 7.31 (d, J = 8.0 Hz, 1 H), 2.43 (s, 3H), 2.08 (t, J = 6.4 Hz, 2H), 1.80-1.74 (m, 2H), 1.56-1.54 (m, 2H), 1.44 (s, 3H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 277 (M + H + ).

Examples 64a and 64b: (±)-(4aR,9aS)-5-chloro-1 ,1 ,4a,6-tetramethyl- 2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)-one (64a) and (+)-(4aK,9aS)-7- chloro-1 ,1 ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aW)-one (64b)

The product of Example 63 (162 mg, 0.59 mmol) was added to methanesulfonic acid (2 mL) and the mixture was stirred at 55 °C for 3 hours. The reaction mixture was added to ethyl acetate (50 mL) and the organic phase was washed with water (30 mL), then brine (30 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 500:1 ) and then preparative thin layer chromatography twice (petroleum ether/ethyl acetate = 50:1) to afford the title compound 64a as a pale yellow solid (32 mg; Yield: 20%). R f = 0.38 (50:1 petroleum ether/ethyl acetate = 50/1 ); 1 H NMR (400 MHz, CDCI 3 ) δ 7.64 (s, 1 H), 7.28 (s, 1 H), 2.47 (s, 3H), 2.18 (s, 1 H), 2.15-2.05 (m, 1 H), 1.70-1.56 (m, 5H), 1.49-1.32 (m, 2H), 1.30 (s, 3H), 1.26 (s, 3H), 0.65 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 277 (M + H + ). As well as the title compound 64b as a yellow solid ( 2 mg, Yield: 7%). Mp = 85-87 °C; R f = 0.7 (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.63 (s, 1 H), 7.27 (s, 1 H), 2.46 (s, 3H), 2.17 (s, 1 H), 2.09-2.05 (m, 1 H), 1.68-1.58 (m, 2H), 1.45-1.33 (m, 3H), 1.25 (s, 3H), 1.21 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 277 (M + H + ). Example 65: (±)-(4aft,9 ?,9aS)-7-chloro-1 ,1 ,4a,6-tetramethyl-2,3 ,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a stirred solution of the product of Example 64a (27 mg, 0.10 mmol) in methanol (2 mL) was added sodium borohydride (31 mg, 0.80 mmol). The reaction was stirred at room temperature for 4 hours. The reaction was quenched by the addition of water (20 mL) and the mixture was extracted with ethyl acetate (15 mL x 4). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 50:1 ) afforded the title compound as a white solid (18 mg, Yield: 65%). Mp = 1 15-1 17 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 7.34 (s, 1 H), 6.96 (s, 1 H), 4.97 (t, J = 8.2 Hz, 1 H), 2.36 (s, 3H), 1.70 (d, J = 8.4 Hz, 1 H), 1.64-1.59 (m, 2H), 1.58-1.52 (m, 1 H), 1.45 (s, 3H), 1.44-1.34 (m, 3H), 1 .17 (s, 3H), 1 .14 (s, 3H), 0.96 (dt, J = 13.5, 3.5 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 261 (M - H 2 0 + H + ).

Example 66: (±)-(4afl,9fl,9aS)-5-chloro-1 ,1 ,4a,6-tetramethyl-2,3,4,4a,9,9a hexahydro-1 W-fluoren-9-ol

To a stirred solution of the product of Example 64b (39 mg 0.14 mmol) in methanol (2 mL) was added sodium borohydride (42 mg, 1.1 1 mmol). The reaction was stirred at room temperature for 60 hours. Water (20 mL) was added to quench the reaction and the mixture was extracted with ethyl acetate (15 mL x 4). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 00:1 -> 50:1) gave a colorless oil (39 mg) which was further purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) to afford the title compound as a colorless oil (24 mg, Yield: 61 %). R f = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.18 (d, J = 7.2 Hz, 1 H), 7.12 (d, J = 7.2 Hz, 1 H), 4.88 (t, J = 7.8 Hz, 1 H), 2.36 (s, 3H), 2.18 (d, J = 14.0 Hz, 1 H), 1.75 (br, H), 1.74 (s, 3H), 1.67 (d, J = 8.8 Hz, 1 H), 1.65-1.58 (m, 1 H), 1.45- 1.32 (m, 3H), 1.20 (s, 3H), 1.17 (s, 3H), 1.09 (dt, J = 13.2, 3.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 261 (M - H 2 0 + H + ).

Example 67: (±)-(3-fluoro-4-methoxyphenyl)(2,6,6-trimethylcyclohex-1 - enyl)methanol

To a stirred solution of (3-fluoro-4-methoxyphenyl)magnesium bromide (0.5 M in tetrahydrofuran, 20 mL, 10.0 mmol) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-l-enecarbaldehyde (609 mg, 4.0 mmol) in dry tetrahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for additional 2 hours. The mixture was cooled down to 0 °C and saturated aqueous ammonium chloride (30 mL) was added slowly followed by addition of water (20 mL). The mixture was then extracted with ethyl acetate (50 mL x 4) and the combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1 :0 -> 25:1 ) gave the title compound as a colorless oil (786 mg, Yield: 70%). = 0.3 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.19-7.14 (m, 2H), 6.94 (t, J = 8.6 Hz, H), 5.36 (d, J = 4.4 Hz, 1 H), 3.91 (s, 3H), 2.01 (t, J = 6.4 Hz, 2H), 1.85 (d, J = 5.6 Hz, 1 H), 1.70-1.64 (m, 2H), 1.58-1.54 (m, 2H), 1.43 (s, 3H), 1.21 (s, 3H), 1.08 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 261 (M - H 2 0 + H + ).

Example 68: (±)-(3-fluoro-4-methoxyphenyl)(2,6,6-trimethylcyclohex-1- enyl) methanone

To a stirred solution of the product of Example 67 (278 mg, 1.0 mmol) in dichloromethane (10 mL) was added manganese dioxide (869 mg, 10.0 mmol). The mixture was stirred overnight. The reaction mixture was filtered and the black solid was washed with dichloromethane (5 mL x 3). The combined filtrate was concentrated in vacuo and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 10:1) to afford the title compound as a colorless oil (196 mg, Yield: 71 %). R, = 0.4 (25:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) 7.76-7.68 (m, 2H), 7.02 (t, J = 8.4 Hz, 1 H), 3.99 (s, 3H), 2.10 (t, J = 6.8 Hz, 2H), 1.81-1.76 (m, 2H), 1.58-1.56 (m, 2H), 1.47 (s, 3H), 1.06 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 277 (M + H + ). Example 69: (±)-(4aR,9aS)-7-fluoro-6-methoxy-1,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1 H-fluoren-9(9aH)-one

A solution of the product of Example 68 (1 17 mg, 0.42 mmol) in methansulfonic acid (2 mL) was stirred at 55 °C for 3 hours. The mixture was diluted with ethyl acetate (50 mL), washed with water (30 mL) and brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 00:1) gave the title compound as a colorless oil (91 mg, Yield: 78%). R f = 0.38 (25 :1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.36 (d, J = 9.6 Hz, 1 H), 6.91 (d, J = 7.2 Hz, 1 H), 4.01 (s, 3H), 2.17 (s, 1 H), 2.07-2.00 (m, 1 H), 1.74-1.62 (m, 2H), 1.54-1.44 (m, 1 H), 1.41-1.37 (m, 2H), 1.30 (s, 3H), 1.24 (s, 3H), 0.70 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 277 (M + H + ).

Example 70: (±)-(4aR,9R,9aS)-7-fluoro-6-methoxy-1 ,1 ,4a-trimethyl- 2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9-ol

To a stirred solution of the product of Example 69 (68 mg, 0.25 mmol) in methanol (2 mL) was added sodium borohydride (38 mg, 1.0 mmol). The reaction was stirred at room temperature for 3 hours after which time additional sodium borohydride (38 mg, 1.0 mmol) was added. The reaction was stirred overnight. Water (20 mL) was added to quench the reaction and the mixture was extracted with ethyl acetate (15 mL x 4). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 10:1) afforded the title compound as a white solid (59 mg, Yield: 86%). Mp = 100-101 °C; R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3+D2O) δ 7.08 (d, J = 10.8 Hz, 1 H), 6.99 (d, J - 7.6 Hz, 1 H), 4.95 (d, J = 8.0 Hz, 1 H), 3.89 (s, 3H), 1.65-1.54 (m, 3H), 1.46 (s, 3H), 1.44-1.30 (m, 3H), 1.18 (s, 3H), 1.15 (s, 3H), 1.01-0.93 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 261 (M - H 2 0 + H + ).

Example 71 : (±)-(thiophen-2-yl(2,6,6-trimethylcyclohex-1 -enyl)methanol To a solution of thiophen-2-ylmagnesium bromide (2M in tetrahydrofuran, 6.6 mL, 13.13 mmol) in tetrahydrofuran (7 mL) at -78 °C was added a tetrahydrofuran solution (10 mL) of 2,6,6-trimethyicyclohex-1- enecarbaldehyde (1 g, 6.57 mmol) drop wise over 5 minutes. The reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction was quenched by pouring into aqueous saturated ammonium chloride (10 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (3 x 20 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel chromatography to afford the title compound as yellow oil (0.92 g, Yield: 59%). = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.21 (dd, J = 6.0, 1.2 Hz, 1 H), 6.95-6.92 (m, 1 H), 6.84 (dd, J = 6.8, 1.2 Hz, 1 H), 5.55 (d, J = 4.8 Hz, 1 H), 2.10 (d, J = 5.2 Hz, 1 H), 2.00 (t, J = 6.4 Hz, 2H), 1.67 - 1.61 (m, 2H), 1.57 (s, 3H), 1.54 - 1.48 (m, 2H), 1.18 (s, 3H), 1.06 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 72: thiophen-2-yl(2,6,6-trimethylcyclohex-1 -enyl)methanone

To a solution of the product of Example 71 (490 mg, 2.07 mmol) in dichloromethane (25 mL) was added manganese dioxide (1.8 g, 20.73 mmol). The reaction mixture was stirred at room temperature for 48 hours. The mixture was filtered and the filtrate was concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography to afford the title compound as a white solid (330 mg, Yield: 68%). Mp = 122-124 °C; R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.64 (dd, J = 4.8, 1.2 Hz, 1 H), 7.56 (dd, J = 4.0, 1.2 Hz, 1 H), 7.10 (dd, J = 4.8, 3.6 Hz, 1 H), 2.06 (t, J = 6.4 Hz, 2H), 1.77-1 .73 (m, 2H), 1.56 - 1.51 (m, 5H), 1.08 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ). Example 73: (±)-(3b ?,7aS)-3b,7,7-trimethyl-5,6,7,7a-tetrahydro-3bH- indeno[2,1 -b]thiophen-8(4H)-one

To the product of Example 72 (100 mg, 0.427 mmol) was added methanesulfonic acid (1.5 mL) and the mixture was stirred at 50 °C for 2 hours. Water (10 mL) was added to the reaction mixture and it was extracted with ethyl acetate (3 x 10 mL). The combined organic phase was washed with brine (5 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a white solid (75 mg, Yield: 75%). Mp = 67-69 °C; R f = 0.4 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.82 (d, J = 4.8 Hz, 1 H), 6.98 (d, J = 5.2 Hz, 1 H), 2.41 (s, 1 H), 1 .93 - 1 .84 (m, 1 H), 1.72-1.68 (m, 2H), 1.58 - 1.50 (m, 1 H), 1.43 - 1.37 (m, 2H), 1.37 (s, 3H), 1.26 (s, 3H), 0.90 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ). Example 74: (±)-(3bR,7aS,8R)-3b,7,7-trimethyl-4,5,6,7,7a,8-hexahydro- 3bH-indeno[2,1 -b]thiophen-8-ol

To a solution of the product of Example 73 (55 mg, 0.24 mmol) in methanol (2 mL) and tetrahydrofuran (1 mL) at 0 °C was added sodium borohydride (71 mg, 1.88 mmol) portion wise. The mixture was warmed to room temperature stirred for approximately 48 hours. The reaction mixture was concentrated in vacuo and the residue was partitioned between water and ethyl acetate (20 mL). The aqueous phase was extracted with additional ethyl acetate (20 mL) and the combined organic phase was washed with brine (10 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid (10 mg, Yield: 18%). Mp = 109-1 1 1 °C; R f = 0.2 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.24 (d, J = 4.8 Hz, 1 H), 6.75 (d, J = 4.8 Hz, 2H), 5.14 (t, J = 8.0 Hz, 1 H), 1.99 (d, J = 8.0 Hz, 1 H), 1.77 (d, J = 8.0 Hz, 1 H), 1.69-1.55 (m, 2H), 1.46 (s, 3H), 1.43 - 1.34 (m, 3H), 1.16 (s, 3H), 1.12 (s, 3H), 1.07 - 0.99 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ). Example 75: (±)-thiophen-3-yl(2,6,6-trimethylcyclohex-1 -enyl)methanol

3-Bromothiophene (0.978 g, 6.0 mmol) was dissolved in dry hexane (9 mL). n-Butyl lithium (3.75 mL, 6.0 mmol) was added slowly at -40 °C. Tetrahydrofuran (0.9 mL) was then slowly added to this mixture at -40 °C, during which time a white solid precipitated. Then more hexane (5 mL) was added. A solution of 2,6,6-trimethylcyclohex-l-enecarbaldehyde (609 mg, 4.0 mmol) in dry hexane (3 mL) was added slowly at this temperature and the reaction was then allowed to warm gradually to room temperature and stirred for 1 hour. Saturated aqueous ammonium chloride (30 mL) was added and the mixture was extracted with ethyl acetate (20 mL x 4). The combined organic phase was washed with brine (40 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 400:1) gave the title compound as a light yellow solid (659 mg, Yield: 70%). Mp = 47-49 °C; = 0.25 in (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 7.26-7.24 (m, 1H), 7.11-7.10 (m, 1H), 7.03-7.01 (m, 1H), 5.38 (d, J = 5.2 Hz, 1 H), 1.97 (t, J = 6.0 Hz, 2H), 1.88 (d, J = 5.6 Hz, 1 H), 1 .66-1.60 (m, 2H), 1.53-1.50 (m, 2H), 1.48 (s, 3H), 1.18 (s, 3H), 1.05 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 76: thiophen-3-yl(2,6,6-trimethylcyclohex-1 -eny l)methanone

To a solution of the product of Example 75 (200 mg, 0.85 mmol) in dichloromethane (5 mL) cooled to 0 °C was added Dess-Martin periodinane (539 mg, 1.27 mmol) in one portion. The reaction was stirred at room temperature for 1 hour. The mixture was filtered and the solid was washed with dichloromethane (10 mL x 3). The combined organic phase was concentrated under reduced pressure and the residue purified by flash column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to obtain the title compound as a white solid (194 mg, Yield: 98%). Mp = 146- 148 °C; R f = 0.6 in (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7. 94 (d, J = 2.4 Hz, 1 H), 7.51 (d, J = 4.4 Hz, 1 H), 7.30-7.28 (m, 1 H), 2.05 (t, J = 6.4 Hz, 2H), 1.78-1.72 (m, 2H), 1.54-1.51 (m, 2H), 1.49 (s, 3H), 1.05 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ). Example 77: (±)-3b,7,7-trimethyl-4,5,6,7-tetrahydro-3bH-indeno[2,1 - 6]thiophene

To a solution of the product of Example 71 (100 mg, 0.423 mmol) in dichloromethane (6 mL) at 0 °C under argon was added stannic chloride (165 mg, 0.64 mmol). The mixture was allowed to warm to room temperature and was stirred for 2 hours. Water (10 mL) was added to the reaction mixture and then it was extracted with diethyl ether (3 x 0 mL). The combined organic phase was washed with brine (5 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel chromatography to afford the title compound as a pale yellow solid (73 mg, Yield: 79%,). Mp = 51-53 °C; R f = 0.9 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.08 (d, J = 4.8 Hz, 1 H), 6.94 (d, J = 4.8 Hz, 1 H), 6.34 (s, 1 H), 2.17-2.11 (m, 1 H), 1.97 - 1.85 (m, 1 H), 1.69 - 1.58 (m, 2H), 1 .35 (s, 3H), 1.28 (s, 3H), 1.23 (s, 3H), 1.16 - 1.08 (m, 1 H), 1.02 (td, J = 13.2, 3.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M + H + ).

Example 78: (±)-(3b ?,7aS,8/?)-3b,7,7-trimethyl-4,5,6,7,7a,8-hexahydro- 3bH-indeno[2,1 -b]thiophen-8-ol

To 1 M borane in tetrahydrofuran (2.23 mL, 2.23 mmol) at -78 °C under argon was added the product of Example 77 (60 mg, 0.28 mmol) and the reaction stirred at that temperature for 10 minutes after which time it was allowed to warm to room temperature and stirring was continued for 20 hours. The mixture was cooled to 0 °C and then 95% ethanol was added to quench the reaction and then a solution of sodium hydroxide (180 mg) in 3.2 mL water and the 1.68 mL of 30% hydrogen peroxide were added slowly with stirring. The reaction mixture was warmed to room temperature and stirring was continued for 2 hours. The reaction mixture was poured into saturated aqueous ammonium chloride, and then extracted with diethyl ether (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), then brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid (35 mg, Yield: 53%). Mp = 1 15-117 X; R, = 0.2 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.25 (del, J = 4.8, 0.8 Hz, 1 H), 6.75 (d, J = 4.8 Hz, 1 H), 5.14 (t, 1 H), 1.98 (d, J - 7.6 Hz, 1 H), 1.77 (d, J = 6.8 Hz, 1 H), 1.69-1.58 (m, 2H), 1.46 (s, 3H), 1.43 - 1.33 (m, 3H), 1.16 (s, 3H), 1.12 (s, 3H), 1.06-1.02 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 79: (±)-(4aS,8a ?)-5,5,8a-trimethyl-4a,5,6,7,8,8a-hexahydro-4H- indeno[1 ,2-b]thiophen-4-one

A solution of the product of Example 76 (148 mg, 0.63 mmol) in methansulfonic acid (4 mL) was stirred at 50 °C for 3 hours. The mixture was added to ethyl acetate (100 mL), washed with saturated aqueous sodium bicarbonate (100 mL * 3), brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 25:1 ) afforded the title compound as a pale yellow oil (128 mg, Yield: 86%). R f = 0.4 (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.24 (d, J = 5.2 Hz, 1 H), 7.07 (d, J = 5.2 Hz, 1 H), 2.40 (s, 1 H), 1.95-1.90 (m, 1 H), 1.81-1.74 (m, 1 H), 1.71-1.67 (m, 1 H), 1.63-1.57 (m, 1 H), 1.42 (s, 3H), 1.41-1.37 (m, 2H), 1.25 (s, 3H), 0.88 (s, 3H) ) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ).

Examples 80a and 80b: (±)-(4R,4aS,8aR)-5,5,8a-trimethyl-4a,5,6,7,8,8a- exahydro-4H-indeno[1,2-b]thiophen-4-ol (80a) and (+)-(4S,4aS,8aR)- 4a,5,5,8a-tetramethyl-4a,5,6,7,8,8a-hexahydro-4A/-indeno[1,2 -b]thiophen- 4-ol (80b)

A stirred solution of the product of Example 79 (62 mg, 0.26 mmol) in anhydrous tetrahydrofuran (3 mL) cooled to 0 °C was added lithium aluminum hydride (30 mg, 0.79 mmol). The reaction was stirred at room temperature for 4 hours. Water (0.12 mL) and 15% aqueous sodium hydroxide (0.03 mL) was added and the mixture was further diluted with water (30 mL) and then extracted with ethyl acetate (15 mL x 4). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 25:1) afforded the title compound (80a) as a white solid (50 mg, Yield: 81%). Mp = 99-101 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.14 (d, J = 5.2 Hz, 1 H), 6.91 (d, J = 4.8 Hz, 1 H), 4.98 (t, J = 7.6 Hz, 1 H), 1.99 (d, J = 7.2 Hz, 1 H), 1.74-1.70 (m, 1 H), 1.65 (d, J = 8.0 Hz, 1 H), 1.63-1.59 (m, 1 H), 1 .51 (s, 3H), 1.45-1.35 (m, 3H), 1.17 (s, 3H), 1.13-1.09 (m, 1 H), 1.12 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ) and the title compound (80b) as a white solid (4.3 mg, Yield: 7%). Mp = 81-83 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.17 (d, J = 4.8 Hz, 1 H), 6.92 (d, J = 4.8 Hz, 1 H), 5.00 (d, J = 5.2 Hz, 1 H), 1.96 (d, J = 4.8 Hz, 1 H), 1.74-1.70 (m, 1 H), 1.63-1.58 (m, 1 H), 1.51 (s, 3H), 1.45-1.35 (m, 3H), 1.16 (s, 3H), 1.11 (s, 3H), 1.12-1.08 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 81 : furan-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanone

Example 81a: furan-3-yl(2,6,6-trimethy lcyclohex-1 -eny l)methanol

To a solution of 3-bromo-furan (661 mg, 4.5 mmol) dissolved in tetrahydrofuran (20 mL) at -78 °C was added n-butyl lithium (2.5 M, 1.8 mL) and the reaction was stirred for 2 hours. To the mixture was added 2,6,6- trimethylcyclohex-1-enecarbaldehyde (456 mg, 3.0 mmol), the mixture was stirred for an additional 2 hours at -78 °C and then allowed to warm to room temperature. The reaction was quenched by the addition of saturated ammonium chloride and then extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford furan-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanol furan-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanol as a colorless oil (545 mg, Yield: 83% yield). 1 H NMR (400 MHz, CDCI 3 ) δ 7.36-7.35 (m, 1 H), 7.26-7.24 (m, 1 H), 6.36 (s, 1 H), 5.31 (s ,1 H), 1.99-1.97 (m, 2H), 1.81 (s, 1 H), 1.61 (s, 3H), 1.61-1.59 (m, 2 H), 1.46-1.42 (m, 2 H), 1.16 (s, 3H), 0.99 (s, 3H). Example 81 : furan-3-yl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 81a (220.0 mg, 1.0 mmol) in dichloromethane (3.0 ml_) cooled to 0 °C was added sodium bicarbonate (84 mg, 1.0 mmol) and Dess-Martin periodinane (848.0 mg, 2.0 mmol). The mixture was stirred for 1 hour, then warmed to room temperature and stirred for an additional 2 hours. The reaction mixture was quenched with the addition of saturated aqueous sodium bicarbonate (20 ml_) and then extracted with dichloromethane, dried over anhydrous magnesium sulfate, filtered, then purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate from 50:1 -> 40:1) to afford the title compound as a yellow solid (170.2 mg, Yield: 77%). Mp = 122-123 °C; R, = 0.6 (20:1 petroleum ether/ethyl acetate; H NMR (400 MHz, CDCI 3 ) δ 7.82-7.80 (m, 1 H), 7.42-7.40 (m, 1 H), 6.76-6.74 (m, 1 H), 2.02 (t, J = 6.0 Hz, 2H), 1.75-1.68 (m, 2H), 1.51 (s, 3 H), 1.51-1.47 (m, 2H), 1.05 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M + H + ).

Example 82: (ti^aS.ea^-S^.ea-trimethyMa.S.e .e.ea-hexah dro^W- indeno[1 ,2-6]furan-4-one

The product of Example 81 (5 0 mg, 0.23 mmol) was dissolved in methansulfonic acid (1 ml_). The mixture was stirred at room temperature for 1 hour and then at 50 °C for 3 hours. After cooling to room temperature, water (6.0 ml_) was added and the organics were extracted with ethyl acetate. The combined the organic phase was dried over anhydrous magnesium sulfate and filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 ->to 40:1) to give the title compound as a colorless oil (54.0 mg, Yield 49%). R f = 0.4 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 7.44 (d, J = 2.0 Hz, 1H), 6.44 (d, J = 2.0 Hz, 1 H), 2.43 (s, H), 1.89- 1.55 (m, 4H), 1.40-1.42 (m, 2 H), 1.41 (s, 3H), 1.27 (s, 3 H), 0.96 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M + H + ). Example 83: (±)-(4R,4aS,8aR)-5,5,8a-trimethyl-4a,5,6,7,8,8a-hexahydro- 4H-indeno[1 ,2-/>]furan-4-ol

To a solution of the product of Example 82 (33.9 mg, 0.16 mmol) in methanol at 0 °C was added sodium borohydride (17.6 mg, 0.47 mmol). The reaction was stirred at room temperature for 4 hours and then the solution was stirred at 50 °C for 2 hours. The reaction was quenched by the addition of water (2 ml_) and then the volatiles were evaporated under reduced pressure, was The residue was extracted with ethyl acetate and the organic phase was dried over anhydrous magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 20:1 - >15:1) to afford the title compound (10.2 mg, Yield 29%). R, = 0.3 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) 7.28 (d, J = 2.0 Hz, 1 H), 6.33 (d, J = 2.0 Hz, 1H), 4.84 (m, 1 H), 1.97 (d, J - 6.4 Hz, 1 H), 1.74-1.75 (m, 1H), 1.58-1.62 (m, 2 H), 1.46 (s, 3H), 1.25-1.38 (m, 3 H) , 1.15 (s, 3 H) , 1.07 (s, 3 H), 1.07-1.02 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 203 (M - H 2 0 + H + ).

Example 84: furan-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanone Example 84a: furan-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanol

Furan ( 0.5 ml_, 6.0 mmol) and tetramethylethylenediamine (0.93 mL, 6.2 mmol) dissolved in tetrahydrofuran (20 mL) and cooled to -78 °C. n-Butyl lithium (2.5 M, 2.5 mL) was added and the reaction was stirred 0.5 hour and then 2,6,6-trimethylcyclohex-1-enecarbaldehyde (0.5 mL, 6.0 mmol) was added. The mixture was stirred for 0.5 hour at -78 °C and then allowed to warmed to room temperature. The reaction was stirred for 1 hour and then the reaction mixture was added to saturated ammonium chloride. The organics were extracted with dichloromethane, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the residue purified by silica gel column chromatography to give the title compound as a light yellow oil (527 mg, Yield: 80% yield). 1 H NMR (400 MHz, CDCI 3 ) δ 7.37 (s, 1 H), 6.30 (t, J = 1.6 Hz, 1 H), 6.11 (d, J = 3.2 Hz, 1 H ), 5.38 (d, J = 3.6 Hz, 1 H), 2.10 (m, 1 H), 2.00 (t, J = 6.4 Hz, 2 H), 1.68 (s, 3 H), 1.64-1.53 (m, 2 H), 1.51-1.46 (m, 2 H), 1.15 (s, 3 H), 0.95 (s, 3H).

Example 84: furan-2-yl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a slurry of manganese oxide (1.04 g, 12.0 mmol) in dichloromethane

(10.0 mL) was added the product of Example 84a (220 mg, 1.0 mmol) and the mixture was stirred overnight. The reaction mixture was filtered, and then concentrated under reduced pressure and the residue was purified by silica gel column chromatography to afford the title compound as a white solid (120 mg, Yield: 55% yield). Mp = 94-95 °C; R f = 0.7 (10: 1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.60 (s, 1 H), 7.04(d, J = 3.2 Hz, 1 H), 6.51 (dd, J = 3.6, 1.6 Hz, 1 H), 2.03 (t, J = 6.4 Hz, 2 H), 1.74-1.71 (m, 2 H ), 1.53-1.50 (m, 5 H), 1.06 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M + H + ).

Example 85: pyridin-3-yl(2,6,6-trimethylcyclo ex-1-enyl)methanol

To a solution of pyridin-3-ylmagnesium bromide (0.25 M in 2- methyltetrahydrofuran) (20 mL, 5.0 mmol) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (305 mg, 2.0 mmol) in tetrahydrofuran (5 mL). Then the mixture was warmed up to room temperature and stirred for 2 hours. The reaction was quenched with saturated aqueous ammonium chloride and the mixture was extracted with ethyl acetate (100 mL). The organic layer was washed with brine (50 mL x χ 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound a white solid (341 mg, Yield: 74%); Mp = 129.6- 130.7 °C; Rf = 0.2 (2:1 petroleum ether/ethyl acetate); 1 H NMR (400MHz, CDCI 3 ) δ 8.62 (t, J = 1.2 Hz, 1 H), 8.46 (d, J = 4.4 Hz, 1 H), 7.78 (d, J = 8.8 Hz, 1 H), 7.26-7.23 (m, 1 H), 5.43 (d, J = 4.0 Hz, 1H), 1.98 (t, J = 6.4 Hz, 2H), 1.92 (d, J = 4.4 Hz, 1 H), 1.66-1.63 (m, 2H), 1 .57-1.52 (m, 2H), 1.37 (s, 3H), 1.20 (s, 3H), 1.08 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 232 (M + H + ). Example 86: pyridin-3-yl(2,6,6-trimethylcyclohex-1 -eny l)methanone

A stirred solution of the product of Example 85 (274 mg, 1.18 mmol) in dichloromethane (6 mL) cooled to 0 °C was added Dess-Martin periodinane (904 mg, 2.13 mmol). The reaction was stirred at room temperature for 1 hour. Dichloromethane (30 mL) was added, the mixture was washed with saturated sodium bicarbonate (30 mL x 3) and brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 5:1) gave the title compound as a yellow oil (206 mg, Yield: 76%); R f = 0.7 (2:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 9.08 (dd, J = 2.4, 0.8 Hz, 1 H), 8.75 (dd, J = 4.8, 2.0 Hz, 1 H), 8.23 (dt, J = 8.0, 2.0 Hz, 1 H), 7.44-7.40 (m, 1 H), 2.09 (dd, J = 6.8, 6.0 Hz, 2H), 1.80-1.74 (m, 2H), 1.58-1.55 (m, 2H), 1.45 (s, 3H), 1.04 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 230 (M + H + ).

Example 87: 1 , ,4a,6-tetramethyl-2,3,4,4a-tetrahydro-1 tf-carbazole

To a solution of 2,2,6-trimethylcyclohexanone (630 mg, 4.5 mmol) in acetic acid (5.0 mL) at room temperature was added p-tolylhydrazine hydrochloride (784 mg, 4.95 mmol). The resulting mixture was stirred in a microwave reactor at 160 °C for 1 hour. The reaction was quenched by the addition of water (5 mL). The volatiles were evaporated under reduced pressure and the organics were extracted with ethyl acetate (20 mL x 2). The combined organic phase was washed with water (20 mL), saturated aqueous sodium bicarbonate and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to yield the title compound as yellow oil (50 mg, Yield: 5%). 1 H NMR (400 MHz, CDCI 3 ) δ 7.44 (d, J = 7.2 Hz, 1 H), 7.16 (s, 1 H), 7.1 (d, J = 7.2 Hz, 1 H), 2.81-2.76 (m, 1 H), 2.54-2.51 (m, 1 H), 2.39 (s, 3H), 1.98- 1.85 (m, 2H), 1.73-1.69 (m, 1 H), 1.31-1.21 (m, 7H), 0.31 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 228 (M + H + ). Example 88: (±)-(4aS,9aS)-1 ^4a ) 6-tetramethyl-2^4,4a,9,9a-hexahydro- 1H-carbazole

To a solution of the product of Example 87 (74 mg, 0.35 mmol) in methanol (3 mL) was added sodium borohydride (40 mg, 1.05 mmol), stirred at room temperature for 2 days. The reaction was quenched with water (2 mL) and mixture was concentrated under reduced pressure. The aqueous residue was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with water (20 mL), then brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a yellow oil (23 mg, Yield: 30%). R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 6.89-6.83 (m, 2H), 6.57 (d, J=8.4 Hz, 1 H), 3.56 (d, J=2.4 Hz, 1 H), 2.28 (s, 3H), 1.82 (m, 3H), 1.43 (m, 2H), 1.30- 1.25 (m, 5H), 0.89 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 230 (M + H + ).

Example 89: (±)-(4aS,9aS)-1 , 1,4a, 6, 9-pentamethyl-2, 3,4,4a, 9,9a- hexahydro-1 H-carbazole

To a mixture of the product of Example 88 (20 mg, 0.087 mmol) and potassium carbonate (30 mg, 0.21 mmol) in acetonitrile (4 mL) was added methyl iodide (210 mg, 1.5 mmol) and the reaction was heated and stirred at reflux for 3 hours. After cooling to room temperature the reaction mixture was concentrated under reduced pressure and then diluted with ethyl acetate (60 mL). The organic phase was washed with water (10 m L x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and then concentrated in vacuo. Purification of the residue by preparative thin layer chromatography afforded the title compound as yellow oil (12 mg, Yield: 50%). R f = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 6.92 (d, J = 7.6 Hz, 1 H), 6.86 (s, 1 H), 6.47 (d, J = 7.6 Hz, 1H), 2.78-2.76 (m, 1 H), 2.59 (s, 3H), 2.28 (s, 3H), 2.03- 1.98 (m, 1 H), 1.67-1.57 (m, 2H), 1.54-1.42 (m, 2H), 1.31 (s, 3H), 1.25-1.12 (m, 1 H) 0.89 (s, 3H), 0.61 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 244 (M + H + ). Example 90: 1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H-carbazole

To a solution of 2,2,6-trimethylcyclohexanone (630 mg, 4.5 mmol) in acetic acid (9.0 mL) at was added phenylhydrazine (715 mg, 5.0 mmol). The resulting mixture was stirred in a microwave reactor at 160 °C for 1 hour. The reaction was quenched by the addition of water (5 mL). The volatiles were evaporated under reduced pressure and the residue extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (20 mL), saturated aqueous sodium bicarbonate and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to yield the title compound as yellow oil (500 mg, Yield: 52%). R f = 0.2 ( 10:1 p etroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.56 (d, J = 7.6 Hz, 1 H), 7.36 (d, J = 7.2 Hz, 1 H), 7.33-7.26 (m, 1 H), 7.19-7.16 (m, 1 H), 2.70-2.62 (m, 1 H), 2.57- 2.54 (m, 1 H), 1.99-1.92 (m, 2H), 1.72-1.65 (m, 1 H), 1.32-1.26 (m, 7H), 0.29 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 214 (M + H + ).

Example 91 : (±)-(4aS,9aS)-1,1,4a-trlmethyl-2,3,4,4a,9,9a-hexahydro-1H- carbazole

The solution of the product of Example 90 (120 mg, 0.56 mmol) in methanol (3 mL) was added sodium borohydride (64 mg, 1.69 mmol) and the reaction was stirred at room temperature over night. The reaction was quenched with water (2 mL) and the mixture was concentrated under reduced pressure. The aqueous residue was extracted with ethyl acetate (30 mL x 3), washed with water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil. (80 mg, Yield: 66%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.08-7.02 (m, 2H), 6.77-6.73 (m, 1 H), 6.67-6.64 (m, 1 H), 3.59-3.58 (m, 2H), 1.83-1.69 (m, 3H), 1.46-1.43 (m, 2H), 1.32 (s, 3H), 1.25 (m, 1 H), 0.89 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 216 (M + H + ). Example 92: (±)-(4aS,9aS)-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro- 1H-carbazole

To a mixture of the product of Example 91 (50 mg, 0.23 mmol) and potassium carbonate (77.14 mg, 0.56 mmol) in acetonitrile (4 mL) was added methyl iodide (554 mg, 3.91 mmol) and the reaction was stirred at reflux for 3 hours. The volatiles were evaporated under reduced pressure and the residue diluted with ethyl acetate (60 mL). The organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a yellow oil (40 mg, Yield; 76%). R f = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 6 7.13 (dt, J = 7.6, 1.2 Hz, 1 H), 7.07 (dd, J = 7.2, 0.8 Hz, 1 H), 6.75 (dt, J = 7.2, 0.8 Hz, 1H), 6.57 (d, J = 8.0 Hz, 1 H), 2.87-2.85 (m, 1 H), 2.64 (s, 3H), 2.07-2.02 (m, 1 H), 1.69-1.60 (m, 2H), 1.50-1.45 (m, 2H), 1.35 (s, 3H), 1.26 (m, H) 0.91 (s, 3H), 0.62 (s, 3H) ) ppm; Mass spectrum (ESI +ve) m/z 230 (M + H + ).

Example 93: (±)-(4aS,9aS)-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1H-carbazole

Example 93a: 6-methoxy-1,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H- carbazole

To a solution of 2,2,6-trimethylcyclohexanone (210 mg, 1.5 mmol) in acetic acid (3.0 mL) at was added (4-methoxyphenyl)hydrazine hydrochloride (288 mg, 1.65 mmol). The resulting mixture was stirred at reflux for 2 hours. The reaction was quenched by the addition of water (5 mL). The volatiles were evaporated under reduced pressure and the aqueous residue was extracted with ethyl acetate (20 mL x 2). The combined organic phase was washed with water (20 mL), saturated aqueous sodium bicarbonate, brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure, and semi purified by silica gel chromatography to yield the title compound as a yellow oil (50 mg) (Mass spectrum (ESI +ve) m/z 244 (M + H + ).

Example 93: (±)-(4aS,9aS)-6-methoxy-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1H-carbazole

To the product of Example 93a (240 mg, 0.21 mmol) in methanol (10 mL) was added sodium borohyd de (113 mg, 0.62 mmol), stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure and then quenched with water (2 mL). The aqueous residue was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as white solid. (180 mg, Yield: 74%). Mp = 81-81.5 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 +D 2 0) δ 6.62-6.57 (m, 3H), 3.75 (s, 3H), 3.06 (s, 1 H), 1.60-1.45 (m, 2H), 1.43-1.40 (m, 2H), 1.40-1.20 (m, 2H), 1.24 (s, 3H),1.04 (s, 3H), 1.02 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 246 (M + H + ). Example 94: (±)-(4aS,9aS)-6-methoxy-1, 1,4a, 9-tetramethyl-2, 3,4,4a, 9,9a- hexahydro-1 W-carbazole

The solution of the product of Example 93 (50 mg, 0.2 mmol) and potassium carbonate (67.6 mg, 0.49 mmol) in acetonitrile (4 mL) was added methyl iodide (0.18 mL, 3.5 mmol) and the reaction was stirred at reflux for 5 hours. The reaction mixture was concentrated under reduced pressure to and then quenched with water (2 mL). The aqueous residue was extracted with ethyl acetate (30 mL x 3) and the combined organic phase washed with water (20 mL), then brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a yellow oil (38 mg, Yield: 74%). R f = 0.8 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3+D2O) δ 6.62-6.55 (m, 2H), 6.27 (s, 1 H), 3.74 (s, 3H), 2.94 (s, 3H), 2.73 (s, 1 H), 1.83 (m, 2H), 1.56-1.45 (m, 4H), 1.24 (m, 4H), 1.07 (s, 3H), 0.76 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 260 (M + H + ).

Example 95: (3-methoxyphenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol To a stirred solution of (3-methoxyphenyl)magnesium bromide (1.0 M in tetrahydrofuran) (10 mL, 10 mmol) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (609 mg, 4.0 mmol) in dry tetrahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for additional 5 hours. The mixture was cooled down to 0 °C and saturated ammonium chloride (30 mL) was added slowly followed by addition of water (20 mL). The mixture was extracted with ethyl acetate (50 mL x 4) and the combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 25:1) gave the title compound as a colorless oil (877 mg, Yield: 84%), R f = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.25 (dd, J = 8.4, 7.6 Hz, 1 H), 7.03 (d, J = 2.4 Hz, 1 H), 7.00 (d, J = 7.6 Hz, 1 H), 6.76 (dd, J = 8.0, 2.4 Hz, 1 H), 5.37 (d, J = 4.8 Hz, 1 H), 3.81 (s, 3H), 1.98 (t, J = 6.2 Hz, 2H), 1.86 (d, J = 4.8 Hz, 1 H), 1.67-1.61 (m, 2H), 1.56-1.52 (m, 2H), 1.40 (s, 3H), 1.19 (s, 3H), 1.08 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M - H 2 0 + H + ).

Example 96: (3-methoxyphenyl)(2,6,6-trimethylcyclohex-1- enyl)methanone

To a stirred solution of the product of Example 95 (391 mg, 1 .5 mmol) in dichloromethane (13 mL) was added manganese dioxide (1043 mg, 12 mmol). The mixture was stirred at room temperature for 72 hours. The reaction mixture was filtered and the filtrate concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) afforded the title compound as a colorless oil (232 mg, Yield: 60%). R f = 0.5 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.51-7.48 (m, 2H), 7.35 (t, J = 8.0 Hz, 1 H), 7.11-7.08 (m, 1 H), 3.86 (s, 3H), 2.07 (t, J = 6.4 Hz, 2H), 1.80-1.75 (m, 2H), 1.56-1.53 (m, 2H), 1.44 (s, 3H), 1.04 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 259 (M + H + ).

Example 97: (±)-(4aA?,9aS)-7-methoxy-1,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1W-fluoren-9(9aW)-one

A solution of the product of Example 96 (162 mg, 0.59 mmol) in methansulfonic acid (2 mL) was stirred at 50 °C for 3 hours. To the mixture was then added ethyl acetate (50 mL) and the organic phase was washed with water (30 mL), brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by prep- HPLC gave the title compound as a pale yellow oil (142 mg, Yield: 88%). R f = 0.4 (25:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.31 (d, J = 8.8 Hz, 1 H), 7.15 (d, J = 8.8 Hz, 1 H), 7.14 (s, 1 H), 3.83 (s, 3H), 2.19 (s, 1 H), 2.05-2.02 (m, 1 H), 1 .70-1.59 (m, 2H), 1 .46-1.34 (m, 3H), 1.26 (s, 3H), 1.22 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 259 (M + H + ).

Examples 98a and 98b: (±)-(4aR,9S,9aS)-7-methoxy-1 ,1 ,4a-trimethyl- 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol (98a) and (±)-(4aR,9R,9aS)-7- methoxy-1,1,4a-trimethyl-2,3,4,4a,9,9a-hexahydro-1W-fluoren- 9-ol (98b) To a stirred solution of the product of Example 97 (63 mg, 0.24 mmol) in anhydrous tetrahydrofuran (3 mL) was added lithium aluminum hydride (73 mg, 1.92 mmol). The reaction was stirred at room temperature for 5 hours. Water (0.03 mL), aqueous sodium hydroxide (15%, 0.03 mL) and water (30 mL) was added sequentially. The mixture was extracted with ethyl acetate (15 mL x 4) and the combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 30:1 ) afforded the title compound (98a) as white solid (9 mg, Yield: 14%). Mp = 75-78 °C; R f = 0.35 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.12 (d, J = 8.8 Hz, 1 H), 6.92 (s, 1 H), 6.87 (d, J = 9.6 Hz, 1 H), 5.04 (d, J = 5.6 Hz, 1 H), 3.80 (s, 3H), 1.81-1.73 (m, 3H), 1.68 (d, J = 8.8 Hz, 1 H), 1.58-1.53 (m, 1 H), 1.41 (s, 3H), 1.41-1.33 (m, 1 H), 1.28 (s, 3H), 1.28-1.25 (m, 1 H), 1.16 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M - H 2 0 + H + ). The title compound (98b) as a white solid (43 mg, Yield: 68%). Mp = 107-1 10 °C =0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.04 (d, J = 8.0 Hz, 1 H), 6.94 (d, J = 2.4 Hz, 1 H), 6.81 (dd, J = 8.8, 2.4 Hz, 1 H), 4.98 (d, J = 9.2 Hz, 1 H), 3.81 (s, 3H), 1.65-1.54 (m, 3H), 1.46 (s, 3H), 1.44-1.32 (m, 3H), 1.19 (s, 3H), 1.15 (s, 3H), 0.98 (dt, J = 10.8, 3.2 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 243 (M - H 2 0 + H + ).

Example 99: (±)-(4a ?,9R,9aS)-1 ,1 ,4a,6,7-pentamethy l-2,3,4,4a,9,9a- hexahydro-1W-fluoren-9-ol

To a stirred solution of the product of Example 55a (23 mg, 0.09 mmol) in anhydrous tetrahydrofuran (2 mL) was added lithium aluminum hydride (27 mg, 0.72 mmol). The reaction was stirred at room temperature for 5 hours. Water (0.02 mL), sodium hydroxide (15%, 0.02 mL) and H 2 0 (30 mL) was added sequentially. The mixture was extracted with ethyl acetate (15 mL x 4) and the combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100: 1) followed by repurification (eluent: petroleum ether/ethyl acetate = 25:1) afforded the title compound as a white solid (16 mg, Yield: 68%). Mp = 127-130 °C; R f = 0.3 (25:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.17 (s, 1 H), 6.91 (s, 1 H), 4.99 (t, J = 8.4 Hz, 1 H), 2.27 (s, 6H), 1.65-1.56 (m, 3H), 1.47 (s, 3H), 1.47- 1.31 (m, 4H), 1.19 (s, 3H), 1.15 (s, 3H), 0.97 (dt, J = 10.8, 3.2 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 241 (M - H 2 0 + H + ).

Example 100: (perfluorophenyl)(2,6,6-trimethylcyclohex-1-enyl)methanol

To a mixture of magnesium (264 mg, 1 1.0 mmol) in tetrahydrofuran (5 mL) was added iodine (5.0 mg), and then to the mixture was added a solution of 1-bromo-2,3,4,5,6-pentafluorobenzene (247 mg, 1.0 mmol) in tetrahydrofuran (0.5 mL). The reaction mixture was stirred and heated to 60 °C, and a solution of 1-bromo-2,3,4,5,6-pentafluorobenzene ( 2.22 g, 9 mmol) in tetrahydrofuran (4.5 mL) was added drop wise. After 2 hours the reaction mixture was cooled to room temperature and used in the next step.

To the solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (452 mg, 3.0 mmol) in tetrahydrofuran (4.0 mL) at 0 °C was added a 0 °C solution of (perfluorophenyl)magnesium bromide (1.0 M, 10 mL). The mixture was allowed to room temperature and stirred overnight. Saturated aqueous ammonium chloride was added to quench the reaction and the organics were extracted with dichloromethane. The organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a wax (660 mg, Yield: 69%). R f = 0.6 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 5.69 (d, J = 4.8 Hz, 1 H), 2.15 (d, J = 4.8 Hz, 1 H), 2.03 (t, J = 6.4 Hz, 2 H), 1.72 (s, 3 H), 1.64-1.59 (m, 2 H), 1.49-1.46 (m, 2 H), 1.13 (s, 3 H), 0.94 (s, 3 H) ppm; Mass spectrum (El +ve) m/z 320 (M + ).

Example 101: (perfluorophenyl)(2,6,6-trimethylcyclohex-1- enyl)methanone

To a solution of the product of Example 100 (160 mg, 0.5 mmol) in dichloromethane (3 mL) at 0 °C was added sodium bicarbonate (42 mg, 0.5 mmol) and Dess-Martin periodinane (424 mg, 1.0 mmol). The reaction was stirred for 0.5 hours and then allowed to warmed to room temperature and stirred for an additional 2 hours. The mixture was poured onto the 2N HCI and extracted with dichloromethane (150 mL). The organic layer was washed by saturated aqueous sodium bicarbonate (50 mL x 2) and brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica column chromatography to give the title compound as a white solid (100 mg, Yield: 63%). Mp = 39-41 °C; R f = 0.7 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 2.10 (t, J = 6.4 Hz, 1 H), 1.73-1.70 (m, 2 H), 1.60 (s, 3 H), 1.52-1.49 (m, 2 H), 1.09 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 319 (M + H + ). Example 100: (±)-(4aR,9R,9aS)-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

The product of Example 41 (50 mg, 0.21 mmol) was added to 1 M borane in tetrahydrofuran (1.7 mL, 1.7 mmol) at -78 °C under argon, and the reaction mixture was stirred at that temperature for 10 minutes and then it was warmed to room temperature and stirred for 20 hours. The mixture was cooled to 0 °C and 95% ethanol was added to quench the reaction and then a solution of sodium hydroxide (136.2 mg) in water (2.4 mL) and 30% hydrogen peroxide (1.26 mL) were sequentially added slowly with stirring. After stirring for 2 hours at room temperature, the mixture was poured into saturated aqueous ammonium chloride, and extracted with diethyl ether (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a white solid (30 mg, Yield: 54%). Mp = 107.2- 107.9 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate; H NMR (400 MHz, CDCI3+D20) δ 7.30 (d, J = 8.0 Hz, 1 H), 7.19 (dd, J = 8.0, 2.0 Hz, 1 H), 7.09 (d, J = 1.6 Hz, 1H), 4.98 (d, J = 8.8 Hz, 1H), 1.67-1.54 (m, 3H), 1.46-1.34 (m, 6H), 1.18 (s, 3H), 1.15 (s, 3H), 1.02-0.96 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M - H 2 0 + H + ).

Example 102: 6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-carbazole

To a solution of 2,2,6-trimethylcyclohexanone (630 mg, 4.5 mmol) in acetic acid (9.0 mL) at room temperature was added (4- fluorophenyl)hydrazine hydrochloride salt (810 mg, 5.0 mmol). The resulting mixture was stirred in a microwave reactor at 160 °C for 1 hour. After cooling, the reaction was quenched by the addition of water (5 mL). The volatile organics were evaporated under reduced pressure the aqueous residue and extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (20 mL), saturated aqueous sodium bicarbonate, brine (20 ml), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by flash column chromatography to give the title compound as a yellow solid (30 mg, Yield: 3 %). 1 H NMR (400 MHz, CDCI 3 ) δ 7.50 (m, 1 H), 7.06 (d, J = 8.4 Hz, 1 H), 7.00 (t, J = 8.8 Hz, 1 H), 2.80 (dd, J = 13.6, 4.4 Hz, 1 H), 2.54 (dt, J = 13.2, 6.4 Hz, 1 H), 2.00-1.86 (m, 2 H), 1.73- 1.61 (m, 1 H), 1.33-1.25 (m, 1 H), 1.26 (s, 3H), 1.25 (s, 3 H), 0.33 (s, 3 H). Example 103: (4-methoxy-3-methylphenyl)(2,6,6-trimethylcyclohex-1- enyl)methanol

To a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (1.2 g, 8 mmol) in tetrahydrofuran (8 mL) at -78 °C was added (4-methoxy-3- methylphenyl)magnesium bromide (12 mmol, 24 mL). The solution was allowed to warm to room temperature and stirred for 2 hours. The reaction was quenched with saturated aqueous ammonium chloride and partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was washed with water (15 mL) then brine (15 mL), dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 25:1 ) to afford the title compound as a colorless oil (952 mg, Yield: 43%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ): δ 7.21 (d, J = 2.8 Hz, 1 H), 7.15 (d, J = 8.0 Hz, 1 H), 6.76 ( d, J = 8.4 Hz, 1 H), 5.36 (d, J = 5.2 Hz, 1 H), 3.82 (s, 3H), 2.22 (s, 3H), 2.02-1.98 (m, 2H), 1.80 (d, J = 4.8 Hz, 1 H), 1.66-1.63 (m, 2H), 1.55-1.51 (m, 2H), 1.45 (s, 3H), 1.18 (s, 3H), 1.02 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 257 (M - H 2 0 + H + ).

Example 104: (4-methoxy-3-methylphenyl)(2,6,6-trimethylcyclohex-1- enyl)methanone

To a solution of the product of Example 103 (280 mg, 1.02 mmol) in dichloromethane (10 mL) was added manganese dioxide (0.9 g, 10.2 mmol). The reaction was stirred overnight at room temperature. The insoluble solid was filtered off and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) to give the title compound as a light yellow oil (55 mg, Yield: 20%). R f = 0.4 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ): δ 7.77 (s, 1 H), 7.76 (d, J = 8.4 Hz, 1 H), 6.83 (d, J = 8.4 Hz, 1 H), 3.89 (s, 3H), 2.24 (s, 3H), 2.07 (t, J = 6.4 Hz, 2H), 1.78-1.74 (m, 2H), 1.56- 1.52 (m, 2H), 1.44 (s, 3H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 273 (M + H + ). Example 105: (±)-(4a ?,9aS)-6-methoxy-1,1 ,4a,7-tetramethyl-2,3,4,4a- tetrahydro-1 tf-fluoren-9(9atf)-one

A solution of the product of Example 104 (180mg, 0.66 m mol) in methansulfonic acid (5 mL) was stirred at 50 °C for 2 hours. After cooling to room temperature, the reaction was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was washed with water (15 mL) and brine (15 mL), dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ ethyl acetate = 50:1) to afford the title compound as white solid (75 mg, Yield: 42%). Mp = 118.1-122.9 °C; R f = 0.4 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 + D 2 0): δ 7.45 (s, 1 H), 6.75 (s, 1 H), 3.93 (s, 3H), 2.21 (s, 3H), 2.13 (s, 1 H), 2.03-2.00 (m, 1 H), 1.71-1.59 (m, 2H), 1.47-1.35 (m, 3H), 1.27 (s, 3H), 1.22 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 273 (M + H + ). Example 106: (±)-5,5,8a-trimethyl-6,7,8,8a-tetrahydro-5W-lndeno[1 ,2- 6]thiophene

To a stirred solution of the product of Example 80 (73 mg, 0.33 mmol) in anhydrous dichloromethane (6 mL) at 0 °C was added thionyl chloride (0. 2 μί). The reaction mixture was stirred at room temperature for 2 hours. Saturated aqueous sodium bicarbonate was added to quench the reaction and the mixture was extracted with dichloromethane (60 mL). The organic phase was washed with water (20 mL x 3), saturated aqueous sodium bicarbonate (20 mL x 2) and brine (30 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether) to give the title compound as a colorless oil (19 mg, Yield: 29%); R f = 0.9 (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.19 (d, J = 4.8 Hz, 1 H), 6.90 (d, J = 4.8 Hz, 1 H), 6.32 (s, 1 H), 2.14-2.10 (m, 1 H), 1.95-1.84 (m, 1 H), 1.66-1.59 (m, 2H), 1.40 (s, 3H), 1.27 (s, 3H), 1.22 (s, 3H), 1.18-1.10 (m, 2H) ppm; Mass spectrum (ESI +ve) m/z 219 (M + H + ).

Example 107: (4-(trifluoromethyl)phenyl)(2,6,6-trimethylcyclohex-1- enyl)methanol

To a solution of 4-bromobenzotrifluoride (1.02 g, 4.5 mmol) in tetrahydrofuran (20 mL) at -78 °C was added dropwise n-butyl lithium (1 .8 mL, 2.5 M). Then to the reaction solution was added 2,6,6-trimethylcyclohex-1- enecarbaldehyde (456 mg, 3.0 mmol) and the reaction was stirred overnight while letting warm slowly to room temperature. The mixture was quenched by the addition of saturated aqueous ammonium chloride (40 mL). The solution was extracted with ethyl acetate and the organic phase dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue was purified by sliica gel column chromatography (eluent: petroleum ether/ ethyl acetate = 50:1 -> 20:1) to afford the title compound as a colorless oil (190 mg, Yield: 22%). R f = 0.3 (10: 1 petroleum ether/ ethyl acetate); H NMR (400 MHz, CDC ) δ 7.57 (d, J = 8.8 Hz, 2H), 7.54 (d, J = 8.8 Hz, 2H), 5.40 (s, 1 H), 1.98 (t, J = 6.0 Hz, 2 H), .87 (d, J = 4.8 Hz, 1 H), 1.68- .61 (m, 2 H), .60- .53 (m, 2 H), 1.32 (s, 3 H), 1.20 (s, 3 H), 1.10 (s, 3 H) ppm; Mass spectrum (El +ve) m/z 298 (M + ).

Example 108: (4-(trifluoromethyl)phenyl)(2,6,6-trimethylcyclohex-1-enyl) methanone

To a solution of the product of Example 107 (160 mg, 0.54 mmol) in dichloromethane (3.0 mL) at 0 °C was added sodium bicarbonate (45.4 mg, 0.54 mmol) and Dess-Martin periodinane (454.8 mg, 1.08 mmol). The mixture was stirred for 1 hour and, then stirred at room temperature for 2 hours. To the reaction mixture was quenched with saturated aqueous sodium bicarbonate (40 mL) and then extracted with dichloromethane, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ ethyl acetate = 80:1 -> 60:1 ) to afford the title compound as a colorless oil (90 mg, Yield 56%). R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 8.04 (d, J = 8.0 Hz, 2 H), 7.22 (d, J = 8.0 Hz, 2 H), 2.10 (t, J = 6.8 Hz, 2 H), 1.80-1.76 (m, 2 H), 1.59- 1.55 (m, 2 H), 1.43 (s, 3 H), 1.03 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 320 (M + Na + ).

Example 109: (±)-(4a ?,9aS)-1 ,1 ,4a-trimethy l-e-ftrifluoromethyl^.S^^a- tetrahydro-1 H-fluoren-9(9aH)-one

The product of Example 108 (50 mg, 0.17 mmol) was dissolved in methansulfonic acid (1.0 mL). The mixture was stirred at room temperature for 1 hour and then at 50 °C for 2 hours. The reaction was cooled to room temperature and water (6 mL) was added. The reaction mixture was extracted with ethyl acetate (10 mL x 2) then combined the organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 -> 50:1) to give the title compound as yellow solid (12 mg, Yield: 24%). Mp = 60-62 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.98 (d, J = 8.0 Hz, 1 H), 7.71 (s, 1 H), 7.62 (d, J = 8.0 Hz, 1 H), 2.26 (s, 1 H), 2.15-2.09 (m, 2 H), 1.75-1.63 (m, 2 H), 1.45-1.36 (m, 3 H), 1.30 (s, 3 H), 1.22 (s, 3 H), 0.63 (s, 3 H) ppm; Mass spectrum (El +ve) m/z 296 (M + ).

Example 110: (3-fluoropheny l)(2,6,6-trimethy lcyclohex-1 -enyl)methanol

To the mixture of magnesium (528 mg, 22.0 mmol) and tetrahydrofuran (10 mL) was added iodine (5.0 mg) and then to this mixture was added a solution of 1-bromo-3-fluorobenzene (350 mg, 2 mmol) in tetrahydrofuran (1.0 mL). The reaction mixture was heated to 60 °C, and a solution of 1-bromo-3- fluorobenzene (3.15 g, 18 mmol) in tetrahydrofuran (9.0 mL) was added by dropwise. Two hours later the homogenous solution was cooled to room temperature and used directly.

To the solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (608 mg, 4.0 mmol) in tetrahydrofuran (4.0 mL) at 0 °C was added a 0 °C solution of (3- fluorophenyl)magnesium bromide (1.0 M, 20 mL). The mixture was allowed to room temperature and stirred overnight. The reaction mixture was quenched by the addition of saturated aqueous ammonium chloride. The reaction was extracted with dichloromethane, dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (700 mg, Yield: 71%). p = 62-52.8 °C; R f = 0.6 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.29-7.23 (m, 1 H), 7.20-7.13 (m, 2 H), 6.89 (t, J = 8.4 Hz, 1 H), 5.36 (d, J = 5.2 Hz, 1 H), 1.98 (t, J = 6.0 Hz, 2 H), 1.81 (d, J = 5.6 Hz, 1 H), 1.67-1.52 (m, 4 H), 1.36 (s, 3 H), 1.19 (s, 3 H), 1 .09 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ).

Example 111 : (3-fluoropheny l)(2,6,6-trimethy lcyclohex-1 -enyl)methanone

To a solution of the product of Example 110 (654.1 mg, 2.66 mmol) in dichloromethane (12 mL) at 0 °C was added sodium bicarbonate (223.4 mg, 2.66 mmol) and Dess-Martin periodinane (2.25 g, 5.32 mmol). The mixture was stirred at this temperature for 1 hour, warmed to room temperature and stirred for an additional 2 hours. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate (100 mL). The reaction mixture was extracted with dichloromethane and the organic phase dried over anhydrous magnesium sulfate, filtered and then concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 70:1 -> 60:1) to afford the title compound as a colorless oil (470 mg, Yield: 71 %). R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.72 (d, J = 7.6 Hz, 1 H), 7.61 (d, J = 9.6 Hz, 1 H), 7.43 (ddd, J = 8.0, 8.0, 5.6 Hz, 1 H), 7.24-7.22 (m, 1 H), 2.08 (t, J = 6.4 Hz, 2 H), 1.79-1.74 (m, 2 H), 1.59-1.54 (m, 1 H), 1.44 (s, 3 H), 1.03 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 269 (M + Na + ).

Examples 112a and 112b: (+)-(4aR,9aS)-7-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1W-fluoren-9(9aW)-one (112a) and (±)-(4a/?,9aS)-5-fluoro- 1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H-fluoren-9(9aW)-one

The product of Example 1 11 (400 mg, 1.62 mmol) was dissolved in methanesulfonic acid (10 mL) and the mixture was stirred at room temperature for 1 hour and then at 50 °C for 2 hours. The reaction was cooled to room temperature and quenched by the addition of water (50 mL). The reaction was extracted with ethyl acetate and the organic phase dried over anhydrous magnesium sulfate, filtered and then concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 50:1) then further purified by Prep-HPLC to afford the title compound (112a) as a colorless oil (200 mg, Yield 50 %). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.38 (dd, J = 8.0, 4.4, 1 H), 7.31 (dd, J = 7.6, 2.4, 1 H), 7.29-7.24 (m, 1 H), 2.22 (s, 1 H), 2.08- 1.97 (m, 2 H), 1.72-1.61 (m, 2 H), 1.47-1.35 (m, 3 H), 1.27 (s, 3 H), 1.21 (s, 3 H), 0.66 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). The title compound (112b) as a white solid (25 mg, Yield 6%). Mp = 41-42 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.50 (d, J = 7.2 Hz, 1 H), 7.34-7.26 (m, 1 H), 7.23-7.19 (m, 1 H), 2.25-2.19 (m, 1 H), 2.15 (s, 1 H), 1.91-1.84 (m, 1 H), 1.74-1.68 (m, 1 H), 1.59-1.51 (m, 1 H), 1.42-1.39 (m, 5 H), 1.25 (s, 3 H), 0.74 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ).

Example 113: (±)-(4a ?,9 ?,9aS)-7-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1W-fluoren-9-ol

To a mixture of lithium aluminum hydride (38.7 mg, 1.02 mmol) in tetrahydrofuran (4 mL) at °C was added dropwise the product of Example 112a (62.7 mg, 0.25 mmol) in tetrahydrofuran (5 mL) and the reaction was stirred for 2 hours. The reaction was quenched by the addition of water (0.2 mL), aqueous 1 N sodium hydroxide (0.2 mL), then water (0.6 mL). The reaction mixture was extracted with ethyl acetate and the organic layer dried over anhydrous magnesium sulfate, filtered and the volatiles were evaporated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 30:1) to afford the title compound as a white solid (45 mg, Yield 72%). Mp = 109.2-1 10 °C; R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.08-7.04 (m, 2 H), 6.92 (dd, J = 9.6, 7.6 Hz, 1 H), 4.99 (dd, J = 8.8, 8.0 Hz, 1 H), 1.72-1.55 (m, 3 H), 1.47 (s, 3 H), 1.45-1.39 (m, 3 H), 1.19 (s, 3 H), 1.16 (s, 3 H), 0.97 (dt, J = 4.4, 12.8 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ).

Example 114: (3-chloro-4-fluorophenyl)(2,6,6-trimethylcyclohex-1-enyl) methanol

To a solution of (3-chloro-4-fluorophenyl)magnesium bromide (20 ml_, 0.5 M in tetrahydrofuran) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (609 mg, 4.0 mmol) in tetrahydrofuran (10 mL). The reaction mixture was warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of saturated aqueous ammonium chloride and the mixture was extracted with ethyl acetate (150 mL). The combined organic phase was washed with brine (50 mL x 2) and dried over anhydrous sodium sulfate. Concentration under reduced pressure and purification of the residue by silica gel column chromatography afforded the title compound as a white solid (823 mg, Yield: 73%). Mp = 109-110 °C; R f = 0.3 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 57.51 (d, J = 5.6 Hz, 1 H), 7.29-7.25 (m, 1 H), 7.10 (t, J = 8.8 Hz, 1 H), 5.35 (d, J = 4.0 Hz, 1 H), 2.02 (t, J = 6.4 Hz, 2H), 1.85 (d, J = 4.8 Hz, 1 H), 1.69-1.66 (m, 2H), 1.60-1.55 (m, 2H), 1.40 (s, 3H), 1.21 (s, 3H), 1.09 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 265 (M - H 2 0 + H + ).

Example 115: (3-chloro-4-fluorophenyl)(2,6,6-trimethy lcyclohex-1 -enyl) methanone

To a solution of the product of Example 114 (200 mg, 0.71 mmol) in dichloromethane (10 mL) was added manganese dioxide (617 mg, 7.1 mmol) and the reaction mixture was stirred overnight. The mixture was filtered, concentrated under reduced pressure and purified by silica gel column chromatography to give the title compound as colorless oil (164 mg, Yield: 82%). R f = 0.6 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 8.03 (dd, J = 7.2, 2.0 Hz, 1 H), 7.86-7.83 (m, 1 H), 7.24 (t, J = 8.6 Hz, 1 H), 2.13-2.10 (m, 2H), 1.82-1.78 (m, 2H), 1 .60-1.57 (m, 2H), 1.47 (s, 3H), 1.06 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 281 (M + H + ). Examples 116a and 116b: (±)-(4aR,9aS)-7-chloro-6-fluoro-1 ,1 ,4a- trimethyl-2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)-one (116a) and (±)- (4a ?,9aS)-5-chloro-6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9a/Y)-one (116b)

A mixture of the product of Example 115 (296 mg, 1.05 mmol) and methanesulfonic acid (6 mL) was stirred at 50 °C for 3 hours. Water (50 mL) was added and the mixture was extracted with ethyl acetate (150 mL). The combined organic phase was washed with water (50 mL x 2) and brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated in vacuo. Purification of the residue by preparative thin layer chromatography afforded the title compound (1 16a) as a white solid (40 mg, Yield: 14%). Mp = 79-81 °C; R f = 0.5 (100:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 67.74 (d, J = 7.6 Hz, 1 H), 7.20 (d, J = 8.4 Hz, 1 H), 2.22 (s, 1 H), 2.06-2.02 (m, 1 H), 1.70-1.65 (m, 2H), 1.44-1.39 (m, 3H), 1.29 (s, 3H), 1.23 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 281 (M + H + ). The title compound 116b as a colorless oil (47 mg, Yield: 16%); R f = 0.45 (100:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ7.62 (dd, J = 8.2, 5.0 Hz, 1 H), 7.15 (t, J = 8.4 Hz, 1H), 2.32-2.25 (m, 1H), 2.17 (s, 1 H), 2.14-2.11 (m, 1 H), 1.77-1.71 (m, 1 H), 1.64-1.58 (m, 1 H), 1.49 (s, 3H), 1.46-1.44 (m, 2H), 1.26 (s, 3H), 0.75 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 281 (M + H + ).

Example 117: (+)-(4a ?,9ft,9aS)-7-chloro-6-fluoro-1 ,1 ,4a-trimethy I- 2,3,4,4a,9,9a-hexahydro-1W-fluoren-9-ol

To a solution of the product of Example 116a (38 mg, 0.14 mmol) in methanol (2 mL) and tetrahydrofuran (1.5 mL) was added sodium borohydride (42.4 mg, 1.12 mmol) and the reaction mixture was stirred at room temperature overnight. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate =100:1->80:1) to afford the title compound as a white solid (32 mg, Yield: 81 %). Mp = 136-138 °C; R f = 0.3 (100 :1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.39 (d, J = 6.4 Hz, 1 H), 6.89 (d, J = 9.6 Hz, 1 H), 4.97 (d, J = 8.8 Hz, 1 H), 1.68 (d, J = 8.8 Hz, 1 H), 1.62-1.53 (m, 2H), 1.47-1.43 (m, 1 H), 1.45 (s, 3H), 1.40-1.34 (m, 2H), 1.18 (s, 3H), 1.15 (s, 3H), 1.00-0.96 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 265 (M - H 2 0 + H + ).

Example 118: (±)-(4a ?,9R,9aS)-5-chloro-6-fluoro-1 ,1 ,4a-trimethyl- 2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 116b (47 mg, 0.14 mmol) in methanol (2 mL) and tetrahydrofuran ( .5 mL) was added sodium borohydride (51.4 mg, 1.36 mmol) and the mixture was stirred at room temperature overnight. The mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate = 100:1-> 60:1) to afford the title compound as a white solid (37 mg, Yield: 77%). Mp = 64-66 °C; R f = 0.2 (50 A petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.26-7.23 (m, 1 H), 7.02 (t, J = 8.8 Hz, 1 H), 4.88 (d, J = 8.0 Hz, 1 H), 2.15-2.11 (m, 1 H), 1.81 (bs, 1 H), 1.75 (s, 3H), 1.72 (d, J = 8.8 Hz, 1 H), 1.65-1.60 (m, 1 H), 1.47-1.34 (m, 3H), 1.21 (s, 3H), 1.17 (s, 3H), 1.13-1.05 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 265 (M - H 2 0 + H + ).

Example 119: o-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (604 mg, 4 mmol) in tetrahydrofuran (8 mL) at -78 °C was added o-tolylmagnesium bromide (8 mL) and the solution was allowed to warm to room temperature and stirred for 2 hours. The solution was quenched with saturated aqueous ammonium chloride and partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was washed with water (15 mL x 2) then brine ( 5 mL), dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/acetone = 100:1 ) to afford the title compound as a yellow oil (362 mg, Yield 37%). R f = 0.4 (20:1 petroleum ether/acetone); 1 H NMR (400 MHz, CDCI3) δ 7.38 (d, J = 7.2 Hz, 1 H), 7.22-7.10 (m, 3H), 5.63 (s, 1 H), 2.53 (s, 3H), 2.20-2.05 (m, 2H), 1.78 (s, 3H), 1.74-1.65 (m, 2H), 1.51-1.45 (m, 2H), 1.15 (s, 3H), 0.80 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 227 (M - H 2 0 + H + ). Example 120: o-tolyl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 119 (0.5 g, 2.5 mmol) in dichloromethane (50 mL) was added manganese dioxide (1.78 g, 20.5 mmol). The reaction mixture was stirred overnight. The solution was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/acetone 200:1 -> 50:1 to give the title compound as a light yellow oil (255 mg, Yield 51 %). R f = 0.5 (100:1 petroleum ether/acetone); 1 H NMR (400 MHz, CDCI 3 ) δ 7.68 (d, J = 8.0 Hz, 1H), 7.35 (t, J - 7.2 Hz, 1H), 7.26-7.20 (m, 2H), 2.63 (s, 3H), 2.06 (t, J = 6.8 Hz, 2H), 1.78-1.71 (m, 2H), 1.54-1.45 (m, 2H), 1.46 (s, 3H), 1.04 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 121 : (±)-(4afl,9aS)-1 ,1 ,4a,8-tetramethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aW)-one

A solution of the product of Example 120 (0.2 g, 0.8 mmol) in methansulfonic acid (5 mL) was stirred at 50 °C for 1 hour. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL), the organic layer was washed with saturated aqueous sodium bicarbonate (10 mL) and brine, dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/acetone (200:1 -> 50:1) to afford the title compound as a light yellow oil (25 mg, Yield 12%). R f = 0.5 (100:1 petroleum ether/acetone); H NMR (400 MHz, CDCI 3 ) δ 7 Λ0 (t, J = 8.0 Hz, 1 H), 7.22 (d, J = 7.6 Hz, 1 H), 7.06 (d, J = 7.6 Hz, 1 H), 2.60 (s, 3H), 2.15-2.1 1 (m, 2H), 1.62-1.59 (m, 2H), 1.36-1.33 (m, 3H), 1.24 (s, 3H), 1.20 (s, 3H), 0.58 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 243 (M + H + ).

Example 122: (+)-(4aR,9R,9aS)-1 ,1 ,4a, 8-tetramethyl-2, 3,4,4a, 9,9a- hexahydro-1 H-fluoren-9-ol

To a stirred solution of the product of Example 121 (55 mg, 0.23 mmol) in anhydrous tetrahydrofuran (3 mL) cooled to 0 °C was added lithium aluminum hydride (62 mg, 1.63 mmol). The reaction was warmed to room temperature and stirred for 5 hours. Water (0.03 mL), aqueous sodium hydroxide (15%, 0.03 mL) and water (0.09 mL) was added sequentially followed by addition of water (30 mL). The mixture was extracted with ethyl acetate (15 mL x 4) and the combined organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) afforded the title compound as a pale yellow solid (28 mg, Yield: 50%). Mp = 76-78 °C; R f = 0.3 (25:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.17 (t, J = 7.2 Hz, 1 H), 7.00 (d, J = 7.6 Hz, 1 H), 6.97 (d, J = 7.2 Hz, 1 H), 5.13 (t, J = 8.0 Hz, 1 H), 2.45 (s, 3H), 1.68 (d, J = 7.6 Hz, 1 H), 1.63-1.50 (m, 3H), 1.49 (s, 3H), 1.41-1.34 (m, 3H), 1.17 (s, 3H), 1.16 (s, 3H); 1.00-0.96 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 227 (M - H 2 0 + H + ).

Example 123: (±)-(4aft,9/?,9aS)-6-chloro-1 ,1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1W-fluoren-9-ol

To a solution of the product of Example 61 (17 mg, 0.065 mmol) in methanol (2 mL) was added sodium borohydride (10 mg, 0.19 mmol) and the reaction mixture was stirred at room temperature for 2 days. The reaction mixture was concentrated under reduced pressure and the residue was portioned between water (2 mL) and ethyl acetate (30 mL). The organic phase was washed with water (20 mL) and brine (20 mL)L, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as white solid (12 mg, Yield 70%). Mp = 1 10.2-110.8 °C; R, = 0.6 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.30 (d, J = 8.0 Hz, 1 H), 7.19 (d, J = 8.0 Hz, 1 H), 7.08 (s, 1 H), 4.99 (t, J = 8.0 Hz, 1 H), 1 .72 (d, J - 7.2 Hz, 1 H), 1.67-1 .60 (m, 2H), 1.58-1.53 (m, 1 H), 1.46 (s, 3H), 1.45- 1.25 (m, 3H), 1.18 (s, 3H), 1.15 (s, 3H), 0.97 (dt, J = 3.2, 13.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M - H 2 0 + H + ). Example 124: (3,4-difluorophenyl)(2,6,6-trimethylcyclohex-1- enyl)methanol

To a mixture of iodine (5 mg) and magnesium (0.26 g, 11 mmol) tetrahydrofuran (5 ml_) was dropwise added 4-bromo-1 ,2-difluorobenzene (1.93 g, 10 mmol) and the reaction was stirred at room temperature 5 minutes, warmed to reflux for about 2 hours and the resulting solution was used directly in the next step.

To a solution of (3,4-difluorophenyl)magnesium bromide in tetrahydrofuran at 0 °C was added 2,6,6-trimethylcyclohex-l-enecarbaldehyde (608 mg, 4.0 mmol). The reaction was stirred at room temperature for 2 hours and then the reaction mixture was quenched with saturated aqueous ammonium chloride (5 mL). The mixture was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid (500 mg, Yield: 47%). Mp = 56-58 °C; R, = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.26-7.21 (m, 1 H), 7.11-7.06 (m, 1 H), 5.32 (d, J = 4.4 Hz, 1 H), 1.98 (t, J = 6.0 Hz, 2H), 1.81 (d, J = 4.8 Hz, 1 H), 1.66-1.62 (M, 2H), 1.57-1.51 (m, 2H), 1.36 (s, 3H), 1.17 (s, 3H), 1.06 (s, 3H) ) ppm; Mass spectrum (El +ve) m/z 266 (M + ).

Example 125: (3,4-difluorophenyl)(2,6,6-trimethylcyclohex-1-enyl) methanone

To a mixture of the product of Example 124 ( 00 mg, 0.46 mmol) and sodium bicarbonate (40 mg, 0.46 mmol) in dichloromethane (10 mL) at 0 °C was added Dess-Martin periodinane (393 mg, 0.93 mmol). The reaction was stirred at 0 °C for 30 minutes and then it was warmed to room temperature and stirred for 4 hours. The reaction was quenched by the addition of 5% HCI (2 mL) and the resulting mixture was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a white solid (40 mg, Yield: 33%). Mp = 39-42 °C; R f = 0.5 (10:1 petroleum ether/ethyl acetate); H > NMR (400 MHz, CDCI3) δ 7.80-7.69 (m, 2H), 7.26-7.21 (m, 1 H), 2.08 (t, J = 6.4 Hz, 2H), 1.79-1.75 (m, 2H), 1.57-1.53 (m, 2H), 1.43 (s, 3H), 1.02 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 265 (M + H + ).

Example 126: (±)-(4aR ) 9aS)-6,7-difluoro-1,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1 W-fluoren-9(9atf)-one

The product of Example 125 (32 mg, 0.12 mmol) in methanesulfonic acid (2 mL) was stirred at 50 °C overnight. Water (2 mL) was added and the resulting solution extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as white solid (10 mg, Yield: 31%). Mp = 87.2-88.5 °C; R, = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.44 (t, J = 8.0 Hz, 1 H), 7.21 (t, J = 7.2 Hz, 1 H), 2.18 (s, 1 H), 2.01-1.95 (m, 1 H), 1.68-1 .64 (m, 2H), 1.45-1.38 (m, 3H), 1.27 (s, 3H), 1.21 (s, 3H), 0.67 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 265 (M + H + ).

Example 127: (3,4,5-trifluorophenyl)(2,6,6-trimethy lcyclohex-1 -enyl) methanol

To a mixture of magnesium (264 mg, 1 1.0 mmol) and tetrahydrofuran (5 mL) was added iodine (5.0 mg) and a solution of 5-bromo-1 ,2,3- trifluorobenzene ( 21 1 mg, 1 mmol) in tetrahydrofuran (0.5 mL). The mixture was heated to 60 °C, and a solution of 5-bromo-1 ,2,3-trifluorobenzene (1.9 g, 9 mmol) in tetrahydrofuran (4.5 mL) was added dropwise. Two hours later the solution was cooled to room temperature and used directly in the next step. To a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (452 mg, 3.0 mmol) in tetrahydrofuran (4.0 mL) at 0 °C was added (3,4,5- trifluorophenyl)magnesium bromide (1.0 M, 10 mL). The mixture was allowed to warm to room temperature and stirred for overnight. The reaction was quenched with saturated aqueous ammonium chloride and then it was extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (582 mg, Yield: 68% yield). Mp = 43.8-44.9 °C; R, = 0.6 (20:1 petroleum ether/ethyl acetate); 1 H N R (400 MHz, CDCI 3 ) δ 7.04 (t, J = 8.0 Hz, 2 H), 5.26 (d, J = 4.4 Hz, 1 H), 1.98 (t, J = 6.4 Hz, 2 H), 1.80 (d, J = 4.8 Hz, 1 H), 1.676-1.49 (m, 4 H), 1.35 (s, 3 H), 1.17 (s, 3 H), 1.07 (s, 3 H) ppm; Mass spectrum (El +ve) m/z 284 (M + ). Example 128: (3,4,5-trifluorophenyl)(2,6,6-trimethylcyclohex-1-enyl) methanone

To a mixture of the product of Example 127 (100 mg, 0.36 mmol) and sodium bicarbonate (31 mg, 0.36 mmol) in dichloromethane (10 mL) at 0 °C was added Dess-Martin periodinane (299 mg, 0.70 mmol) and the reaction was stirred for 30 minutes, warmed to room temperature and stirred for an additional 4 hours. The reaction was quenched by the addition of 5% HCI (2 mL) and the resulting solution was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), and brine (20 mL) then dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by preparative thin layer chromatography to give the title compound as colorless oil (60 mg, Yield: 59%). R f = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.57 (t, J = 7.6 Hz, 2H), 2.08 (t, J = 6.4 Hz, 2H), 1.78-1.74 (m, 2H), 1.58-1.55 (m, 2H), 1.43 (s, 3H), 1.01 (s, 6H) ) ppm; Mass spectrum (ESI +ve) m/z 283 (M + H + ). Example 129: (±)-(4aR,9aS)-5,6,7-trifluoro-1 ,1 ,4a-trimethy l-2,3,4,4a- tetrahydro-1 W-fluoren-9(9aH)-one

A mixture of the product of Example 128 (50 mg, 0.17 mmol) in methanesulfonic acid (2 mL) was stirred at 50 °C overnight. Water (2 mL) was added to quench the reaction and the solution was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a yellow solid (10 mg, Yield: 20%). Mp = 47-50 °C; R, = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.31 (t, J = 6.8 Hz, 1 H), 2.17-2.12 (m, 2H), 1.91-1.87 (m, 1 H), 1.78-1.62 (m, 1 H), 1.58-1.48 (m, 1 H), 1.43 (m, 5H), 1.24 (s, 3H), 0.75 (s, 3H) ppm; Mass spectrum (El +ve) m/z 282 (M + ).

Example 130: (4-(trifluoromethoxy )pher»y ( )(2,6,6-trimethy lcyclohex-1 - enyl) methanol

To a solution of 2,6,6-trimethylcyclohex-1-enecarbaidehyde (0.93 g, 6.0 mmol) in tetrahydrofuran (10.0 mL) at -78 °C was added (4- (trifluoromethoxy)phenyl)magnesium bromide (0.5 M, 24 mL, 12 mmol). The mixture was stirred for 1 hour at this temperature and then allowed to warm to room temperature and stirred for an additional 2 hours. The reaction was quenched by the addition of saturated aqueous ammonium chloride and then the solution was extracted with ethyl acetate (2 x 40 mL). The organic layer was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a light yellow oil (1 .64 g, Yield: 87%), R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.44 (d, J = 8.0 Hz, 2H), 7.15 (d, J = 8.0 Hz, 2H), 5.38 (d, J = 6.0 Hz, 1 H), 1.98 (t, J = 6.8 Hz, 2H), 1.84 (d, J = 6.0 Hz, 1 H), 1.66-1.51 (m, 4H), 1.35 (s, 3 H), 1.18 (s, 3 H),1.07 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 297 (M - H 2 0 + H + ). Example 131 : (4-(trifluoromethoxy)pheny l)(2,6,6-trimethy lcyclohex-1 - enyl) methanone

To a slurry of manganese dioxide (0.869 g, 10.0 mmol) in dichloromethane (10.0 mL) was added the product of Example 130 (314 mg, 1.0 mmol) and the mixture was stirred overnight. The reaction was filtered and the filtrate concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a yellow oil (200 mg, Yield 64%). R f = 0.8 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 7.99-7.96 (m, 2H), 7.28-7.26 (m, 2H), 2.08 (t, J = 6.4 Hz, 2H), 1.78-1.74 (m, 2H), 1.56-1.42 (m, 2H), 1.43 (s, 3 H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 313 (M + H + ).

Example 132: (±)-1 ,1 ,4a-trlmethy l-6-(trifluoromethoxy)-2,3,4,4a- tetrahydro-1 H-fluorene

To a stirred solution of the product of Example 130 (100 mg, 0.318 mmol) in dichloromethane (6.0 mL) at 0 °C was added stannic chloride (124 mg, 0.47 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was quenched by the addition of wateT (2 mL) and then it was concentrated under reduced pressure. The aqueous residue was extracted with diethyl ether (20 mL x 2) and the combined organic phase was washed with water (20 mL), brine (20 mL), dried over anhydrous sodium suifate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a yellow oil (90 mg, Yield 96%). R f = 0.9 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CD3OD) δ 7.22 (d, J = 8.4 Hz, 1 H), 7.07 (s, 1 H), 7.04 (d, J = 8.0 Hz, 1H), 6.34 (s, 1 H), 2.13-2.11 (m, 1H), 1.94 (m, 1 H), 1.66-1.64 (m, 2H), 1.36 (s, 3H), 1.29 (s, 3H), 1.25 (s, 3H), 1.12-1.09 (m, 1 H), 1.00-0.98 (m, 1 H); ppm; Mass spectrum (ESI +ve) m/z 297 (M + H + ). Example 133: (±)-(4aA?,9a/?)-1,1,4a-trimethyl-6-(trifluoromethoxy)-2,3,4 ,4a- tetrahydro-1 H-fluoren-9(9aH)-one

To a mixture of the product of Example 132 (80 mg, 0.27 mmol) and sodium bicarbonate (69 mg, 0.82 mmol) in dichloromethane (6 mL) at 0 °C was added metachloroperbenzoic acid (70 mg, 0.41 mmol) and the reaction was stirred for 30 minutes, warmed to room temperature and stirred for an additional for 2 hours. The reaction was cooled to 0 °C and saturated sodium carbonate (2 mL) was added to quench the reaction. The reaction was concentrated under reduced pressure and the aqueous residue extracted with diethyl ether (20 mL x 3). The combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was solvated in dichloromethane (6 mL), cooled to 0 °C and boron trifluoride etherate (0.1 mL, 0.788 mmol) was added. The reaction was stirred at 0 °C for 30 minutes, warmed to room temperature and stirred for an additional 2 hours. The mixture was cooled to 0 °C and water (2 mL) was added to quench the reaction. The reaction mixture was extracted with diethyl ether (20 mL x 3) and the combined organic phase was washed with water (10 mL x 2), saturated aqueous sodium bicarbonate (10 mL x 2), water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a colorless oil (30mg, Yield: 36%). R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.28-7.26 (m, 1 H), 7.09-7.08 (m, 1 H), 7.00 (s, 1 H), 3.07 (s, 1 H), 1.70-1.69 (m, 2H), 1.37-1.34 (m, 5H), 1.25-1.21 (m, 4H), 1.04 (s, 3H), 1.04-0.98 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 313 (M + H + ). Example 134: (±)-(4af?,9aS)-1 ,1 ,4a-trimethyl-6-(trifluoromethoxy)- 2,3,4,4a, 9,9a-hexahydro-1W-fluorene

A mixture of the product of Example 132 (30 mg, 0.1 mmol) and 20% Pd/C (6 mg, 20%) in methanol (2 mL) was stirred overnight at room temperature under hydrogen. The reaction mixture was filtered, and the solids washed with methanol followed by dichloromethane. The filtrate was dried over anhydrous sodium sulfate and then concentrated under reduced pressure to afford the title compound product as a colorless oil (25 mg, Yield: 84 %). R f = 0.6 (petroleum ether); 1 H NMR (400 MHz, CDC ) δ 7.19 (d, J = 8.4 Hz, 1 H), 6.97 (d, J = 8.0 Hz, 1 H), 6.93 (s, 1 H), 2.80-2.77 (m, 2 H), 1.93 (t, J = 10.0 Hz, 1 H), 1.71-1.60 (m, 1 H), 1.46-1.21 (m, 8 H), 1.14 (s, 3 H), 0.95 (s, 3 H) Mass spectrum (ESI +ve) m/z 299 (M + H + ). Example 135: (±)-6-fluoro-1 ,1 ,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluorene

To a stirred solution of the product of Example 56 (51.7 mg, 0.21 mmol) in dry dichloromethane (2 mL) at 0 °C was added stannic chloride (0.04 mL 0.32 mmol). The mixture was allowed to warm to room temperature and stirred for 2 hours. The reaction was cooled to 0 °C and quenched with water (20 mL). The aqueous layer was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether) to afford the title compound as a colorless liquid (40 mg, Yield: 83%). R f = 0.8 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.19 (dd, J = 8.0, 5.2 Hz, 1 H), 6.96 (d, J = 8.8 Hz, 1 H), 6.90 (t, J = 8.8 Hz, 1 H), 6.34 (s, 1 H), 2.13 (d, J = 12.8 Hz, 1 H), 2.03-1.93 (m, 1 H), 1 .69-1.63 (m, 2H), 1.38 (s, 3H), 1.31 (s, 3H), 1.26 (s, 3H), 1.13 (dt, J = 13.4, 4.0 Hz, 1 H), 1.02 (dt, J = 13.2, 3.6 Hz, H) ppm; Mass spectrum (El +ve) m/z 230 (M + ).

Example 136: (±)-(4aR,9aS)-6-fluoro-1 Λ ,4a-trimethyl-2, 3,4,4a, 9,9a- hexahydro-1 H-fluorene

To a solution of the product of Example 135 (24 mg, 0.1 mmol) in methanol (2 mL) was added 20% Pd/C (6 mg). The reaction was stirred under an atmosphere of hydrogen at room temperature overnight. The mixture was filtered and the solid washed with methanol. The organic phase was concentrated under reduced pressure. The residue was purified by Prep- HPLC to afford the title compound as a colorless oil (1 1 mg, Yield: 47%). R f = 0.9 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.10 (t, J = 6.4 Hz, 1 H), 6.80-6.75 (m, 2H), 2.76-2.71 (m, 2H), 1.89 (t, J = 9.4 Hz, 1 H), 1.63-1.56 (m, 1 H), 1.45-1.34 (m, 3H), 1.43 (s, 3H), 1.30-1.19 (m, 2H), 1.1 1 (s, 3H), 0.92 (s, 3H) ppm; Mass spectrum (El +ve) m/z 232 (M + ). Example 137: cyclohexyl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a stirred solution of cyclohexylmagnesium bromide (5 mL, 1 M in tetrahydrofuran, 5 mmol) cooled to -78 °C was slowly added a solution of 2,6,6-trimethylcyclohex-l-enecarbaldehyde (304 mg, 2.0 mmol) in tetrahydrofuran (4 mL). The reaction was then allowed to warm gradually to room temperature and stirred for 2 hours. The reaction was cooled to 0 °C and quenched with saturated aqueous ammonium chloride (10 mL). Water (45 mL) was added and the mixture was extracted with ethyl acetate (40 mL x 4). The combined organic phase was washed with brine (60 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 -> 40:1) afforded the title compound as a colorless oil (352 mg, Yield: 75%). R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, DMSO-d6) δ 4.24 (d, J = 4.0 Hz, H), 3.71 (bs, 1 H), 2.15 (d, J - 13.2 Hz, 1 H), 1.89-1.87 (m, 2H), 1.72-1.61 (m, 7H), 1.52-1.31 (m, 5H), 1.26-1.09 (m, 3H), 1.04 (s, 3H), 0.92 (s, 3H), 0.86-0.70 (m, 2H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 138: cyclohexyl(2,6,6-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 137 (85 mg, 0.382 mmol) in dichloromethane (5 mL) at 0 °C was added sodium bicarbonate (32 mg, 0.382 mmol) followed by Dess-Martin periodinane (324 mg, 0.764 mmol) at 0 °C. The resulting mixture was stirred at room temperature for 2 hours after which the reaction mixture was washed with saturated aqueous sodium bicarbonate (5 mL) and the organic layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (65 mg, Yield: 78%). R f = 0.7 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 3.07 - 2.96 (m, 1 H), 1.97 (t, J = 6.8 Hz, 2H), 1.92 - 1.85 (m, 2H), 1.80 - 1.71 (m, 4H), 1.70 - 1 .64 (m, 3H), 1.58-1.55 (m, 2H), 1.57 (s, 3H), 1.46 - 1.40 (m, 2H), 1.08 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 221 (M + H + ). Example 139: cyclopentyl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a solution of cyclopentylmagnesium bromide (2 M in tetrahydrofuran, 2.1 mL, 4.11 mmol) in tetrahydrofuran (3 mL) at 0 °C was added a the solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (250 mg, 1.64 mmol) in tetrahydrofuran (2 mL) dropwise over 5 minutes. The reaction mixture was allowed to stir at room temperature for 1.5 hours and then the reaction mixture was poured into aqueous saturated ammonium chloride solution (20 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (3 x 15 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (130 mg, Yield: 36%). R f = 0.3 (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, DMSO) δ 4.27 (d, J = 4 Hz, 1 H), 3.89-3.85 (m, 1 H), 2.35-2.25 (m, 1 H), 1.90-1.78 (m, 3H), 1.72 (s, 3H), 1.56-1.26 (m, 1 1 H), 1.05 (s, 3H), 0.96 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 205 (M - H 2 0 + H + ).

Example 140: cyclopentyl(2,6,6-trimethylcyclohex-1 -enyl)methanone

To a solution of the product of Example 139 (85 mg, 0.382 mmol) in dichloromethane (5 mL) at 0 °C was added sodium bicarbonate (32 mg, 0.382 mmol) followed by Dess-Martin periodinane (324 mg, 0.764 mmol). The resulting mixture was warmed to room temperature and stirred for 2 hours. The reaction mixture was washed with saturated aqueous sodium bicarbonate (5 mL) and the organic layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (65 mg, Yield: 78%). R f = 0.7 in (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 3.07 - 2.96 (m, 1H), 1.97 (t, J = 6.8 Hz, 2H), 1.92 - 1.85 (m, 2H), 1.80 - 1.71 (m, 4H), 1.70 - 1.64 (m, 3H), 1.58-1.55 (m, 2H), 1.57 (s, 3H), 1.46 - 1.40 (m, 2H), 1.08 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 221 (M + H + ). Example 141 : (±)-(4aR,9R,9aS)-1 ,1 ,4a-trimethyl-6-(trifluoromethoxy)- 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol

A solution of the product of Example 132 (200.0 mg, 0.68 mmol) in borane-tetrahydrofuran (1.0 M, 8.16 mL) was stirred at 30 °C for 48 hours. The reaction was cooled to 0 °C and 95% ethanol (5 mL), followed by a solution of sodium hydroxide (462.4 mg, 11.6 mmol) in water (9 mL) was added. After 15 minutes, 30% hydrogen peroxide (4.28 mL) was added slowly. The reaction was quenched by the addition of saturated aqueous ammonium chloride (25 mL) and the mixture then extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 -> 20:1) to afford the title compound as a white solid (165 mg, Yield: 77 %). Mp = 120-123 °C; R f = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.39 (d, J = 8.4 Hz, 1 H), 7.07 (d, J = 8.0 Hz, 1 H), 6.95 (s, 1 H), 5.02 (t, J = 7.8 Hz, 1 H), 1.77 (d, J = 7.6 Hz, 1 H), 1.70 (d, J = 9.2 Hz, 1 H), 1.67-1.55 (m, 2 H), 1.48 (s, 3 H), 1.45-1.35 (m, 3 H), 1.19 (s, 3 H), 1.16 (s, 3 H), 0.99 (dt, J = 13.6, 3.6 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 297 (M - H 2 0 + H + ).

Example 142: (+)-(4a ?,9aS)-1,1,4a-trimethyl-6-(trifluoromethoxy)-2,3,4,4a- tetrahydro-1W-fluoren-9(9aH)-one

To a solution of the product of Example 141 (310 mg, 0.99 mmol) in dichloromethane (5.5 mL) at 0 °C was added sodium bicarbonate (82.9 mg, 0.99 mmol) and Dess-Martin periodinane (836.4 mg, 1.98 mmol). The mixture was stirred at this temperature for 1 hour and then stirred at room temperature for 2 hours. Aqueous saturated aqueous sodium bicarbonate (50 mL) was added to the reaction mixture and then the mixture was extracted with dichloromethane. The organic phase was dried over anhydrous magnesium sulfate, filtered and then concentrated under reduced pressure, The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1) to afford the title compound as a white solid (260 mg, Yield: 84 %). Mp = 36-38 °C; R, = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.71 (d, J = 8.0 Hz, 1 H), 7.22 (s, 1 H), 7.17 (d, J = 8.4 Hz, 1 H), 2.22 (s, 1 H), 2.07-2.01 (m, 1 H), 1.73-1.62 (m, 2 H), 1.50-1.35 (m, 3 H), 1.29 (s, 3 H), 1.22 (s, 3 H), 0.65 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 313 (M + H + ).

Example 143: (±)-(4a ?,9S,9aS)-1,1,4a,9-tetramethyl-6-(trifluoromethoxy)- 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol

To a solution of the product of Example 142 (60 mg, 0.19 mmol) in dry tetrahydrofuran (4 ml_) under argon at -78 °C was added methyl lithium dropwise (0.13 ml_, 0.38 mmol). The solution was stirred at -78 °C for 2 hours. The reaction mixture was warmed to 0 °C and then quenched by the addition of saturated aqueous ammonium chloride (10 mL). The mixture was extracted with ethyl acetate and the organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 ) to afford the title compound as a white solid (46 mg, Yield: 74 %). Mp = 66-68 °C; R f - 0.45 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.33 (d, J = 8.0 Hz, 1 H), 7.08 (d, J = 8.0 Hz, 1 H), 6.99 (s, 1 H), 1.76 (s, 3H), 1.81-1.60 (m, 2 H), 1.54-1.52 (m, 1 H), 1.47 (s, 3 H), 1.37 (s, 3 H), 1.35-1.29 (m, 1 H), 1.27-1.24 (m, 2 H), 1.18 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 3 1 (M - H 2 0 + H + ).

Example 144: (±)-(4afi,9fl,9afiM ,1 ,4a,9-tetramethy l-6-(trifluoromethoxy)- 2,3,4,4a,9,9a-hexahydro-1tf-fluoren-9-ol

To a solution of methylmagnesium iodide (3.0 M in diethyl ether, 0.34 mL, 1.01 mmol) in dry tetrahydrofuran (2 mL) under an atmosphere of argon at -78 °C was added dropwise a solution of the product of Example 133 (70 mg, 0.22 mmol) in dry tetrahydrofuran (2 mL). The solution was stirred at -78 °C for 2 hours. The reaction mixture was warmed to 0 °C and quenched by the addition of saturated aqueous ammonium chloride ( 0 mL). The mixture was extracted with ethyl acetate and the organic phase dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 70:1 -> 60:1 ) to afford the title compound as a colorless oil (50 mg, Yield: 69 %). R f = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDC ) δ 7.17 (d, J = 8.0 Hz, 1 H), 6.93 (d, J = 8.0 Hz, 1 H), 6.83 (s, 1 H), 2.58 (s, 1 H), 2.03-1.91 (m, 2 H), 1.51 (s, 1 H), 1.43 (s, J = 13.6 Hz, 1 H), 1.32-1.24 (m, 2 H), 1.24 (s, 3 H), 1.20 (s, 3 H), 1.15 (s, 3 H), 0.94 (s, 3 H), 0.78-0.67 (m, 1 H) ppm; Mass spectrum (El +ve) m/z 328 (M + ).

Example 145: (±)-(4afl,9ft,9afl)-1 ,1 ,4a-trimethyl-6-(trifluoromethoxy)- 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol

To a solution of the product of Example 133 (40 mg, 0.13 mmol) in methanol (1.5 mL) at 0 °C was added sodium borohydride (14.5 mg, 0.38 mmol). The reaction was warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of water (2 mL) and the organic volatiles evaporated under reduced pressure. The aqueous mixture was extracted with ethyl acetate and the organic phase dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 50:1 -> 20:1) to afford the title compound as a colorless oil (28 mg, Yield: 68 %). R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCb) δ 7.19 (d, J = 8.0 Hz, 1 H), 6.97 (d, J = 8.4 Hz, 1 H), 6.70 (s, 1 H), 4.23 (t, J = 6.4 Hz, 1 H), 2.89 (d, J = 7.6 Hz, 1 H), 1.94-1.85 (m, 3 H), 1.49 (d, J = 13.6 Hz, 1 H), 1.34-1.27 (m, 2 H), 1.32 (s, 3 H), 1.25 (s, 3 H), 0.92 (s, 3 H), 0.76-0.67 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 297 (M - H 2 0 + H + ).

Example 146: (±)-(4aS,9aS)-6-fluoro-1 ,1 ,4a-trimethyl-2, 3,4,4a, 9,9a- hexahydro-1 H-carbazole

To a solution of the product of Example 102 (23.1 mg, 0.1 mmol) in methanol (1.0 ml) was added sodium borohydride (7.6 mg, 0.2 mmol) and the reaction was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure and then partitioned between water (20 mL) and ethyl acetate (60 mL). The organic phase was washed with water (20 ml), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a white solid (20 mg, Yield: 86%). Mp = 47-49 °C; R f = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 6.79 (d, J = 8.0 Hz, 1 H), 6.72 (t, J = 8.8 Hz, 1 H), 6.56- 6.53 (m, 1 H), 3.60-3.40 (m, 2 H), .85-1.66 (m, 3 H), 1 .48-1.30 (m, 2 H), 1.28 (s, 3 H), 1.28-1.22(m, 1 H), 0.88 (s, 3 H), 0.69 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 234 (M + H + ).

Example 147: (±)-(4a ?,9aR)-4,4,4a,9-tetramethyl-4,4a,9,9a-tetrahydro-1H- carbazol-2(3W)-one

Example 147a: 1 ,3-dimethyl-1W-indole

To a stirred solution of 3-methyl-1 /-/-indole (523 mg, 3.96 mmol) in anhydrous dimethylforamide (5 mL) was added sodium hydride (198 mg, 4.97 mmol). The mixture was stirred at room temperature for 30 minutes. The reaction was cooled to 0 °C and methyl iodide (0.7 mL, 11.46 mmol) was added in one portion. The mixture was stirred at room temperature for 5 hours. The mixture was concentrated under high vacuum to and the residue was taken up in ethyl acetate. The solution was washed with water (30 mL x 2), brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether) gave the title compound as a colorless oil (573 mg, Yield: 99%). R f = 0.9 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) 6 7.56 (d, J = 8.0 Hz, 1 H), 7.27 (d, J = 8.4 Hz, 1 H), 7.21 (t, J = 7.6 Hz, 1 H), 7.10 (t, J = 7.4 Hz, 1 H), 6.81 (s, 1 H), 3.72 (s, 3H), 2.32 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 146 (M + H + ).

Example 147: (±)-(4aR,9aR)-4,4,4a,9-tetramethy l-4,4a,9,9a-tetrahydro-1 H- carbazol-2(3H)-one

To a stirred solution of the product of Example 147a (210 mg, 1.45 mmol) in acetonitrile (2 mL) at 0 °C was added concentrated sulfuric acid (0.2 mL) and one minute later mesityl oxide (405 mg, 4.14 mmol) was added. The reaction was stirred at 0 °C for 30 minutes and then allowed to warm to room temperature and stirred for 1.5 hours. The mixture was then added to a suspension of sodium bicarbonate (1.3 g) in water (10 mL). The resulting mixture was extracted with diethyl ether (30 mL x 3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/dichloromethane = 0:1 - > 5:1) afforded the title compound as a pale yellow solid (133 mg, Yield:

38%). Mp = 92-94 °C; R f = 0.5 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.15 (t, J = 7.6 Hz, 1 H), 7.08 (d, J = 7.6 Hz, 1 H), 6.76 (t, J = 7.6 Hz, 1 H), 6.53 (d, J = 7.6 Hz, 1 H), 3.36 (dd, J = 5.2, 2.8 Hz, 1 H), 2.76 (dd, J = 17.2, 2.8 Hz, 1 H), 2.69 (dd, J = 17.2, 5.2 Hz, 1 H), 2.63 (s, 3H), 2.34 (d, J = 15.2 Hz, 1 H), 2.21 (d, J = 15.2 Hz, 1 H), 1.47 (s, 3H), 1.07 (s, 3H), 0.82 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 244 (M + H + ).

Example 148: (+)-(4aR,9aR)-4,4,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro- 1W-carbazole

To a solution of the product of Example 147 (51 mg, 0.21 mmol) in diethylene glycol (2 mL) was added solid potassium hydroxide (13 mg, 0.23 mmol) and 64% hydrazine hydrate (0.15 ml). The reaction was refluxed for 2.5 hours. The mixture was then slowly distilled until the temperature of the liquid reached 210 °C and refluxing was continued for 3 hours. The mixture was cooled and water was added. The solution was extracted with diethyl ether (30 mL x 3) and the combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether) gave the title compound as a colorless oil (22 mg, Yield: 46%). R f = 0.8 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.12 (t, J = 7.6 Hz, 1 H), 7.05 (d, J = 6.8 Hz, 1 H), 6.74 (t, J = 6.8 Hz, 1 H), 6.56 (d, J = 7.6 Hz, 1 H), 2.84 (d, J = 2.8 Hz, 1 H), 2.63 (s, 3H), 2.03 (d, J = 14.4 Hz, 1 H), 1.70-1.58 (m, 2H), 1.52-1.40 (m, 2H), 1.33 (s, 3H), 1.23 (d, J = 14.8 Hz, 1 H), 0.90 (s, 3H), 0.60 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 230 (M + H + ). Example 149: (±)-(4aA?,9a ?)-4,4,4a,9-tetramethyl-2,3,4,4a,9,9a-hexahydro- 1 Y-carbazol-2-ol

To a solution of the product of Example 147 (50 mg, 0.21 mmol) in methanol (5.0 mL) at 0 °C was added sodium borohydride (19 mg, 0.51 mmol). The reaction mixture was stirred at room temperature overnight after which time it was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (36.8 mg, Yield: 73%); R f = 0.4 (5:1 petroleum ether/ethyl acetate); 1 H NMR(400 MHz, CDCI 3 ) δ 7.16-7.12 (m, 2 H), 6.79 (t, J = 7.2 Hz, 1 H), 6.60 (d, J = 7.6 Hz, 1 H), 4.04 (m, 1 H), 3.05 (t, J = 4.0 Hz, 1 H), 2.74 (s, 3 H), 2.62 (br, s, 1 H), 2.10-1.96 (m, 2H), 1.70 (dd, J = 13.6, 4.8 Hz, 1 H), 1.62- 1.58 (m, 1 H), 1.30 (s, 3 H), 0.98 (s, 3 H), 0.87 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 246 (M + H + ). Example 150: (±)-(4aR,9aR)-4,4,4a,9-tetramethyl-2-methylene- 2,3,4,4a,9,9a-hexahydro-1H-carbazole

To a solution of the product of Example 147 (50 mg, 0.21 mmol) in tetrahydrofuran (2 mL) at 0 °C was added Tebbe's Reagent (0.41 mL of 0.5M, 0.21 mmol). The reaction mixture was warmed to room temperature and stirred for 1 hour. Additional Tebbe's Reagent (0.41 mL, 0.21 mmol) was added and stirring was continued for 1 hour. A final aliquot of Tebbe's Reagent (0.41 mL, 0.21 mmol) was added and stirring was continued at room temperature for 1 day. Diethyl ether (5 mL) was added and then methanol (10 drops) and the reaction mixture was stirred for 5 minutes. The mixture was vacuum filtered through celite and the solids washed with diethyl ether. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography to give the title compound as a yellow oil (21 mg, Yield: 43%). R, = 0.3 (80:1 petroleum ether/ethyl acetate); 1 H NMR(400 MHz, CDCI 3 ) δ 7.11 (t, J = 7.6 Hz, 1 H), 7.06 (d, J = 7.2 Hz, 1 H), 6.74 (t, J = 7.2 Hz, 1 H), 6.53 (d, J = 8.0 Hz, 1 H), 4.80 (s, 1 H), 4.68 (s, 1 H), 2.99 (d, J = 5.6 Hz, 1 H), 2.62 (d, J = 15.6 Hz, 1 H), 2.41 (dd, J, = 14.8 Hz, J 2 = 4.4 Hz, 1 H), 2.28 (d, J = 13.2 Hz, 1 H), 1.84 (d, J = 12.8 Hz, 1 H), 1.40 (s, 3 H), 0.96 (s, 3 H), 0.56 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 242 (M + H + ).

Example 151 : (±)-(4a ?,9 ?,9aS)-1 ,1 ,4a-trimethy l-6-(trifluoromethyl)- 2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 109 (60 mg, 0.2 mmol) in methanol (2 mL) at 0 °C was added sodium borohydride (23.0 mg, 0.61 mmol) in several portions. The reaction was warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of water (2 mL). The organic volatiles were evaporated under reduced pressure and the reaction mixture was extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 50:1 -> 20:1) to give the title compound as a white solid (50 mg, Yield: 84%). Mp = 92-94 °C; R f 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.49 (s, 2 H), 7.36 (s, 1 H), 5.05 (t, J = 8.4 Hz, 1 H), 1.85 (d, J = 7.6 Hz, 1 H), 1.70 (d, J = 8.4 Hz, 1 H), 1.68-1.57 (m, 2 H), 1.51 (s, 3 H), 1.48-1.36 (m, 3 H), 1.20 (s, 3 H), 1.17 (s, 3 H), 1.00-0.94 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 281 (M - H 2 0 + H + ).

Example 152: (±)-(4a ,9a/?)-2-methoxy-4,4,4a,9-tetramethyl-2,3,4,4a,9,9a- hexahydro-1/Y-carbazole

To a solution of the product of Example 149 (34.4 mg, 0.14 mmol) in tetrahydrofuran (2 mL) at 0 °C was added sodium hydride (7 mg, 0.175 mmol) and the mixture was warmed and stirred at room temperature for 30 minutes. Methyl iodide (0.026 mL, 0.42 mmol) was added and the reaction mixture was stirred at room temperature for 20 hours. Additional sodium hydride (7 mg, 0.175 mmol) and methyl iodide (0.026 mL, 0.42 mmol) were added and the reaction mixture was stirred at room temperature for 2 days. The reaction was quenched with water and then extracted with ethyl acetate. The combined organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a colorless oil (20 mg, Yield: 56%). R, = 0.5 (8:1 petroleum ether/ethyl acetate); H NMR(400 MHz, CDCI3+D2O) δ 7.15 (d, J - 7.2 Hz, 1 H), 7.08 (t, J = 7.2 Hz, 1 H), 6.65 (t, J = 7.2 Hz, 1 H), 6.44 (d, J = 7.6 Hz, 1 H), 3.51-3.48 (m, 1 H), 3.32 (s, 3 H), 3.20 (t, J = 7.2 Hz, 1 H), 2.72 (s, 3 H), 2.19-2.15 (m, 1 H), 1.73 (dd, J 1 = 13.2 Hz, J2 = 5.2 Hz,1 H), 1.61-1.57 (m, 1 H), 1.51-1.45 (m, 1 H), 1 .24 (s, 3 H), 1.08 (s, 3 H), 1.01 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 260 (M + H + ).

Example 153: (3,3-dimethylcyclohex-1-enyl)(4-fluorophenyl)methanol

Example 153a: 2-(hydroxymethylene)-6-methylcyclohexanone

To an ice cold suspension of powdered sodium methoxide (24.5 g, 454 mmol) in toluene (450 mL) was added 2-methylcyclohexanone (60 g, 178 mmol) and ethyl formate (79.2 g, 1069 mmol). The mixture was stirred at room temperature overnight. Ice water and toluene were added and the phases were separated. The organic phase was washed with 10% sodium hydroxide (100 mL x 2). The aqueous layer was acidified with dilute hydrochloric acid to pH -3 and then extracted with diethyl ether (200 mL x 3). The combined organic phase was washed with water (100 mL x 2), brine (100 mL x 2) and dried over anhydrous sodium sulfate and concentration under reduced pressure gave the title compound as a light orange oil (50 g, Yield: 66%).

Example 153b: 2-(isopropoxymethylene)-6-methylcyclohexanone

To a solution of the product of Example 153a (50 g, 357 mmol) in acetone (500 mL) was added potassium carbonate (74.1 g, 536 mmol) and 2- iodopropane (45 ml, 446 mmol) and the reaction mixture was refluxed overnight. After concentration under reduced pressure the residue was diluted with ether (800 mL) and the organic layer was washed with 5% aqueous sodium hydroxide (100 mL x 2), brine (100 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound as a light yellow oil (55 g, Yield: 84%), which was used in the next step without any further purification. Example 153c: (E)-6-(tert-butoxymethylene)-2,2-dimethylcyclohexanone and (E)-6-(isopropoxymethylene)-2,2-dimethylcyclohexanone

To a solution of potassium tert-butoxide (106 g, 945 mmol) in tetrahydrofuran (550 mL) cooled to 0 °C was added the product of Example 153b (55 g, 302 mmol). The mixture was stirred at 0 °C for 10 minutes and then methyl iodide (141 g, 993 mmol) was added. The mixture began to reflux and when reflux ceased the cooling bath was removed and the mixture was then stirred at room temperature for 1 hour. The mixture was filtered and then concentrated under reduced pressure. The residue was diluted with diethyl ether (600 mL), washed with 10% aqueous sodium hydroxide (100 mL x 2), brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 45 g of an orange oil. Purification of 6.0 g of this material by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1 :0 -> 30:1) gave a 4:1 mixture of (E)-6-(tert-butoxymethylene)-2,2- dimethylcyclohexanone and (£)-6-(isopropoxymethylene)-2,2- dimethylcyclohexanone as a colorless oil (3.52) H NMR (400 MHz, CDCI 3 ) Major: δ 7.58 (t, J = 2.0 Hz, 1 H), 2.41-2.39 (m, 2H), 1.70-1.66 (m, 4H), 1.35 (s, 9H), 1.12 (s, 6H); Minor: δ 7.37 (t, J = 1.8 Hz, 1 H), 4.23-4.15 (m, 1 H), 2.41-2.39 (m, 2H), 1.70-1.66 (m, 4H), 1.29 (d, J = 6.4 Hz, 6H), 1.12 (s, 6H).

Example 153d: 3,3-dimethylcyclohex-1-enecarbaldehyde

To a stirred solution of a mixture of the product of Example 153c (2.10 g, 10.0 mmol) in dry diethyl ether (50 mL) at -20 °C was added lithium aluminum hydride (0.57 g, 15.0 mmol) portionwise. The resulting mixture was stirred for 1 hour during which time the reaction temperature was allowed to warm to room temperature. The reaction was diluted with ethyl acetate (5 mL) and then saturated aqueous ammonium chloride (5 mL) was added and the mixture was further diluted with diethyl ether (300 mL). The organic phase was dried over anhydrous magnesium sulfate, filtered and then concentrated under reduced pressure to give a colorless liquid. The material was dissolved in acetone (20 mL) and 2N HCI (0.05 mL) was added and the solution was shaken for 1 minute. Solid sodium bicarbonate (50 mg) was added and the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography give the title compound as a colorless liquid (1.15 g, Yield: 83%). 1 H NMR (400 MHz, CDCI 3 ) δ 9.39 (s, 1 H), 6.47 (s, 1 H), 2.14 (t, J = 6.0 Hz, 2H), 1.69-1.63 (m, 2H), 1.53-1.50 (m, 2H), 1.10 (s, 6H).

Example 153: (3,3-dimethylcyclohex-1-enyl)(4-fluorophenyl)methanol

To a 25 mL three necked round bottom flask was added (4- fluorophenyl)magnesium bromide (8.2 ml of 0.8 M in tetrahydrofuran, 6.53 mmol). The solution was cooled to -78 °C and a solution of the product of Example 153d (360 mg, 2.61 mmol) in tetrahydrofuran (5 mL) was added. The resulting mixture was stirred at -78 °C for 15 minutes, after which time the reaction was warmed to room temperature and stirred for 1 hour. The mixture was cooled to 0 °C and saturated aqueous ammonium chloride (30 mL) was added. The resulting mixture was extracted with ethyl acetate (25 mL x 4) and the combined organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 50:1) gave the title compound as a colorless oil (490 mg, Yield: 80%). R, = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.31 (dd, J = 8.0, 6.0 Hz, 2H), 7.01 (t, J = 8.6 Hz, 2H), 5.61 (s, 1H), 5.04 (s, 1 H), 1.83-1.77 (m, 1 H), 1.68- 1.54 (m, 3H), 1.45-1.34 (m, 2H), 1.02 (s, 3H), 1.01 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 2M (M - H 2 0 + H + ). Example 154: (3,3-dimethylcyclohex-1-enyl)(4-fluorophenyl)methanone

To a stirred solution of the product of Example 153 (180 mg, 0.77 mmol) in dichloromethane (8 mL) at 0 °C was added Dess-Martin periodinane (587 mg, 1.38 mmol). The reaction was stirred at room temperature for 1 hour. The mixture was then concentrated under reduced pressure and the residue dispersed in petroleum ether, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography affording a light yellow oil (152 mg) which was then purified by preparative HPLC affording the title compound as light yellow oil (120 mg, Yield: 67%). = 0.6 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.67 (dd, J = 8.0, 6.0 Hz, 2H), 7.11 (t, J = 8.4 Hz, 2H), 6.20 (s, 1 H), 2.35 (t, J = 6.2 Hz, 2H), 1.77-1.71 (m, 2H), 1.52 (t, J = 5.8 Hz, 2H), 1.07 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 255 (M + Na + ).

Example 155: (±)-(4aS,9aS)-6-fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro- H- fluoren-9(9aH)-one

The product of Example 154 (218 mg, 0.94 mmol) and methansulfonic acid (12 mL) were mixed and the resulting solution was stirred at 80 °C for 6 hours. The mixture was cooled and ethyl acetate (150 mL) was added and the organic phase was washed with water (80 mL x 2), saturated aqueous sodium bicarbonate (80 mL), brine (80 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a light yellow solid (72 mg, Yield: 33%). Mp = 49.3-50.3 °C; R, = 0.4 (30:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.76 (dd, J = 8.0, 5.6 Hz, 1 H), 7.18 (d, J = 8.4 Hz, 1 H), 7.07 (t, J = 8.8 Hz, 1 H), 3.16 (d, J = 7.2 Hz, 1 H), 2.70 (t, J = 7.6 Hz, 1 H), 2.44 (d, J = 13.6 Hz, 1 H), 1.56-1.34 (m, 5 H), 1.16 (s, 3 H), 0.13 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 233 (M + H + ).

Example 156: (4-fluorophenyl)(2,3,3-trimethylcyclohex-1 -enyl)methanol

Example 156a: 6-(dimethoxymethyl)-2,2-dimethylcyclohexanone

To a solution of the product of Example 153c (3.0 g, 14.3 mmol) in methanol (60 mL) at 0 °C was added ethanol (500 mg, 10.8 mmol) and acetyl chloride (2.0 mL, 28.6 mmol). The mixture was stirred for 4 hours at room temperature. The mixture was concentrated under reduced pressure and then diluted with ethyl acetate (200 mL). The organic layer was washed with water (100 mL), brine (10 0 mL x 3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) gave the title compound as light yellow oil (830 mg, Yield: 29%). R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 4.65 (d, J = 5.2 Hz, 1 H), 3.45 (s, 3H), 3.44 (s, 3H), 2.90-2.84 (m, 1 H), 2.16 (d, J = 13.2 Hz, 1 H), 1.85-1.74 (m, 3H), 1.56-1.48 (m, 2H), 1.19 (s, 3H), 1.05 (s, 3H).

Example 156b: 6-(dimethoxymethyl)-1,2,2-trimethylcyclohexanol

To a stirred solution of methyl lithium (2.74 mL, 3M in diethoxymethane) at 0 °C was added dropwise a solution of the product of Example 56a (823 mg, 4.1 1 mmol) in diethyl ether (10 mL). The mixture was warmed to room temperature and stirred for 1.5 hours. The reaction mixture was cooled to 0 °C and 0. N HCI (70 mL) was added slowly. The resulting mixture was extracted with diethyl ether (150 mL) and the organic layer was washed with water (100 mL x 2), brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 - > 20:1) gave the title compound as light yellow oil (588 mg, Yield: 66%). R, = 0.5 (20. petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) Major, δ 4.34 (d, J = 8.4 Hz, 1 H), 4.13 (s, 1 H), 3.42 (s, 3H), 3.32 (s, 3H), 2.11-2.07 (m, 1 H), 1.88-1.72 (m, 1 H), 1.60-1.49 (m, 2H), 1.45-1.20 (m, 2H), 1.09-1.02 (m, 1H), 1.13 (s, 3H), 1.02 (s, 3H), 0.93 (s, 3H); Minor, δ 4.47 (d, J = 2.4 Hz, 1 H), 3.55 (s, 1 H), 3.47 (s, 3H), 3.44 (s, 3H), 1.88-1.72 (m, 2H), 1.60-1.49 (m, 2H), 1.45-1.20 (m, 2H), 1.09-1.02 (m, 1 H), 1.19 (s, 3H), 0.95 (s, 3H), 0.90 (s, 3H).

Example 156c: 2,3,3-trimethylcyclohex-l-enecarbaldehyde

A solution of the product of Example 156b (620 mg, 2.87 mmol) in hydrochloric acid/acetone (0.17 M, 10 mL) was refluxed for 3 hours. The reaction was cooled to room temperature and the mixture was extracted with diethyl ether (150 mL). The organic layer was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound as a yellow oil (420 mg, Yield: 96%). R f = 0.7 in (30:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 10.16 (s, 1 H), 2.18-2.16 (m, 2H), 2.13 (s, 3H), 1.61-1.60 (m, 2H), 1.52-1.50 (m, 2H), 1.12 (s, 6H). Example 156: (4-fluorophenyl)(2,3,3-trimethylcyclohex-1-enyl)methanol

To a 25 ml three necked round bottom flask was added (4- fluorophenyl)magnesium bromide (0.8 M in tetrahydrofuran, 6.16 mL, 4.92 mmol). The solution was cooled to -78 °C and a solution of the product of Example 156c (300 mg, 1.97 mmol) in tetrahydrofuran (5 mL) was added and the resulting mixture was stirred at -78 °C for 15 minutes, after which time it was allowed to warm to room temperature and stirred for 2 hours. The mixture was cooled to 0 °C, saturated aqueous ammonium chloride (30 mL) was added and the resulting mixture was extracted with ethyl acetate (25 mL x 4). The combined organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 40:1) afforded the title compound as a colorless oil (446 mg, Yield: 81%.) = 0.3 in (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.28 (dd, J = 8.4, 6.0 Hz, 2H), 7.01 (t, J = 8.4 Hz, 2H), 5.73 (s, 1 H), 2.06-2.03 (m, 1H), 1.77 (s, 3H), 1.60- 1.38 (m, 5H), 1.06 (s, 3H), 1.05 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ). Example 157: (4-fluorophenyl)(2,3,3-trimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 156 (270 mg, 1.08 mmol) in dichloromethane (5 mL) at 0 °C at was added Dess-Martin periodinane (824 mg, 1.94 mmol). The mixture was stirred for at room temperature for 2 hours. The mixture was diluted with dichloromethane (100 mL) and the organic layer was washed with saturated aqueous sodium bicarbonate (50 mL x 3), brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification by Prep-HPLC afforded the title compound as white solid (155 mg, Yield: 58%). Mp = 48.7-50.1 °C; R, = 0.5 in (50:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.92 (dd, J = 8.0, 6.0 Hz, 2H), 7.13 (t, J = 8.2 Hz, 2H), 2.16 (br, 2H), 1.74-1.73 (m, 2H), 1.62- 1.59 (m, 2H), 1.50 (s, 3H), 1.12 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 158: (±)-(4aS,9aS)-6-fluoro-4,4,4a-trimethyl-2,3,4,4a-tetrahydro - 1 H-fluoren-9(9aH)-one

A mixture of the product of Example 157 (121 mg, 0.49 mmol) in methanesulfonic acid (3 mL) was stirred at 50 °C overnight. Water (20 mL) was added and the mixture was extracted with ethyl acetate (50 mL) and the combined organic layer was washed by water (30 mL x 2), brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography gave the title compound as a white solid (10 mg, Yield: 8%). Mp = 75-78.8 °C; R f = 0.5 (40:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.75 (dd, J = 8.0, 5.6 Hz, 1 H), 7.19 (d, J = 8.8 Hz, 1 H), 7.05 (t, J = 8.4 Hz, 1 H), 2.42-2.35 (m, 2H), 1.63-1.41 (m, 4H), 1.52 (s, 3H), 1.25-1.20 (m, 1 H), 1.01 (s, 3H), 0.12 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 159: (±)-(4aR,9R,9aS)-1 ,1 ,4a-trimethyl-6-(trifluoromethyl)- 2,3,4,4a,9,9a-hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 109 (60 mg, 0.2 mmol) in methanol (2 mL) at 0 °C was added sodium borohydride (23.0 mg, 0.61 mmol) in several portions. The reaction was warmed and stirred at room temperature for 2 hours. The reaction was quenched by the addition of water (2 mL) and the organic volatiles were evaporated under reduced pressure. The mixture was extracted with ethyl acetate and the organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 - >20:1) to afford the title compound as a white solid (50 mg, Yield 84%). Mp = 92-94 °C; R f = 0.2 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.49 (s, 2 H), 7.36 (s, 1 H), 5.05 (t, J = 8.4 Hz, 1 H), 1.85 (d, J = 7.6 Hz, 1 H), 1.70 (d, J = 8.4 Hz, 1 H), 1.68-1.57 (m, 2 H), 1.51 (s, 3 H), 1.48-1.36 (m, 3 H), 1.20 (s, 3 H), 1 .17 (s, 3 H), 1.00-0.94 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 281 (M - H 2 0 + H + ). Example 160: (2,6,6-trimethylcyclohex-1-enyl)methyl)benzene

Into a reaction flask containing a stirred mixture of 280 mg of lithium (280 mg) in anhydrous diethyl ether (10 mL) was slowly added a solution of bromobenzene (790 mg (5.0 mmol) in diethyl ether (7 mL). After 1 hour a solution of 2,6,6-trimethylcyclohex-1-enecarbaldehyde (380 mg, 2.5 mmol) in 8 mL diethyl ether (8 mL) was slowly added and the mixture was stirred for an additional 1 hour. Ammonia (ca. 25 ml) was carefully distilled into the mixture and, once the dark blue color of the mixture was established, saturated aqueous ammonium chloride was cautiously added to discharge the blue color and the ammonia was allowed to evaporate. After the residue had been partitioned between brine and diethyl ether, the organic phase was dried over anhydrous magnesium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (380 mg, Yield: 71%). R f = 1.0 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.27-7.22 (m, 2H), 7.16-7.13 (m, 3H), 3.47 (s, 2H), 2.05 (t, J = 6.0 Hz, 2H), 1.68-1.65 (m, 2H), 1.54 (s, 3H), 1.48 (t, J = 6.0 Hz, 2H), 0.91 (s, 6H) ppm; Mass spectrum (APCI +ve) m/z 214 (M + ).

Example 161 : (+)-6-fluoro-1 ,1,4a-trimethyl-2,3,4,4a-tetrahydro-1H- fluorene

To a solution of the product of Example 56 (50 mg, 0.20 mmol) in dry dichloromethane (3 mL) at 0 °C was added stannic chloride (35 μΐ, 0.30 mmol). The mixture was stirred for 5 minutes and then allowed to warm to room temperature and stirred for an additional 1.5 hours. The reaction was cooled to 0 °C and quenched with water (1 mL). The mixture was extracted with dichloromethane (20 mL x 2). The combined organic phase was washed with water (30 mL x 2), brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography (eluent: petroleum ether) gave the title compound as a colorless oil (4.5 mg, Yield: 10%). R f = 0.8 (petroleum ether); 1 H NMR (400 MHz, CDCI 3 ) δ 7.15-7.09 (m, 2H), 6.89 (t, J = 8.8 Hz, 1 H), 6.28 (s, 1 H), 2.61-2.57 (m, 1 H), 2.27-2.22 (m, 1 H), 1.78-1.74 (m, 2H), 1.50-1.48 (m, 1 H), 1.24 (s, 3H), 1.20 (s, 3H), 1.24-1.20 (m, 1 H), 0.35 (s, 3H) ppm.

Example 162: (±)-(4a ?,9 ?,9aS)-6-fluoro-1,1,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

A solution of the product of Example 161 (1 1 mg, 0.05 mmol) in borane tetrahydrofuran (1 ml_, 1.0 M) was stirred at room temperature under argon for 2 days. The mixture was cooled to 0 °C and 95% ethanol (1 ml_) was added to quench the remaining borane. A solution of aqueous sodium hydroxide (1.42 N, 0.6 ml_) and hydrogen peroxide (0.3 mL, 8.45 mmol) were added slowly and the mixture was warmed to room temperature and stirred for 2 hours. The reaction was quenched with saturated aqueous ammonium chloride (5 mL) and then the mixture was extracted with ethyl acetate (30 mL). The organic phase was washed with water (30 mL x 2), brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50: 1 - >20:1) afforded the title compound as a colorless oil (5.6 mg, Yield: 45%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.34 (t, J = 6.8 Hz, 1 H), 6.94 (t, J = 8.6 Hz, 1 H), 6.88 (d, J = 9.2 Hz, 1 H), 5.00 (d, J = 8.8 Hz, 1 H), 2.02 (d, J = 13.6 Hz, 1 H), 1.96-1.92 (m, 1 H), 1.68-1.55 (m, 4H), 1 .36 (s, 3H), 1.25- 1.22 (m, 1 H), 0.94 (s, 3H), 0.34 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ).

Example 163: (±)-(4a ,9aR)-4,4,9-trimethyl-4a-propyl-4,4a,9,9a- tetrahydro-1 H-carbazol-2(3W)-one

Example 163a: 1-methyl-3-propyl-1H-indole

To a solution of pentanal (64 mg, 0.74 mmol) in acetic acid (1.2 mL) was slowly added 1-methyl-1-phenylhydrazine (500 mg, 4.01 mmol) and the reaction mixture was placed in microwave reactor at 200 °C for 1 minute. The reaction was cooled to room temperature and additional pentanal (64 mg, 0.74 mmol) was added. The mixture was placed in a microwave reactor at 200 °C for 1 hour. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was partitioned between saturated aqueous sodium bicarbonate and ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography gave the title compound as a colorless oil (127 mg, Yield: 49%). R f = 0.5 (100:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.59 (d, J = 8.0 Hz, 1 H), 7.29-7.25 (m, 1 H), 7.20 (t, J = 7.6 Hz, 1 H), 7.08 (t, J = 7.4 Hz, 1 H), 6.82 (s, 1 H), 3.74 (s, 3H), 2.72 (t, J = 7.6 Hz, 2H), 1.75-1.69 (m, 2H), 0.99 (t, J = 7.6 Hz, 3H) ppm; Mass spectrum (ESI +ve) /77/z 174 (M + H + ). Example 163: (±M4a/?,9a/?MA9-trimethyMa^ropyl-4,4a,9,9a- tetrahydro-1 V-carbazo 1-2(3/-/) -one

To a stirred solution of the product of Example 163a (127 mg, 0.73 mmol) in acetonitrile (1.3 ml_) at 0 °C was added concentrated sulfuric acid (0.13 mL) followed by mesityl oxide (201 mg, 2.05 mmol). The reaction was stirred at 0 °C for 30 minutes after which time the temperature was allowed to rise to room temperature and stirring was continued overnight. The reaction mixture was added slowly with cooling and stirring to a suspension of sodium bicarbonate (1.0 g) in water (10 mL). The solution was extracted with diethyl ether (30 mL x 3) and the combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography gave the title compound as a light yellow solid (138 mg, Yield: 69%). Mp =

1 10.2-1 12.1 °C; R f = 0.5 (5:1 petroleum ether/ethyl acetate); 1 H NMR(400 MHz, CDCI 3 ) δ 7.12 (t, J = 7.6 Hz, 1 H), 6.97 (d, J = 7.2 Hz, 1 H), 6.72 (t, J = 7.6 Hz, 1 H), 6.49 (d, J = 7.6 Hz, 1 H), 3.55 (d, J = 3.2 Hz, 1 H), 2.72-2.69 (m, 2H), 2.62 (s, 3H), 2.21 (s, 2H), 1.93-1.89 (m, 1 H), 1.61 -1.55 (m, 1 H), 1.15- 1.10 (m, 1 H), 1.09 (s, 3H), 1.00-0.96 (m, 1 H), 0.90 (t, J = 7.2 Hz, 3H), 0.88 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 272 (M + H + ).

Example 164: (±)-((4aA?,9a ?)-4,4,4a,9-tetramethyl-2,3,4,4a,9,9a- hexahydro-1 Y-carbazol-2-yl)methanol

A mixture of the product of Example 150 (125 mg, 0.52 mmol) and 1.0 M borane tetrahydrofuran (4 mL, 4 mmol) under argon was stirred at -78 °C for 10 minutes and then the temperature was raised to room temperature and stirring was continued for 20 hours. The mixture was cooled to 0 °C, 95% ethanol was added to quench the reaction, and a solution of sodium hydroxide (337 mg) in water (6 mL) along with 30% hydrogen peroxide (3.15 mL) were added slowly with stirring. After stirring for 2 hours at room temperature, the mixture was poured into saturated aqueous ammonium chloride. The reaction mixture was extracted with diethyl ether and the organic phase was washed with water, brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography followed by preparative HPLC to afford the title compound as a light yellow oil (32 mg, Yield: 48%). R f = 0.5 (5:2 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.14-7.09 (m, 2 H), 6.71 (t, J = 7.4 Hz, 1 H), 6.52 (d, J = 7.6 Hz, 1 H), 3.53-3.42 (m, 2 H), 3.09 (t, J = 5.4 Hz, 1 H), 2.70 (s, 3 H), 2.62 (br, s, 1 H), 2.15-2.10 (m, 1 H), 2.05-2.01 (m, 1 H), 1.50- 1.45 (m, 1 H), 1.30-1.28 (m, 2 H), 1.26 (s, 3 H), 1.09 (s, 3 H), 1.02 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 260 (M + Η +

Example 165: (±)-(4a ?,9aR)-2-(methoxymethyl)-4,4,4a,9-tetramethyl- 2, 3,4,4a, 9,9a-hexahydro-1H-carbazole

To the solution of the product of Example 164 (50 mg, 0.19 mmol) in tetrahydrofuran (2 mL) at 0 °C was added sodium hydride (8 mg, 0.24 mmol) and the reaction was warmed to room temperature and stirred for 30 minutes. The reaction was recooled to 0 °C, methyl iodide (80 mg, 0.57 mmol) was added and then it was allowed to warm to room temperature and stirred for 2 days. The reaction was added to water and then extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as light yellow oil (20 mg, Yield: 38%). R f = 0.4 (30:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.17 (d, J = 7.2 Hz, 1 H), 7.08 (t, J = 7.6 Hz, 1 H), 6.63 (t, J = 7.4 Hz, 1 H), 6.41 (d, J = 7.6 Hz, 1 H), 3.32 (s, 3 H), 3.24-3.12 (m, 3 H), 2.71 (s, 3 H), 2.04-1.96 (m, 2 H), 1.42 (dd, J, = 3.2 Hz, J 2 = 4.4 Hz, 1 H), 1.23 (s, 3 H), 1.23-1.11 (m, 2 H), 1.09 (s, 3 H), 1.05 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 274 (M + H + ).

Example 166: (±)-(4aS,9S,9aS)-6-fluoro-4,4-dimethyl-2,3,4,4a,9,9a- hexahydro-1 -fluoren-9-ol

To a stirred solution of the product of Example 155 (20 mg, 0.086 mmol) in methanol (2 mL) at 0 °C was added sodium borohydride (8 mg, 0.22 mmol). The resulting mixture was allowed to warm to room temperature and stirred overnight. The mixture was concentrated under reduced pressure and the residue was purified by preparative thin layer chromatography to give the title compound as a colorless oil (17 mg, Yield: 85%). R f = 0.5 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.32 (t, J = 6.6 Hz, 1 H), 7.19 (d, J = 9.2 Hz, 1 H), 6.91 (t, J = 8.6 Hz, 1 H), 4.96 (t, J = 7.2 Hz, 1 H), 2.78 (d, J = 6.0 Hz, 1 H), 2.70-2.67 (m, 1 H), 1.68 (d, J = 7.6 Hz, 1 H), 1.64- 1.60 (m, 1 H), 1.55-1.50 (m, 1 H), 1.37-1.26 (m, 3 H), 1.17-1.14 (m, 7 H) ppm; Mass spectrum (ESI +ve) m/z 217 (M - H 2 0 + H + ). Example 167: (+)-(4a/?,9S,9aS)-6-fluoro-1,1,4a,9-tetramethyl-2,3,4,4a,9,9 a- hexahydro-1 W-fluoren-9-ol

To a stirred solution of the product of Example 58 (31 mg, 0.13 mmol) in anhydrous tetrahydrofuran (2 mL) at -78 °C was slowly added methyl lithium (0.26 mmol). The reaction was stirred at -78 °C for 1 hour and then the reaction was allowed to warm to room temperature and stirred for 3 hours. The mixture was cooled to -78 °C and additional methyl lithium (0.13 mmol) was added and the reaction was stirred at room temperature for 1 more hour. Saturated aqueous ammonium chloride (30 mL) was added to quench the reaction and the mixture was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) to afford the title compound as a light yellow solid. (9 mg, Yield: 26%). Mp = 88-90 °C; R f = 0.3 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.30-7.26 (m, 1 H), 6.92 (t, J = 9.0 Hz, 1 H), 6.84 (d, J = 9.2 Hz, 1 H), 1.77-1.64 (m, 7H), 1.57-1.53 (m, 1 H), 1.45 (s, 3H), 1.36 (s, 3H), 1.34-1.23 (m, 2H), 1.15 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 245 (M - H 2 0 + H + ).

Example 168: (±)-(4a ?,9aA?)-6-fluoro-1,1,4a-trimethyl-2,3,4,4a-tetrahydro- 1 W-fluoren-9(9aH)-one

A stirred solution of the product of Example 135 (280 mg, 1.22 mmol) and sodium bicarbonate (3.8 mg, 3.66 mmol) in dry dichloromethane (10 mL) cooled to 0 °C was added metachloroperbenzoic acid (367 mg, 1.83 mmol). After 25 minutes, the reaction was allowed to warm to room temperature and stirred 1.5 hours. The reaction was quenched by adding saturated aqueous sodium bicarbonate solution. The solution was concentrated under reduced pressure and water (20 mL) was added. The aqueous layer was extracted with diethyl ether (50 mL x 3) and the combined organic phase was washed with saturated aqueous sodium bicarbonate (30 mL x 2), brine (30 ml x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was dissolved in anhydrous dichloromethane (10 mL) and cooled to 0 °C. Boron trifluoride etherate (0.47 ml, 3.66 mmol) was added and after 40 minutes the reaction was allowed to warm to room temperature and stirred overnight. Water was added to quench the reaction and then the organic volatiles were evaporated under reduced pressure. Water (20 mL) was added and the aqueous layer was extracted with diethyl ether (50 mL x 3). The organic layer washed with saturated aqueous sodium bicarbonate (20 mL x 2), brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 200:1) to give the semi pure title compound (270 mg) as a pale yellow solid. The material (50 mg) was purified again by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 30:1) to afford the title compound as a light yellow solid (46 mg, Yield: 83%). Mp = 80-83 °C; R f = 0.7 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.21 (dd, J = 7.8, 5.4 Hz, 1 H), 6.92 (t, J - 8.8 Hz, 1 H), 6.86 (d, J = 8.8 Hz, 1 H), 3.05 (s, 1 H), 1.72-1.63 (m, 2H), 1.39-1.31 (m, 5H), 1.26-1.18 (m, 4H), 1.03 (s, 3H), 1.01-0.95 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 169: (±)-(4a ?,9/?,9a )-6-fluoro-1,1,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 W-fluoren-9-ol

A stirred solution of the product of Example 168 (63 mg, 0.26 mmol) in methanol (4 mL) and tetrahydrofuran (2 mL) was added sodium borohydride (79 mg, 2.08 mmol). The reaction was stirred at room temperature for 2 days. Water (20 mL) was added and the mixture was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 100:1) afforded the title compound as a white solid (53 mg, Yield: 82%). Mp = 43-46 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.13 (t, J = 6.6 Hz, 1 H), 6.82 (t, J = 8.8 Hz, 1 H), 6.76 (d, J = 8.8 Hz, 1 H), 4.23 (t, J = 6.8 Hz, 1 H), 2.86 (d, J - 7.6 Hz, 1 H), 1.92-1.82 (m, 3H), 1.48 (d, J = 14.0 Hz, 1 H), 1.33-1.25 (m, 2H), 1.31 (s, 3H), 1.23 (s, 3H), 0.91 (s, 3H), 0.79-0.69 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ).

Example 170: (+)-(4aR,9R,9aR)-6-fluoro-1 ,1 ,4a,9-tetramethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

A stirred solution of the product of Example 169 (60 mg, 0.24 mmol) in anhydrous tetrahydrofuran (4 mL) cooled to -78 °C was slowly added methyl lithium (0.48 mmol). The reaction was stirred at -78 °C for 1.5 hours and then saturated aqueous ammonium chloride (30 mL) was added to quench the reaction. Water (20 mL) was added and the mixture was extracted with ethyl acetate (50 mL x 3) and the combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 200:1 ) to afford the title compound as a white solid (61 mg, Yield: 97%). Mp = 35-37 °C; R f = 0.5 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.10 (dd, J - 8.0, 5.6 Hz, 1 H), 6.77 (t, J = 8.6 Hz, 1 H), 6.69 (d, J = 7.6 Hz, 1 H), 2.55 (s, 1 H), 2.02-1.91 (m, 2H), 1.50 (s, 1 H), 1.43 (d, J = 14.0 Hz, 1 H), 1.32-1.26 (m, 2H), 1.23 (s, 3H), 1.19 (s, 3H), 1.15 (s, 3H), 0.93 (s, 3H), 0.81-0.70 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 245 (M - H 2 0 + H + ).

Example 171 : (±)-(4af?,9af?)-2,4,4,4a,9-pentamethyl-2, 3,4,4a, 9,9a- hexahydro-1 W-carbazol-2-ol

To a solution of the product of Example 147 (60 mg, 0.25 mmol) in tetrahydrofuran (2 mL) at -78 °C was added methyl magnesium bromide (3.0 M, 0.21 mL, 0.62 mmol) and the reaction was then warmed to room temperature and stirred overnight. Additional methyl magnesium bromide (3.0 M, 2.1 mL, 6.2 mmol) was added and the mixture was stirred at 30 °C for 6 hours and then at room temperature overnight. Saturated aqueous ammonium chloride was slowly added into the reaction mixture and the mixture was then extracted with ethyl acetate. The combined organic phase was washed brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a light yellow solid (28 mg, Yield: 45%). Mp = 98.8-100.8 °C; R f = 0.5 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.17 (t, J = 7.2 Hz, 1 H), 7.1 1 (d, J = 7.2 Hz, 1 H), 6.86 (t, J = 7.2 Hz, 1 H), 6.69 (d, J = 8.0 Hz, 1 H), 3.98 (s, 1 H), 2.98 (s, H), 2.72 (s, 3H), 2.19 (d, J = 15.2 Hz, 1 H), 1.69 (dd, J = 15.2, 3.6 Hz, 1 H), 1.57 (s, 2H), 1.33 (s, 3H), 1 .24 (s, 3H), 0.93 (s, 3H), 0.68 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 260 (M + H + ). Example 172: (3,4-dihydro-2H-pyran-6-yl)(2,6,6-trimethylcyclohex-1- enyl)methanol

To a stirred solution of 3, 4-dihydro-4H-pyran (504.7 mg, 6.0 mmol) in anhydrous tetrahydrofuran (10 mL) at 0 °C under nitrogen was slowly added n-butyl lithium (3.13 mL, 1.6 M). The reaction mixture was warmed to room temperature stirred for 6 hours. Then to the reaction solution was cooled to - 78 °C and 2,6,6-trimethylcyclohex-1-enecarbaldehyde (608.0 mg, 4.0 mmol) was added. The mixture was stirred at -78 °C for 3 hours. The mixture was warmed to 0 °C and quenched by the addition of saturated aqueous ammonium chloride (20 mL). The reaction was extracted with ethyl acetate, dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 -> 15:1) to afford the title compound as a colorless oil (420 mg, Yield: 45%). R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 4.73 (s, 2 H), 4.10-3.98 (m, 2 H), 2.24 (s, 1 H), 2.03-1.96 (m, 4 H), 1.81-1.80 (m, 2 H), 1.73 (s, 3 H), 1.61- 1.53 (m, 2 H), 1.49-1.43 (m, 2 H), 1.10 (s, 3 H), 0.94 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 2 9 (M - H 2 0 + H + ). Example 173: (3,4-dihydro-2W-pyran-6-yl)(2,6,6-trimethylcyclohex-1- enyl)methanone

To a solution of the product of Example 172 (280 mg, 1.18 mmol) in dichloromethane (6.0 mL) at 0 °C was added sodium bicarbonate (99.5 mg, 1.18 mmol) and Dess-Martin periodinane (1.0 g, 2.36 mmol). The mixture was stirred at room temperature for 2 hours. Aqueous saturated sodium bicarbonate (60 mL) was added and then the mixture was extracted with dichloromethane and the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1 -> 30:1) to afford the title compound as a yellow solid (70 mg, Yield: 25%). Mp = 54-57 °C; R f = 0.25 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 5.95 (t, J = 4.4 Hz, 1 H), 4.10 (t, J = 4.4 Hz, 2 H), 2.25- 2.21 (m, 2 H), 1.97 (t, J = 6.6 Hz, 2 H), 1.88-1.82 (m, 2 H), 1.71-1.65 (m, 2 H), 1.52 (s, 3 H), 1.48-1.45 (m, 2 H), 1.03 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ).

Example 174: (±M4bR,8aSMb,8,8-trimethyl-3,4,4b,5,6,7,8,8a- octahydroindeno[2,1-6]pyran-9(2H)-one

The product of Example 173 (50 mg, 0.17 mmol) was dissolved in methansulfonic acid ( .0 mL). The mixture was stirred at room temperature for 1 hour and then at 50 °C for 2 hours. To the solution was added water (6 mL) and the reaction was cooled to room temperature. The mixture was extracted with ethyl acetate and the organic layer was dried over anhydrous magnesium sulfate, filtered, and then concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 - >15:1) to afford the title compound as a white solid (40 mg, Yield: 62%). Mp = 61-62 °C; R f = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 4.07 (t, J = 5.2 Hz, 2 H), 2.31-2.20 (m, 2 H), 1.96-1.90 (m, 2 H), 1.81 (s, 1 H), 1.65-1.52 (m, 4 H), 1.39-1.36 (m, 2 H), 1.22 (s, 3 H), 1.17 (s, 3 H), 0.93 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 235 (M + H + ).

Example 175: (±)-(4b/?,8aS,9R)-4b,8,8-trimethyl-2,3,4,4b,5,6,7,8,8a,9- decahydroindeno[2,1-b]pyran-9-ol

To a mixture of lithium aluminum hydride (14.6 mg, 0.38 mmol) in anhydrous tetrahydrofuran (2 mL) at 0 °C was dropwise added a solution of the product of Example of 174 (30 mg, 0.12 mmol) in anhydrous tetrahydrofuran (3 mL). The reaction was stirred at room temperature for 2 hours. The reaction was quenched by the addition of water (0.1 mL), aqueous sodium hydroxide (0.1 mL) and water (0.3 mL). The mixture was extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 15:1) to afford the title compound as a colorless oil (14 mg, Yield: 49%). R f = 0.2 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 4.50-4.49 (m, 1 H), 4.08-4.05 (m, 1 H), 3.98-3.93 (m, 1 H), 2.03-1.97 (m, 1 H), 1.88-1.83 (m, 1 H), 1.80-1.72 (m, 2 H), 1.65-1.60 (m, 1 H), 1 .50-1.25 (m, 6 H), 1.15 (s, 3 H), 1.07 (s, 6 H), 0.98-0.90 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 219 (M - H 2 0 + H + ).

Example 176: (±)-(4a/?,9a ?)-9-ethyl-4,4,4a-trimethyl-4,4a,9,9a-tetrahydro- 1W-carbazol-2(3W)-one

Example 176a: 1-ethyl-3-methyl-1H-indole

To a stirred solution of 3-methy 1-1 /-/-indole (656 mg, 5 mmol) in anhydrous dimethylforamide (20 mL) at 0 °C was added sodium hydride (60%, 250 mg, 6.25 mmol). The mixture was warmed to room temperature and stirred for 30 minutes after which ethyl iodide (1.2 mL, 15 mmol) was added in one portion. The mixture was stirred at room temperature for 3 hours. The mixture was concentrated under reduced pressure and the residue was taken up in ethyl acetate. The organic solution was washed with water (80 mL x 2), brine (80 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether) afforded the title compound as a colorless oil (744 mg, Yield: 93%). R f = 0.5 (100:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.56 (d, J = 8.0 Hz, 1 H), 7.30 (d, J = 8.4 Hz, 1 H), 7.20 (t, J = 7.2 Hz, 1 H), 7.10 (t, J = 7.2 Hz, 1 H), 6.88 (s, 1 H), 4.11 (q, J = 6.8 Hz, 2 H), 3.33 (s, 3H), 1.43 (t, J = 7.2 Hz, 3H) ppm; Mass spectrum (ESI +ve) m/z 160 (M + H + ).

Example 176: (±)-(4a ?,9aR)-9-ethyl-4,4,4a-trimethyl-4,4a,9,9a-tetrahydro- 1H-carbazol-2(3H)-one

To a stirred solution of the product of Example 176a (740 mg, 4.65 mmol) in acetonitrile (6.4 mL) cooled to 0 °C was added concentrated sulfuric acid (0.64 mL) follow by mesityl oxide (1.28 g, 13 mmol). The reaction was stirred at 0 °C for 30 minutes after which time the reaction mixture was allowed to rise to room temperature and stirring was continued overnight. The mixture was added slowly with cooling and stirring to a suspension of sodium bicarbonate (5.0 g) in water (50 mL). The solution was extracted with diethyl ether (100 mL x 3). The organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a yellow oil (1.102 g, Yield: 92%); = 0.5 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.12 (dt, J ? = 7.6 Hz, J 2 = 1.2 Hz, 1 H), 7.06 (d, J = 7.6 Hz, H), 6.71 (dt, Ji = 7.6 Hz, J 2 = 0.8 Hz, 1 H), 6.48 (d, J = 7.6 Hz, 1 H), 3.67 (t, J = 4.2 Hz, 1 H), 3.24-3.19 (m, 1 H), 3.08-3.02 (m, 1 H), 2.71 (d, J = 4.0 Hz, 2H), 2.29 (d, J = 15.6 Hz, 1 H), 2.22 (d, J = 16.4 Hz, 1 H), 1.45 (s, 3H), 1.07 (s, 3H), 1.04 (t, J = 7.0 Hz, 3H), 0.85 (s, 3H) ) ppm; Mass spectrum (ESI +ve) m/z 258 (M + H + ).

Example 177: (3,3-dimethylcyclohex-1-enyl)(2,3,4- trifluorophenyl)methanol

1-Bromo-2,3,4-trifluorobenzene (1.53 g, 7.24 mmol) was dissolved in anhydrous tetrahydrofuran (8 mL). To a stirred mixture of magnesium (173.6 mg, 7.24 mmol) and iodine (5 mg) under an argon atmosphere was added a portion of the solution of 1-bromo-2,3,4-trifluorobenzene (2 mL). The reaction mixture was warmed to 40 °C and the remaining solution of then 1-bromo- 2,3,4-trifluorobenzene (6 mL) was added dropwise. The reaction was then heated to reflux for 2 hours. Then to the reaction solution was cooled to -78 °C and the product of Example 153d (200 mg, 1.45 mmol) was added. The reaction was stirred at -78 °C for 2 hours and then warmed to room temperature and stirred for 1 hour. The mixture was quenched by the addition of saturated aqueous ammonium chloride (20 mL). The solution was extracted with ethyl acetate, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 20:1) to afford the title compound as a colorless oil (165 mg, Yield: 42 %) = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.20-7.14 (m, 1 H), 6.99-6.92 (m, 1 H), 5.59 (s, 1 H), 5.33 (d, J = 3.6 Hz, 1 H), 1.89 (d, J = 4.0 Hz, 1 H), 1.85-1.79 (m, 1 H), 1.76-1 .70 (m, 1 H), 1.61-1.57 (m, 2 H), 1.41-1.37 (m, 2 H), 1.00 (s, 3 H), 0.98 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 253 (M - H 2 0 + H + ).

Example 178: (3,3-dimethy lcycIohex-1 -eny l)(2,3,4-triff uoropheny I) methanone

To a solution of the product of Example 177 (155 mg, 0.57 mmol) in dichloromethane (3 mL) at 0 °C was added Dess-Martin periodinane (486.2 mg, 1.14 mmol). The mixture was warmed to room temperature and stirred for 2 hours and then quenched by the addition of saturated aqueous sodium bicarbonate (20 mL), The mixture was extracted with dichloromethane and the organic phase dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 80:1 -> 60:1) to afford the title compound as a colorless oil (100 mg, Yield: 65 %). R f = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.16-7.10 (m, 1 H), 7.07-7.00 (m, 1 H), 6.25 (m, 1 H), 2.36-2.32 (m, 2 H), 1.75-1.69 (m, 2 H), 1.52-1.49 (m, 2 H), 1.04 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 269 (M + H + ). Example 179: (3,3-dimethylcyclohex-1-enyl)(4-fluoro-2-methylphenyl) methanol

2-Bromo-5-fluorotoluene (1.17 g, 7.23 mmol) was dissolved in anhydrous tetrahydrofuran (8 mL). To a stirred mixture of magnesium (173.6 mg, 7.23 mmol) and iodine (5 mg) under argon was added a portion of the solution of 2-bromo-5-fluorotoluene (2 mL). The reaction was warmed to 40 °C the remaining solution of 2-bromo-5-fluorotoluene (6 mL) was added. The reaction was then refluxed for 2 hours after which time it was cooled to -78 °C and the product of Example 153d (200 mg, 1.45 mmol) was added. The reaction was stirred at -78 °C for 2 hours and then warmed to room temperature and stirred for an additional hour. The mixture was quenched by the addition of saturated aqueous ammonium chloride (20 mL). The mixture was extracted with ethyl acetate and the organic phase dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 20:1) to afford the title compound as a colorless oil (259.1 mg, Yield: 72 %). R, = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDC ) δ 7.43-7.39 (m, 1 H), 6.91-6.82 (m, 2 H), 5.52 (s, 1 H), 5.18 (d, J = 3.6 Hz, 1 H), 2.27 (s, 3 H), 1.86-1.80 (m, 1 H), 1.72 (d, J = 3.2 Hz, 1 H), 1.68-1.57 (m, 3 H), 1.44-1.32 (m, 2 H), 1.00 (s, 3 H), 0.98 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 271 (M + Na + ).

Example 180: (3,3-dimethylcyclohex-1-enyl)(4-fluoro-2-methylphenyl) methanone

To a solution of the product of Example 179 (240 mg, 0.97 mmol) in dichloromethane (5 mL) at 0 °C was added Dess-Martin periodinane (819.5 mg, 1.93 mmol). The mixture was warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of aqueous sodium bicarbonate (30 mL) and then extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 80:1 -> 60:1) to afford the title compound as a colorless oil (162 mg, Yield: 68 %). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.19-7.16 (m, 1 H), 6.93-6.87 (m, 2 H), 6.12 (s, 1 H), 2.35-2.32 (m, 2 H), 2.27 (s, 3 H), 1.75-1.69 (m, 2 H), 1.50-1.47 (m, 2 H), 1.01 (s, 6 H) ) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 181: 6-fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1H-fluorene

To a stirred solution of the product of Example 166 (39 mg, 0.17 mmol) in dichloromethane (5 mL) at 0°C was added thionyl chloride (0.1 mL). The reaction was stirred at 0°C for 30 minutes and then warmed to room temperature and stirred for an additional 4 hours. The reaction was quenched by addition of aqueous sodium hydroxide (0.5 N, 30 mL). The resulting mixture was extracted with dichloromethane (30 mL) and the organic phase was washed with saturated aqueous sodium chloride (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether) afforded the title compound as a colorless oil (29 mg, Yield: 81 %). R f = 0.8 (petroleum ether); H NMR (400 MHz, CDCI 3 ) δ 7.17 (dd, J = 9.2, 2.4 Hz, 1 H), 7.14 (dd, J = 8.0, 5.2 Hz, 1 H), 6.93-6.88 (m, 1 H), 6.34 (s, 1 H), 2.93 (s, 1 H), 2.71-2.66 (m, 1 H), 2.33-2.25 (m, 1 H), 1.81-1.75 (m, 1 H), 1.60-1.56 (m, 1H), 1.55-1.46 (m, 2H), 1.40 (s, 3H), 0.35 (s, 3H) ppm; Mass spectrum (APCI +ve) m/z 216 (M + ).

Example 182: (±)-(4aS,9aS)-6-fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1H- fluoren-9(9aW)-one oxime

To a stirred solution of the product of Example 155 (51 mg, 0.22 mmol) in pyridine (3 mL) was added hydroxyl amine hydrochloride (61 mg, 0.88 mmol). The reaction was heated to reflux for 1 hour. The mixture was concentrated under reduced pressure and the residue was purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 8:1 ) to yield a yellow solid (32 mg), which was recrystallized from cold petroleum ether to afford the title compound as a light yellow solid (12 mg, Yield: 22%). Mp = 163-166 °C; R f = 0.4 (8:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 8.34 (dd, J = 8.4, 5.6 Hz, 1 H), 7.52 (bs, 1 H), 7.07 (d, J = 8.8 Hz, 1 H), 6.99 (dd, J = 8.4, 8.0 Hz, 1 H), 3.06 (t, J = 5.8 Hz, 1 H), 2.89 (d, J - 6.8 Hz, 1 H), 2.28-2.25 (m, 1 H), 1.61-1.42 (m, 4H), 1.35-1.26 (m, 1 H), 1.08 (s, 3H), 0.29 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 248 (M + H + ). Example 183: (2-deutero-3,3-dimethylcyclohex-1-enyl)(4-fluorophenyl) methanol

Example 183a: 2-deutero-3,3-dimethylcyclohex-1-enecarbaldehyde

To a stirred solution of a mixture of the product of Example 153c (7 5 mg, 3.4 mmol) in dry diethyl ether (15 mL) at -20 °C under argon was added lithium aluminum deuteride (214 mg, 5.1 mmol). The resulting mixture was stirred for 1 hour during which time the reaction temperature was raised to room temperature gradually. The reaction was quenched by cooling to 0 °C slowly adding ethyl acetate (2 mL). Saturated aqueous ammonium chloride (2 mL) was added to the mixture and then it was diluted with ether (100 mL). The organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to give a colorless liquid. The material was dissolved in acetone (10 mL) and hydrochloric acid (2N, 0.05 mL) was added and the solution was shaken for 1 minute. Sodium bicarbonate (50 mg) was added and the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) gave the title compound as a colorless liquid (190 mg, Yield: 40%). R f - 0.7 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 9.40 (s, 1 H), 2.14 (t, J = 6.2 Hz, 2H), 1.67-1.64 (m, 2H), 1.52- 1.50 (m, 2H), 1.10 (s, 6H) ppm.

Example 183: (2-deutero-3,3-dimethylcyclohex-1-enyl)(4-fluorophenyl) methanol

To a stirred solution of the product of Example 183a (190 mg, 1.36 mmol) in anhydrous tetrahydrofuran (10 mL) under argon at -78 °C was added (4-fluorophenyl)magnesium bromide (0.8 M in tetrahydrofuran, 3.5 ml, 2.8 mmol). The reaction was warmed to room temperature and stirred for 2 hours. The reaction was quenched by addition of saturated aqueous ammonium chloride (10 mL). Water (20 mL) and ethyl acetate (20 mL) were added and the layers were separated. The aqueous layer was extracted with ethyl acetate (25 mL x 2) and the combined organic layers were washed with brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 25: 1) afforded the title compound as a colorless oil (136 mg, Yield: 42%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.32- 7.26 (m, 2H), 7.01 (t, J = 8.4 Hz, 2H), 5.04 (s, 1 H), 1.85-1.76 (m, 1 H), 1.68- 1.54 (m, 3H), 1.44-1.34 (m, 2H), 1.02 (s, 3H), 1.01 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 218 (M - H 2 0 + H + ). Example 184: (3,4-difluoropheny l)(3,3-dimethylcyclohex-1 -eny l)methanol

To a mixture of iodine (5 mg) and magnesium (0.26 g, 1 1 mmol) in tetrahydrofuran (5 mL) was added 4-bromo-1 ,2-difluorobenzene (2.47 g, 10 mmol) dropwise. The mixture was stirred at room temperature for 5 minutes and refluxed for 2 hours. To the in situ generated solution of (3,4- difluorophenyl)magnesium bromide in tetrahydrofuran at -78 °C was added the product of Example 153d (200 mg, 1.45 mmol). The reaction was stirred at -78 °C for 2 hours. The reaction mixture was quenched by the addition of saturated ammonium chloride (5 mL) and the mixture was extracted with ethyl acetate (30 mL x 3). The organic phase was washed with water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford a semi solid which was further purified by Prep- HPLC to give the title compound as a colorless oil (200 mg, Yield: 55%). = 0.3 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3+D20) δ 7.20-7.05 (m, 3H), 5.59 (s, 1 H), 5.02 (s, 1 H), 1.88-1.81 (m, 1H), 1.65-1 .54 (m, 3H), 1.42-1.37 (m, 2H), 1.02 (s, 3H), 1.01 (s, 3H) ppm; Mass spectrum (El +ve) m/z 252 (M + ). Example 185: (3,4-difluoropheny l)(3,3-dimethy Icy clohex- - enyl)methanone

The solution of the product of Example 184 (140 mg, 0.55 mmol) in dichloromethane (8 mL) at 0 °C was added sodium bicarbonate (47 mg, 0.55 mmol) and Dess-Martin periodinane (471 mg, 1.11 mmol). The reaction was warmed to room temperature and stirred overnight. The reaction mixture was quenched with 5% hydrochloric acid (5 mL) and then the mixture was extracted with dichloromethane (20 mL x 3). The organic phase was washed with saturated aqueous sodium bicarbonate (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (120 mg, Yield: 87%). R f = 0.7 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.52-7.47 (m, 1 H), 7.41-7.40 (m, 1 H), 7.25-7.20 (m, 1 H), 6.22 (s, 1 H), 2.36-2.32 (m, 2H), 1.75-1.71 (m, 2H), 1.54-1.51 (m, 2H), 1.08 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 251 (M + H + ).

Example 186: 6-fluoro-4,4-dimethyl-2,3,4,4a,9,9a-hexahydro-1H-fluorene To a stirred solution of the product of Example 181 (20 mg, 0.092 mmol) in methanol (2 mL) was added 5% palladium on carbon (5 mg). The reaction was stirred under hydrogen at room temperature overnight.. The mixture was filtered through celite and the celite washed with additional methanol. The filtrate was concentrated under reduced pressure. Purification of the residue by flash silica gel chromatography (eluent: petroleum ether) gave the title compound as a colorless oil (15.7 mg, Yield: 78%). R f = 0.83 (petroleum ether); H NMR (400 MHz, CDCI 3 ) δ 7.12 (dd, J = 8.4, 5.6 Hz, 1 H), 7.00 (dd, J = 9.6, 2.4 Hz, 1 H), 6.85-6.79 (m, 1 H), 2.71 (d, J = 6.8 Hz, 1 H), 2.67-2.65 (m, 2H), 2.61-2.53 (m, 1 H), 1.66-1.58 (m, 2H), 1.55-1.49 (m, 2H), 1.44-1.38 (m, 1 H), 1.30-1.23 (m, 1 H), 1.06 (s, 3H), 0.62 (s, 3H) ppm; Mass spectrum (El +ve) m/z 218 (M + ).

Example 187: (±)-(4aS,9aS)-6-fluoro-4,4-dimethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aW)-one O-methyl oxime

To a stirred solution of the product of Example 182 (18 mg, 0.073 mmol) in anhydrous tetrahydrofuran (2 mL) under argon at 0 °C was added sodium hydride (5 mg, 0.125 mmol). The reaction was warmed to room temperature and stirred for 2 hours after which time it was recooled to 0 °C and methyl iodide (0.05 ml, 0.8 mmol) was added. The reaction mixture was warmed to room temperature and stirred for 2 hours. The reaction was quenched by addition of water (1 ml). The mixture was then concentrated under reduced pressure and the residue diluted with ethyl acetate (30 mL). The organic phase washed with water (20 mL) and brine (20 ml), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1.0 -> 25:1 ) gave yellow oil (12 mg) which was further purified by preparative thin layer chromatography to afford a mixture of geometric isomers of the title compound as a yellow oil (9.5 mg, Yield: 50%). R f = 0.6 (25:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) {Major isomer) δ 8.22 (dd, J = 8.8, 5.6 Hz, 1 H), 7.06 (dd, J = 8.8, 2.4 Hz, 1 H), 7.01-6.93 (m, 1 H), 3.98 (s, 3H), 3.04 (t, J = 7.6 Hz, 1 H), 2.86 (d, J = 7.2 Hz, 1 H), 2.31-2.26 (m, 1 H), 1.62-1.25 (m, 5H), 1.07 (s, 3H), 0.29 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 262 (M + H + ).

Example 188: (2-deutero-3,3-dimethy lcyclohex-1 -eny l)(4-fluoropheny I) methanone

A stirred solution of the product of Example 183 (126 mg, 0.54 mmol) and pyridine (0.2 mL) in dichloromethane (4 mL) was treated with Dess-Martin periodinane (344 mg, 0.81 mmol). The reaction was stirred for 30 minutes after which water (5 mL) was added to the reaction mixture followed by aqueous sodium hydroxide (1.0 M, 5 ml). The bilayer mixture was allowed to stir vigorously for 10 minutes. Dichloromethane (20 mL) and water (20 mL) were added and the layers were separated. The aqueous layer was extracted with ethyl acetate (20 mL x 3). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to afford the title compound as a colorless oil ( 08 mg, Yield: 86%). R f = 0.6 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.69-7.65 (m, 2H), 7.13-7.08 (m, 2H), 2.35 (t, J = 6.2 Hz, 2H), 1.77-1.71 (m, 2H), 1.53-1.50 (m, 2H), 1.07 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 234 (M + H + ). Example 189: (±)-(4aS,9aS)-4a-deutero-6-fluoro-4,4-dimethyl-2,3,4,4a- tetrahydro-1 H-fluoren-9(9aH)-one

A solution of the product of Example 188 (130 mg, 0.56 mmol) in methansulfornic acid (2 mL) under argon was heated to 80 °C for 5 hours. The material was cooled to room temperature and poured into ice water (30 mL). The mixture was then extracted with ethyl acetate (20 mL x 3) and the combined organic phase was washed with aqueous sodium hydroxide (1 N, 25 mL x 2) and brine (30 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1 :0 -> 100: 1) to afford the title compound as a white solid (45 mg, Yield: 35%). Mp = 62-64 °C; R f = 0.4 (30:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.78-7.74 (m, 1 H), 7.18 (d, J = 8.8 Hz, 1 H), 7.09-7.05 (m, 1 H), 2.69 (d, J = 7.6 Hz, 1 H), 2.44 (d, J = 13.6 Hz, 1 H), 1.56-1.49 (m, 2H), 1.43-1.34 (m, 3H), 1.16 (s, 3H), 0.13 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 234 (M + H + ).

Example 190: (±)-(4aR,9aS)-4a-ethyl-6-fluoro-1,1-dimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a stirred solution of the product of Example 189 (30 mg, 0.13 mmol) in methanol (3 mL) at 0 °C was added sodium borohydride (14 mg, 0.39 mmol). The reaction was stirred at room temperature overnight. Water (1 ml) was added to quench the reaction and then the mixture was concentrated under reduced pressure. The residue was dissolved in ethyl acetate (25 mL) and then it was washed with water (20 mL x 2) and brine (20 mL x 2). The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. Purification of the residue by flash column chromatography (eluent: petroleum ether/ethyl acetate = 30:1) afforded a mixture of the alcohol isomers of the title compound as a colorless oil (28.6 mg, Yield: 93%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.34-7.31 (m, 1 H), 7.21-7.18 (m, 1 H), 6.94- 6.89 (m, 1 H), 4.95 (d, J = 6.0 Hz, 1 H), 2.69-2.65 (m, 1 H), 1.65-1.59 (m, 1 H), 1.56-1.49 (m, 2H), 1.37-1.26 (m, 2 H), 1.21-1.13 (m, 7H) ppm; Mass spectrum (ESI +ve) m/z 218 (M - H 2 0 + H + ).

Example 191 : 4-(hydroxy(2,6,6-trimethylcyclohex-1- enyl)methyl)benzonitrile

To a solution of isopropylmagnesium bromide (1.0 M in tetrahydrofuran, 4.75 mL, 4.75 mmol) was added 4-bromobenzonitrile (720 mg, 3.96 mmol) in tetrahydrofuran (5 mL) and the mixture was stirred at room temperature for 30 minutes. The reaction was cooled to 0 °C and 2,6,6- trimethylcyclohex-1-enecarbaldehyde (500 mg, 3.3 mmol) was added and then the reaction was warmed to room temperature and stirred overnight. The mixture was quenched with saturated aqueous ammonium chloride and the organics extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a light yellow solid (80 mg, Yield: 9.5Q). Mp = 131.1-132.9 °C; R f = 0.5 (5:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.61 (d, J = 8.4 Hz, 2 H), 7.54 (d, J = 8.0 Hz, 2 H), 5.38 (d, J = 3.2 Hz, 1 H), 1.97-1.96 (m, 2 H), 1.86 (d, J = 4.4 Hz, 1 H), 1.66-1.52 (m, 4 H), 1.29 (s, 3 H), 1.19 (s, 3 H), 1.10 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 238 (M - H 2 0 + H + ).

Example 192: 4-(2,6,6-trimethy(cyclohex-1-enecarbonyi)benzonitriIe

To a solution of the product of Example 191 (36 mg, 0.14 mmol) in dichloromethane (2 mL) at 0 °C was added Dess-Martin periodinane (162 mg, 0.38 mmol). The reaction was warmed to room temperature and stirred for 1 hour. Petroleum ether (20 mL) was added and the precipitated solid was filtered off. The filtrate was concentrated under reduced pressure and the residue purified by preparative thin layer chromatography to give the title compound as a light yellow solid (32 mg, Yield: 91 LT). Mp = 122.0-123.4 °C; = 0.4 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 8.02 (d, J = 8.4 Hz, 2 H), 7.76 (d, J = 8.4 Hz, 2 H), 2.10 (t, J = 6.4 Hz, 2 H), 1.79- 1.76 (m, 2 H), 1.58-1.55 (m, 2 H), 1.42 (s, 3 H), 1.02 (s, 6 H) ) ppm; Mass spectrum (ESI +ve) m/z 254 (M + H + ). Example 193: (2,4-difluorophenyl)(3,3-dimethylcyclohex-1-enyl)methanol

To the mixture of magnesium (158 mg, 6.51 mmol) and iodine (5 mg) in anhydrous tetrahydrofuran (5 mL) at room temperature was added -bromo- 2,4-difluorobenzene (1.26 g, 6.51 mmol) and then the mixture was warmed up to reflux for 3 hours. The reaction was cooled to -78 °C and a solution of the product of Example 153d (180 mg, 1.30 mmol) in anhydrous tetrahydrofuran (5 mL) was added. The reaction mixture was allowed to warm to room temperature and then was stirred for 2 hours. The reaction was quenched by the addition of saturated aqueous ammonium chloride. The organics were extracted with ethyl acetate (100 mL) and the organic layer was washed with water (50 mL x 2) and brine (50 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 30:1) gave a colorless oil which was which was further purified by Prep-HPLC to afford the title compound as a colorless oil (89 mg, Yield: 27%). R f = 0.4 ( 0:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.44-7.38 (m, 1 H), 6.89-6.85 (m, 1 H), 6.80-6.74 (m, 1H), 5.58 (s, 1H), 5.33 (d, J = 3.6 Hz, 1 H), 1.85 (d, J = 4.0 Hz, 1 H), 1.80-1.75 (m, 2H), 1.60-1 .57 (m, 2H), 1.41-1.37 (m, 2H), 1.00 (s, 3H), 0.98 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 235 (M - H 2 0 + H + ).

Example 194: pyrimidin-5-yl(2,6,6-trimethylcyclohex-1-enyl)methanol

To a stirred solution of 5-bromopyrimidine (954 mg, 6.0 mmol) in dry tetrahydrofuran (15 mL) cooled to -78 °C was slowly added n-butyl lithium (2.5 M in hexanes, 2.4 ml, 6.0 mmol). The mixture was stirred for 15 minutes at - 78 °C. 2,6,6-trimethylcyclohex-1-enecarbaldehyde (457 mg, 3.0 mmol) was added, and the reaction was stirred at -78 °C for 2.5 hours and then warmed to room temperature and stirred overnight. Saturated aqueous ammonium chloride was added to quench the reaction followed by water dissolve the solids. The mixture was extracted with ethyl acetate (50 mL x 3) and the combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 200:1 -> 2:1) afforded the title compound as yellow solid (243 mg, Yield: 35%). Mp = 141-143 °C; R f = 0.3 (2:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 9.10 (s, 1 H), 8.78 (s, 2H), 5.46 (s, 1 H), 2.00 (t, J = 6.0 Hz, 2H), 1.68-1.60 (m, 3H), 1.58-1.53 (m, 2H), 1.38 (s, 3H), 1.20 (s, 3H), 1.09 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 233 (M + H + ). Example 195: (3-fluoro-4-(trifluoromethoxy)phenyl)(2,6,6- trimethylcyclohex-1-enyl)methanol

4-Bromo-2-fluoro-1-(trifluoromethyl) benzene (3.08 g, 15.0 mmol) was dissolved in dry tetrahydrofuran (10 ml_). To a stirred mixture of magnesium (360 mg, 15 mmol) and iodine (5.0 mg) in dry tetrahydrofuran (2 ml_) at 50 °C under argon was added 2 ml_ of solution of the 4-bromo-2-fluoro-1- (trifluoromethyl) benzene solution and stirring was continued for 10 minutes then the remaining 4-bromo-2-fluoro-1-(trifluoromethyl) benzene solution (8 mL) was added dropwise at 50 °C. When the addition was complete, the reaction was refluxed for 1.5 hours. The Grignard solution was cooled to -78 °C and 2,6,6-trimethylcyclohex-1-enecarbaldehyde was added (457 mg, 3.0 mmol). The reaction then allowed to warm to room temperature and it was stirred for 1 hour. Saturated aqueous ammonium chloride was added to quench the reaction followed by water to dissolve the solids. The mixture was extracted with ethyl acetate (50 mL x 3) and the combined organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) gave 836 mg of a colorless oil 71 mg of which was further purified by prep-HPLC to afford the title compound as a colorless oil (52 mg, Yield: 61 %). R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +H 2 0) δ 7.30-7.26 (m, 1 H), 7.23-7.18 (m, 2H), 5.33 (s, 1 H), 1.98 (t, J = 6.0 Hz, 2H), 1.67-1.61 (m, 2H), 1.57-1.52 (m, 2H), 1.35 (s, 3H), 1.18 (s, 3H), 1.08 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 315 (M - H 2 0 + H + ).

Example 196: (±)-(4aS,9aS)-6-fluoro-4,4,8-trimethyl-2,3,4,4a-tetrahydro- 1 H-fluoren-9(9aH)-one

The product of Example 180 (147 mg, 0.60 mmol) was dissolved in methansulfonic acid (3.0 mL) and then mixture was warmed to 80 °C and stirred overnight. The reaction was cooled to room temperature and water (15 mL) was added. The organics were then extracted with ethyl acetate and the organic phase was dried over anhydrous sodium sulfate and then it was concentrated under reduced pressure. The residue was repeatedly purified by preparative thin layer chromatography (eluent: petroleum ether/dichloromethane = 5:1) to give a colorless oil which was taken up in ethyl acetate (5 mL) washed with 1 N hydrochloric acid (5 mL x 3), aqueous sodium hydroxide (5 mL), and then with water (5 mL) to afford 25 mg colorless oil. To the oil (25 mg, 0.10 mmol) in methanol (1 mL) at 0 °C was added sodium borohydride (11.5 mg, 0.3 mmol). The reaction was allowed to warm to room temperature and stirred for 3 hours. The reaction was quenched by the addition of water (2 mL) and then the mixture was concentrated under reduced pressure. The aqueous mixture was then extracted with ethyl acetate, dried over anhydrous sodium sulfate and then concentrated in vacuo. The residue (20 mg) was dissolved in dichloromethane (1.0 mL) cooled to 0 °C and Dess- artin periodinane (68.3 mg, 0.161 mmol) was added. The mixture was stirred at room temperature for 2 h. To the reaction mixture was added aqueous sodium bicarbonate (10 mL) and then it was extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a colorless oil (10 mg). R f = 0.5 (10:1 petroleum ether/ethyl acetate);

1H N R (400 MHz, CDCI 3 ) δ 6.98 (d, J = 8.8 Hz, 1 H), 6.82 (d, J = 9.6 Hz, 1 H), 3.09 (d, J = 7.2 Hz, 1 H), 2.67-2.63 (m, 1 H), 2.63 (s, 3H), 2.42 (d, J = 13.6 Hz, 1 H), 1.56-1.47 (m, 2H), 1.42-1.32 (m, 3H), 1.14 (s, 3H), 0.14 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 247 (M + H + ). Example 197: (il-^aS.SS.SaSl-e-fluoro^Ae-trimethyl^.a^^a.S.Sa- hexahydro-1 W-fIuoren-9-ol

To a solution of the product of Example 196 (25 mg, 0.10 mmol) in methanol (1 mL) at 0 °C was added sodium borohydride (11.5 mg, 0.3 mmol). The reaction was warmed to room temperature and stirred for 3 hours. The reaction was quenched by the addition of water (2 mL) and then it was concentrated under reduced pressure. The aqueous residue was extracted with ethyl acetate and then the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1) to afford the title compound as white solid (24 mg, Yield: 97%). Mp = 70-73 °C; R f = 0.3 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 6.89 (d, J = 9.2 Hz, 1 H), 6.73 (d, J - 9.2 Hz, 1 H), 4.99 (t, J = 6.0 Hz, 1 H), 2.80 (d, J = 4.0 Hz, 1 H), 2.47-2.40 (m, 1 H), 2.40 (s, 3H), 1.85-1.81 (m, 1 H), 1.71-1.61 (m, 3H), 1.54-1.41 (m, 2H), 1.31-1.25 (m, 1 H), 1.16 (s, 3H), 0.66 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 231 (M - H 2 0 + H + ). Example 198: (2,4-difluorophenyl)(3,3-dimethylcyclohex-1- enyl)methanone

To a solution of the product of Example 193 (109 mg, 0.43 mmol) in tetrahydrofuran (6 mL) at 0 °C was added sodium bicarbonate (36 mg, 0.43 mmol) and Dess-Martin periodinane (365 mg, 0.86 mmol). The mixture was then warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of 5% hydrochloric acid and extracted with dichloromethane (60 mL). The organic phase was washed water (50 mL x 2) and brine (50 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography afforded the title compound as a colorless oil (61 mg, Yield: 57%). = 0.6 (30:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.42-7.36 (m, 1 H), 6.96-6.91 (m, 1 H), 6.87-6.82 (m, 1 H), 6.23 (s, 1 H), 2.34 (t, J = 5.6 Hz, 2H), 1.73-1.70 (m, 2H), 1.51-1.48 (m, 2H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 251 (M + H + ).

Example 199: pyrimidin-5-yl(2,6,6-trimethylcyclohex-1- enyl)methanone

To a stirred solution of the product of Example 194 (117 mg, 0.51 mmol) in dichloromethane (3 mL) cooled to 0 °C was added Dess-Martin periodinane (427 mg, 1.02 mmol). After 45 min, the reaction was warmed to room temperature and stirred for 1.5 hours. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate (20 mL) and the mixture was extracted with dichloromethane (30 ml x 3). The combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 5:1) gave the title compound as a light yellow solid (89 mg, Yield: 76%). Mp = 46-48 °C; R f = 0.6 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ). δ 9.35 (s, 1 H), 9.18 (s, 2H), 2.11 (t, J = 6.2 Hz, 2H), 1.81- 1.75 (m, 2H), 1.61-1.56 (m, 2H), 1.47 (s, 3H), 1.05 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 231 (M + H + ).

Example 200: (3-fluoro-4-(trifluoromethoxy)phenyl)(2,6,6- trimethylcyclohex-1-enyl)methanone

A stirred solution of the product of Example 195 (227 mg, 0.68 mmol) in dichloromethane (4 mL) cooled to 0 °C was added Dess-Martin periodinane (579 mg, 1.37 mmol). After 30 minutes the reaction was warmed to room temperature and stirred for 1.5 hours. The reaction was quenched by the addition of saturated aqueous sodium bicarbonate (20 mL) and the mixture was extracted with dichloromethane (30 ml x 3). The combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 250:1) gave the title compound as a colorless oil (195 mg, Yield: 87%). R f = 0.8 (20:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.78-7.73 (m, 2H), 7.38 (dd, J = 8.0, 7.6 Hz, 1 H), 2.09 (t, J = 6.4 Hz, 2H), 1.80-1.74 (m, 2H), 1.57-1.54 (m, 2H), 1.44 (s, 3H), 1.03 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 331 (M + H + ).

Example 201 : (tetrahydro-2H-pyran-4-y l)(2,6,6-trimethy lcyclohex- - enyl)methanol

4-Bromotetrahydro-2H-pyran, (1.48 g, 9 mmol) was dissolved in tetrahydrofuran (6 mL). To magnesium (216 mg, 9 mmol) and iodine (5 mg) in tetrahydrofuran (2.5 mL) warmed to 50 °C was added a portion of the 4- bromotetrahydro-2/-/-pyran solution (1.5 mL) and the reaction was stirred at 50 °C for 30 minutes. The remaining 4-bromotetrahydro-2 -/-pyran solution (4.5 mL) was then added dropwise into the reaction mixture and stirring was continued at 50 °C for 1 hour. The reaction mixture was cooled to 0 °C and 2,6,6-trimethylcyclohex-l-enecarbaldehyde (456 mg, 3 mmol) in tetrahydrofuran (1.5 mL) was added dropwise into the reaction mixture. The reaction was then warmed to room temperature and stirred overnight. The mixture was quenched with saturated aqueous ammonium chloride and then extracted with ethyl acetate. The combined organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (266 mg, Yield: 37%). Mp = 1 11.5- 112.6 °C; R f = 0.2 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 4.03 (dd, Ji = 11.2 Hz, J 2 = 4.0 Hz, 1 H), 3.96 (dd, ^ = 11.2 Hz, J 2 = 3.6 Hz, 1 H), 3.90 (d, J = 9.6 Hz, 1 H), 3.40 (t, J = 11.8 Hz, 1 H), 3.31 (t, J = 11 .6 Hz, 1 H), 2.18-2.15 (m, 1 H), 2.06 (d, J = 13.2 Hz, 1 H), 1.96 (s, 2 H), 1.82 (s, 3 H), 1.56-1.21 (m, 8 H), 1.10 (s, 3 H), 0.96 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 221 (M - H 2 0 + H + ). Example 202: (±)-(4aR,9aS)-1,1,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1H-fluorene-6-carbonitrile

Example 202a: (±)-(4aR,9aS)-1 ,1 ,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-6-yl trifluoromethanesulfonate To a solution of the product of Example 4 (120 mg, 0.49 mmol) and triethylamine (0.11 ml) in dichloromethane (10 mL) at -20 °C was added trifluoromethanesufonic anhydride (210 mg, 0.12 mL, 0.74 mmol). The reaction mixture was warmed to 0 °C and stirred for 30 minutes after which it was slowly warmed to room temperature. The reaction mixture was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound as a yellow oil (190 mg, Yield: 100%,). R f = 0.8 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.76 (d, J = 8.4 Hz, 1 H), 7.31-7.23 (m, 2H), 2.25 (s, 1 H), 2.06-2.02 (m, 1 H), 1.75-1.64 (m, 2H), 1.47-1.36 (m, 3H), 1.31 (s, 3H), 1.22 (s, 3H), 0.66 (s, 3H) ppm; Mass spectrum (El +ve) m/z 377 (M + H + ).

Example 202: (±)-(4aR,9aS)-1 ,1 ,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1H-fluorene-6-carbonitrile

A mixture of the product of Example 202a (100 mg, 0.27 mmol), dicyanozinc (41 mg, 0.35 mmol) and palladium tetrakistriphenylphosphine (31 mg, 0.027 mmol) in dimethylformamide/water (4 mL/ mL) was stirred at 100 °C overnight under an argon atmosphere. Saturated aqueous ammonium chloride was added to the mixture and then it was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid (56 mg, Yield: 82%). Mp =157.2-157.5 °C; R f = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CD 3 OD) δ 8.01 (s, 1 H), 7.77 (s, 2H), 2.31 (s, 1 H), 2.20-2.14 (m, 1 H), 1.81-1.69 (m, 2H), 1.50-1.41 (m, 3H), 1.32 (s, 3H), 1.22 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (El +ve) m/z 276 (M + Na + ).

Example 203: (±)-(4a/?,9/?,9aS)-9-hydroxy-1 , 1 ,4a-trimethyl-2,3,4,4a,9,9a- hexahydro-1 - -fluorene-6-carbonitrile

To a solution of the product of Example 202 (35 mg, 0.14mmol) in methanol (5 mL) at 0 °C was added sodium borohydride (6.1 mg, 0.17 mmol). The reaction mixture was stirred for 30 minutes and then warmed to slowly to room temperature. Water (5 mL) was added to the reaction and then the mixture was extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound as a white solid (25 mg, Yield: 70%). Mp = 177-177.2 °C; R, = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CD 3 OD) δ 7.59 (d, J = 7.6 Hz, 1 H), 7.54 (s, 1 H), 7.51 (d, J = 8.0 Hz, 1 H), 5.02 (d, J = 9.6 Hz, 1 H), 1.79- 1.62 (m, 3H), 1.55 (s, 3H), 1.51-1.40 (m, 3H), 1.20 (s, 6H), 1.06-0.99 (dt, J = 13.4, 3.2 Hz, 1 H) ppm; Mass spectrum (ESI +ve) m/z 256 (M + H + ).

Example 204: (tetrahydro-2W-pyran-4-y l)(2,6,6-trimethy lcyclohex-1 - enyl)methanone

To a solution of the product of Example 201 (100 mg, 0.42 mmol) in dichloromethane (2 mL) 0 °C was added Dess-Martin periodinane (480 mg, 1.13 mmol). The reaction mixture was warmed to room temperature and stirred for 1 hour. Petroleum ether (20 mL) was added and the precipitated solid was filtered off. The filtrate was concentrated under reduced pressure and the residue purified by silica gel column chromatography to give the title compound as a colorless oil (94.6 mg, Yield: 95%). R f = 0.6 (5:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 4.03 (dd, J, = 11.2 Hz, J 2 = 3.6 Hz, 2 H), 3.39 (dt, .Λ = 1 1.6 Hz, J 2 = 2.0 Hz, 2 H), 2.66-2.60 (m, 1 H), 1.99 (t, J = 6.4 Hz, 2 H), 1.83 (dd, J, = 13.2 Hz, J 2 = 1.6 Hz, 2 H), 1.74-1.64 (m, 4 H), 1.56 (s, 3 H), 1.46-1.43 (m, 2 H), 1.08 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 237 (M + H + ).

Example 205: 2-fluoro-4-(hydroxy(2,6,6-trimethylcyclohex-1-enyl)methyl) benzonitrile

To a stirred solution of 4-bromo-2-fluorobenzonitrile (300.0 mg, 1.50 mmol) in terahydrofuran (6 mL) at room temperature was added isopropylmagnesium chloride (0.75 ml, 1.50 mmol) and the mixture was stirred for 30 minutes after which time a solution of 2,6,6-trimethylcyclohex-1 - enecarbaldehyde (175.6 mg, 1.15 mmol) in tetrahydrofuran (1 mL) was added and stirring was continued for 1 hour. Hydrochloric acid (0.1 N. 10 mL) was added to the reaction and then the mixture was extracted with ethyl acetate (10 mL x 3). The combined organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1 -> 10:1 ) to afford the title compound as a light yellow solid (74.5 mg, Yield: 24%). Mp = 120-121 °C; R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.55 (t, J = 7.2 Hz, 1 H), 7.34-7.29 (m, 2H), 5.34 (d, J - 4.4 Hz, 1 H), 1.98-1.96 (m, 2H), 1.84 (d, J = 4.4 Hz, 1 H), 1.66-1.61 (m, 2H), 1.57-1.54 (m, 2H), 1.30 (s, 3H), 1.18 (s, 3H) .11 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 274 (M + H + ).

Example 206: 2-fluoro-4-(2,6,6-trimethylcyclohex-1-enecarbonyl) benzonitrile

To a solution of the product of Example 205 (50 mg, 0.183 mmol) in dichloromethane (3 mL) at 0 °C was portionwise added Dess-Martin periodinane (139.6 mg, 0.329 mmol) and the mixture was then warmed to room temperature and stirred for 1 hour. Additional dichloromethane (10 mL) was added and the mixture was washed with saturated aqueous sodium bicarbonate and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to afford the title compound as a colorless oil (43 mg, Yield: 87%). R^ = 0.7 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ): δ 7.79-7.73 (m, 3H), 2.10 (t, J = 6.4 Hz, 2H), 1.79-1.76 (m, 2H), 1.58-1.55 (m, 2H), 1.42 (s, 3H), 1.02 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 272 (M + H").

Example 207: (+)-(4aR,9aS)-7-fluoro-1,1,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1H-fluorene-6-carbonitrile Example 207a: (±)-(4a ?,9aS)-7-fluoro-6-hydroxy-1 ,1 ,4a-trimethyl-2,3,4,4a- tetrahydro-1 H-fluoren-9(9aH)-one

To a solution of the product of Example 69 (340 mg, 1.23 mmol) in dichloromethane (4.0 mL) at -78 °C was added boron tribromide solution (6.2 mL, 1.0 M solution in dichloromethane). The reaction mixture was stirred at - 78 °C for 30 minutes and then slowly warmed to room temperature. After 12 hours at room temperature the solution was poured into ice and the mixture was extracted with dichloromethane (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash column chromatography to give the title compound as a yellow solid ( 80 mg, Yield: 56%). R f = 0.2 (10:1 petroleum ether/ethyl acetate); *H NMR (400 MHz, CD 3 OD) δ 7.24 (d, J = 9.6 Hz, 1 H), 6.94 (d, J = 7.6 Hz, 1 H), 2.12 (s, 1 H), 2.05-1.99 (m, 1 H), 1.70-1.60 (m, 2H), 1.50-1.35 (m, 3H), 1.23 (s, 3H), 1.17 (s, 3H), 0.64 (s, 3H) ppm.

Example 207b: (±)-(4afl,9aS)-7-fluoro-1 ,1 ,4a-trimethyl-9-oxo- 2,3,4,4a, 9, 9a-hexahydro-1H-fluoren-6-yl trifiuoromethanesulfonate

To a solution of the product of Example 207a (180 mg, 0.69 mmol) and triethylamine (0.14 ml_) in dichloromethane (10 mL) at -20 °C was added trifluoromethansulfonic anhydride (310 mg, 0.18 mL, 1.1 mmol). The reaction mixture was stirred for 30 minutes and then was slowly warmed to room temperature. The organic phase was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford the title compound as a yellow oil (293 mg, Yield: 100% yield), R f = 0.8 (10:1 petroleum ether/ethyl acetate); Mass spectrum (ESI +ve) m/z 395 (M + H + ). Example 207: (±)-(4aA?,9aS)-7-fluoro-1 ,1,4a-trimethyl-9-oxo-2,3,4,4a,9,9a- hexahydro-1tf-fluorene-6-carbonitrile

A mixture of the product of Example 207b (290 mg, 0.74 mmol) dicyanozinc (112.4 mg, 0.96 mmol) and palladium tetrakistriphenylphosphine (85 mg, 0.074 mmol) in dimethylformamide/water (8 mL 12 mL) was stirred at 100 °C for 4 hours under argon. Saturated aqueous ammonium chloride was added to the mixture, then it was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (100 mg, Yield: 50%). Mp = 150.7- 151.4 °C; R f = 0.7 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.69 (d, J = 9.2 Hz, 1 H), 7.45 (d, J = 7.2 Hz, 1 H), 2.27 (s, H), 2.08- 2.02 (m, 1 H), 1.75-1.64 (m, 2H), 1.45-1.32 (m, 3H), 1.29 (s, 3H), 1.20 (s, 3H), 0.64 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 272 (M + H + ).

Example 208: 7-fluoro-1 ,1 ,4a-trimethyl-6-(trifluoromethoxy)-2,3,4,4a- tetrahydro-IH-fluorene

To a stirred solution of the product of Example 195 (87 mg, 0.26 mmol) in dry dichloromethane (5 mL) cooled to 0 °C was added stannic chloride (0.1 mL 0.39 mmol). After 1 hour, the mixture was allowed to warm to room temperature and stirred for an additional hour. The reaction was cooled to 0 °C and quenched with water (2 mL). The mixture was concentrated under reduced pressure. Water (20 mL) was added and the aqueous layer was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (30 mL x 2), dried over anhydrous sodium sulfate , and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (eluent: petroleum ether) to give title compound as a pale yellow oil (62 mg, Yield: 75%). R f = 0.9 (petroleum ether); H NMR (400 MHz, CDCI 3 ) δ 7.11 (d, J = 7.2 Hz, 1 H), 7.04 (d, J = 10.4 Hz, 1 H), 6.30 (s, 1 H), 2.17-2.11 (m, 1 H), 2.00-1.90 (m, 1 H), 1.69-1.62 (m, 2H), 1.35 (s, 3H), 1.29 (s, 3H), 1.24 (s, 3H), 1.10 (dt, J = 13.2, 3.6 Hz, 1 H), 0.98 (dt, J = 13.2, 4.0 Hz, 1H) ppm; Mass spectrum (APCI +ve) m/z 314 (M + ).

Example 209: (6,6-dimethy Icy clohex-1 -eny l)(4-fluoropheny l)methanol

Example 209a: ethyl 6-hydroxy-2,2-dimethylcyclohexanecarboxylate

To a solution of ethyl 2,2-dimethyl-6-oxocyclohexanecarboxylate (4.95 g, 25 mmol) in MeOH (20 mL) was added sodium borohydride (1.42 g, 37.5 mmol) portion-wise at -78 °C. Then warmed up to -30 °C and quenched with 5% hydrochloric acid (3 mL). The reaction was concentrated under reduced pressure and the aqueous residue extracted with ethyl acetate (200 mL). The organic layer was washed water (100 mL x 2) and brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 10:1) gave the title compound as a colorless oil (2.83 g, Yield: 57%). R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 4.24-4.00 (m, 3H), 3.01 (bs, 1 H), 2.38 (s, H), 1.96-1.90 (m, 1 H), 1.84-1.78 (m, 1 H), 1.63-1.60 (m, 1 H), 1.52-1.40 (m, 2H), 1.29 (t, J = 7.2 Hz, 3H), 1.23-1.16 (m, 1 H), 1.07 (s, 3H), 0.99 (s, 3H) ppm.

Example 209b: ethyl 6,6-dimethylcyclohex-1-enecarboxylate

To a solution of the product of Example 209a (400 mg, 2.0 mmol) in dichloromethane (10 mL) at 0 °C was added dropwise thionyl chloride (0.58 mL, 7.96 mmol) and then pyridine (1.93 mL, 23.86 mmol) was added dropwise slowly. The mixture was warmed to room temperature and stirred for 5 hours. Water (10 mL) was added and the mixture was extracted with dichloromethane (100 mL). The organic phase was washed with saturated aqueous sodium bicarbonate (50 mL x 2) and brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) afforded the title compound as a light yellow oil (248 mg, Yield: 68%). R = 0.8 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) 6.81 (t, J = 4.0 Hz, 1 H), 4.17 (q, J = 7.2 Hz, 2H), 2.13 (t, J = 6.4 Hz, 2H), 1.64-1.60 (m, 2H), 1.50-1.47 (m, 2H), 1.29 (t, J = 7.2 Hz, 3H), 1.22 (s, 6H) ppm.

Example 209c: (6,6-dimethylcyclohex-1-enyl)methanol

To a stirred solution of the product of Example 209b (1.41 g, 7.7 mmol) in dry dichloromethane (80 mL) at -78 °C was added diisobutyl aluminum hydride (1.0 M in hexane, 31 mL, 31 mmol). The reaction was allowed to warm to 0 °C gradually over 1.5 hours and then the mixture was diluted with diethyl ether (100 mL). Tetrahydrofuran/water (5/1 , 30 mL) was added slowly at 0 °C followed by magnesium sulfate. The resulting mixture was stirred for 1 hour and then filtered and concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 5:1) afforded the title compound as a colorless oil (1.03 g, Yield: 95%). 1 H NMR (400 MHz, CDCI 3 ) 5.67 (t, J = 4.0 Hz, 1 H), 4.1 1 (s, 2H), 2.02 (t, J = 6.2 Hz, 2H), 1.64-1.61 (m, 2H), 1.48-1.45 (m, 2H), 1.13 (bs, 1 H), 1.07 (s, 6.H) ppm.

Example 209d: 6,6-dimethylcyclohex-l-enecarbaldehyde

A stirred solution of the product of Example 209c (1.52 g, 7.67 mmol) and pyridine (1 mL) in dichloromethane (40 mL) at 0 °C was added Dess- artin periodinane (4.66 g, 1 .0 mmol). The reaction was stirred at 0 °C for 30 minutes and then at room temperature for 30 minutes. Water (40 mL) was added to the reaction mixture followed by aqueous sodium hydroxide solution (1.0 M, 45 mL). The bilayer mixture was allowed to stir vigorously for 10 minutes. The layers were separated and the aqueous layer was extracted with diethyl ether (40 mL x 3). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 1 :0 -> 100:1) gave the title compound as a colorless liquid (750 mg, Yield: 49%). H NMR (400 MHz, CDCI 3 ) 9.33 (s, 1H), 6.69 (t, J = 4.0 Hz, 1H), 2.30 (m, 2H), 1.64 (m, 2H), 1.48 (m, 2H), 1.22 (s, 6H) ppm. Example 209: (6,6-dimethylcyclohex-1-enyl)(4-fluorophenyl)methanol

A stirred solution of (4-fluorophenyl)magnesium bromide (0.8 M in tetrahydrofuran) (4.6 mL, 3.63 mmol) cooled to -78 °C was added a solution of the product of Example 209d (200 mg, 1.45 mmol) in dry tetrahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for 1 hour. The mixture was cooled down to 0 °C and saturated aqueous ammonium chloride solution (30 mL) was added slowly and then water (20 mL). The mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1) gave an oil (210 mg) of which a portion (32 mg) was further purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 20:1) to give the title compound as a white solid (24 mg, yield: 46%). Mp = 69-71 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ). δ 7.36-7.32 (m, 2H), 7.01 (t, J = 8.8 Hz, 2H), 5.69 (t, J = 3.8 Hz, 1 H), 5.33 (d, J ~ 2.8 Hz, 1 H), 2.07-2.03 (m, 2H), 1.67- 1.60 (m, 3H), 1.51-1.46 (m, 2H), 1.19 (s, 3H), 0.92 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 217 (M - H 2 0 + H + ).

Example 210: (6,6-dimethy Icy clohex-1 -eny l)(4-fluoropheny l)methanone

A stirred solution of the product of Example 209 (173 mg, 0.74 mmol) in dichloromethane (3 mL) cooled to 0 °C was added Dess-Martin periodinane (628 mg, 1.48 mmol). After 30 min, the reaction was warmed to room temperature and stirred for 1.5 hours. Saturated aqueous sodium bicarbonate (20 mL) was added to quench the reaction and then it was extracted with dichloromethane (30 mL x 3). The combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by column silica gel chromatography (eluent: petroleum ether/ethyl acetate = 200:1) gave a pale yellow oil (124 mg) which was further purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to give the title compound as a pale yellow oil (101 mg, Yield: 59%). R f = 0.7 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ). δ 7.75 (dd, J = 8.8, 5.6 Hz, 2H), 7.08 (t, J = 8.8 Hz, 2H), 6.10 (t, J = 3.8 Hz, 1 H), 2.23-2.19 (m, 2H), 1.76- 1.70 (m, 2H), 1.58-1.55 (m, 2H), 1.26 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 233 (M + H + ). Example 211 : (2-deutero-6,6-dimethylcyclohex-1-enyl)(4-fluorophenyl) methanone

Example 211a: ethyl 2-deutero-2-hydroxy-6,6-dimethylcyclohexane- carboxylate

To a solution of ethyl 2,2-dimethyl-6-oxocyclohexanecarboxylate (1.58 g, 8 mmol) in MeOH (20 mL) was added sodium borodeuteride (500 mg, 1 1.9 mmol) portion-wise at -78°C. The reaction was warmed gradually to -30°C and then quenched by the addition of 5% hydrochloric acid (3 mL). The reaction was concentrated under reduced pressure and the aqueous residue extracted with ethyl acetate (200 mL). The organic layer was washed water (100 mL x 2) and brine (100 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 -> 10:1 ) gave the title compound as a colorless oil (914 mg, Yield: 57%). R f = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 4.24-4.10 (m, 2H), 3.01 (bs, 1 H), 2.38 (s, 1 H), 1.96-1.90 (m, 1 H), 1.84-1.78 (m, 1 H), 1.63-1.60 (m, 1 H), 1.52-1.40 (m, 2H), 1.29 (t, J = 7.2 Hz, 3H), 1.23-1.16 (m, 1 H), 1.07 (s, 3H), 0.99 (s, 3H) ppm.

Example 211b: ethyl- 2-deutero-6,6-dimethylcyclohex-1-enecarboxylate

To a solution of the product of Example 21 1a (909 mg, 4.5 mmol) in dichloromethane (20 mL) at 0°C was added dropwise thionyl chloride (1.31 mL, 18.1 mmol) and then pyridine (4.38 mL, 54.2 mmol) was added dropwise slowly at that temperature. The mixture was warmed to room temperature and stirred for 5 hours. Water (20 mL) was added and the mixture was extracted with dichloromethane (200 mL). The organic layer was washed with saturated aqueous sodium bicarbonate (100 mL x 2) and brine (100 mL x 2) dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1) afforded the title compound as a light yellow oil (560 mg, Yield: 68%). R f = 0.8 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) 6.81 (t, J = 4.0 Hz, 1 H), 4.17 (q, J = 7.2 Hz, 2H), 2.13 (t, J = 6.4 Hz, 2H), 1.64-1.60 (m, 2H), 1.50-1.47 (m, 2H), 1.29 (t, J = 7.2 Hz, 3H), 1.22 (s, 6H) ppm.

Example 211c: (2-deutero-6,6-dimethylcyclohex-1-enyl)methanol

To a stirred solution of the product of Example 211b (552 mg, 3.0 mmol) in dry dichloromethane (44 mL) at -78°C was added diisobutyl aluminum hydride (1.0 M in hexane, 9.0 mL, 9.0 mmol). The reaction was allowed to warm to 0°C gradually over 1.5 hours and then the mixture was diluted with diethyl ether (30 mL). Tetrahydrofuran/water (5/1 , 10 mL) was added slowly at 0°C followed by magnesium sulfate. The resulting mixture was stirred for 1 hour and then filtered and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5:1) afforded the title compound as a colorless oil (294 g, Yield: 69%). R f = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 4.11 (s, 2H), 2.02 (t, J = 6.2 Hz, 2H), 1.64-1.61 (m, 2H), 1.48-1.45 (m, 2H), 1.13 (bs, 1H), 1.07 (s, 6H) ppm.

Example 211d: 2-deutero-6,6-dimethylcyclohex-1-enecarbaldehyde

To a stirred solution of the product of Example 211c (290 mg, 2.05 mmol) and sodium bicarbonate (172 mg, 2.05 mmol) in dichloromethane (15 mL) at 0°C was added Dess-Martin periodinane (1.74 g, 4.1 mmol). The reaction was stirred at 0°C for 30 minutes and then at room temperature for 2 hours. The mixture was concentrated under reduced pressure and the residue purified by flash chromatography (eluent: petroleum ether/ethyl acetate = 100:1) to afford the title compound as a colorless oil (250 mg, yield: 88%). R = 0.8 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) 9.33 (s, 1 H), 2.30 (t, J = 6.2 Hz, 2H), 1.66-1.63 (m, 2H), 1.49-1.46 (m, 2H), .21 (s, 6H) ppm.

Example 211 : (2-deutero-6,6-dimethylcyclohex-1 -enyl)(4-fluoropheny I) methanone

A stirred solution of (4-fluorophenyl)magnesium bromide (0.8 M in tetrahydrofuran) (2.7 mL, 2.15 mmol) cooled to -78 °C was added a solution of the product of Example 211d (120 mg, 0.86 mmol) in dry tetrahydrofuran (5 mL). The reaction mixture was warmed gradually to room temperature and stirred for 2 hours. The mixture was cooled down to 0°C and saturated aqueous ammonium chloride solution (15 mL) was added slowly and then water (10 mL). The mixture was extracted with ethyl acetate (50 mL). The organic phase was washed with water (30 mL x 2), and brine 30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 50:1) afforded the title compound as a colorless oil (144 mg, Yield: 71 %). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) 7.34 (dd, J = 8.4, 5.6 Hz, 2H), 7.01 (t, J = 8.8 Hz, 2H), 5.32 (d, J = 4.0 Hz, 1 H), 2.04 (t, J = 6.2 Hz, 2H), 1.67 (d, J = 4.4 Hz, 1 H), 1.64-1.60 (m, 2H), 1.51-1.46 (m, 2H), 1.19 (s, 3H), 0.92 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 218 (M - H 2 0 + H + ).

Example 212: (2-deutero-6,6-dimethylcyclohex-1-enyl)(3,4- difluorophenyl) methanone

To a mixture of magnesium (104.5 mg, 4.3 mmol) and iodine (5 mg) in tetrahydrofuran (5 mL) at room temperature was added 4-bromo-1 ,2- difluorobenzene (830 mg, 4.3 mmol). The mixture was warmed up to reflux for 3 hour. The reaction was cooled to -78°C then a solution of the product of Example 21 1d (120 mg, 0.86 mmol) in tetrahydrofuran (5 mL) was added and the reaction was allowed to warm to room temperature and stirred for 2 hours. The reaction was quenched with saturated aqueous ammonium chloride and extracted with ethyl acetate (50 mL). The organic layer was washed with water (30 mL x 2) and brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100:1 -> 5:1) afforded the title compound as a white solid (42 mg, Yield: 19%). Mp = 96-98 °C; R, = 0.4 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) 7.23-7.18 (m, 1 H), 7.1 1-7.07 (m, 2H), 5.29 (s, 1 H), 2.03 (t, J = 6.2 Hz, 2H), 1.72 (bs, 1 H), 1.65-1.58 (m, 2H), 1 .51-1.47 (m, 2H), 1.18 (s, 3H), 0.97 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 236 (M - H 2 0 + H + ).

Example 213: (±)-(4aR,9aS)-7-fluoro-1,1,4a-trimethyl-6-

(trifluoromethoxy)-2,3,4,4a-tetrahydro-1H-fluoren-9(9aH)- one

A solution of the product of Example 200 (149 mg, 0.45 mmol) in methansulfonic acid (3 mL) was stirred at 60°C overnight. Saturated aqueous sodium bicarbonate solution (20 mL) was added slowly and then the mixture was extracted with ethyl acetate (30 mL x 3). The organic layer was washed with brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 250:1 ) gave 58 mg of material which was further purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 50:1 ) and then by

Prep-HPLC to give the title compound as a white solid (33 mg, Yield: 22%). Mp = 53-54 °C; R f = 0.7 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ). δ 7.47 (d, J = 8.4 Hz, 1 H), 7.34-7.32 (m, 1 H), 2.22 (s, 1 H), 2.01- 1.98 (m, 1 H), 1.74-1.63 (m, 2H), 1.48-1.39 (m, 3H), 1.29 (s, 3H), 1.22 (s, 3H), 0.68 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 331 (M + H + ). Example 214: (±)-(4aR,9 ,9aS)-7-fluoro-9-hydroxy-1,1,4a-trimethyl- 2,3,4,4a,9,9a-hexahydro-1H-fluorene-6-carbonitrile

To a solution of the product of Example 207 (70 mg, 0.26mmol) in methanol (5 mL) at 0°C was added sodium borohydride (1 1.4 mg, 0.31 mmol). The reaction mixture was stirred for 30 minutes before it was slowly warmed to room temperature. Water (5 mL) was added to the mixture then it was extracted with ethyl acetate (20 mL x 3). The organic phase was washed with water (20 mL) and brine (20 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a white solid (55 mg, Yield: 78% yield). Mp = 187.2-188.1 °C. R, = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.33 (d, J = 5.6 Hz, 1 H), 7.22 (d, J = 8.4 Hz, 1 H), 5.03 (t, J = 8.4 Hz, 1 H), 1.87 (d, J = 7.6 Hz, 1 H), 1.73 (d, J = 9.2 Hz, 1 H), 1.68-1.58 (m, 2H), 1.48 (s, 3H), 1.47-1.32 (m, 3H), 1.19 (s, 3H), 1.16 (s, 3H), 0.99-0.92 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 274 (M + H + ).

Example 215: 4-((3,3-dimethylcyclohex-1- enyl)(hydroxy)methyl)benzonitrile

To a solution of 4-bromobenzonitrile (790.2 mg, 4.34 mmol) in dry tetrahydrofuran (16 mL) at -78°C under argon was added n-butyl lithium (2.71 mL, 4.34 mmol). After stirring at -78°C for 30 minutes the product of Example 153d was added. The reaction was allowed to warm gradually to room temperature and stirred for 2 hours. The mixture was quenched by the addition of saturated aqueous ammonium chloride (20 mL) and the solution was extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 70:1 -> 20.1 ) to give the title compound as a white solid (1 10 mg, Yield: 10%). Mp = 93-95 °C. R, = 0.3 (6:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.62 (d, J = 8.8 Hz, 2 H), 7.46 (d, J = 8.0 Hz, 2 H), 5.61 (s, 1 H), 5.12 (d, J - 3.2 Hz, 1 H), 1.91 (d, J = 3.2 Hz, 1 H), 1.86-1.81 (m, 1 H), 1.60-1.53 (m, 3 H), 1.45-1.32 (m, 2 H), 1.01 (s, 3 H), 1.01 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 242 (M + H + ).

Example 216: 4-(3,3-dimethylcyclohex-1-enecarbonyl)benzonitrile

To a solution of the product of Example 215 (45 mg, 0.19 mmol) in dichloromethane (1.5 mL) at 0 °C was added sodium bicarbonate (16.0 mg, 0.19 mmol) and Dess-Martin periodinane (156.9 mg, 0.37 mmol). The mixture was stirred warmed to room temperature and stirred for .5 hours. Saturated aqueous sodium bicarbonate (10 mL) was added and then the mixture was extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a white solid (30 mg, Yield 66 %). Mp = 77-79 °C. R f = 0.3 (6:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.74-7.72 (dd, J = 6.4, 2.0 Hz, 2 H), 7.68-7.66 (dd, J = 6.4, 1.6 Hz, 2 H), 6.22 (s, 1 H), 2.36 (t, J = 6.4 Hz, 2 H), 1.77-1.71 (m, 2 H), 1.55-1.51 (m, 2 H), 1 .06 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 240 (M + H + ).

Example 217: 4-((3,3-dimethylcyclohex-1-enyl)(hydroxy)methyl)-2- fluorobenzonitrile

To a stirred solution of 4-bromo-2-fluorobenzonitrile (300 mg, 1.50 mmol) in tetrahydrofuran (6 mL) at 0°C was added a solution of isopropylmagnesium chloride (0.75 mL, 1.50 mmol) in tetrahydrofuran. The mixture was warmed to room temperature and stirred at for 1 hour. Then a solution of the product of Example 153d (159.5 mg, 1.15 mmol) in tetrahydrofuran (1 mL) was added and the mixture was stirred for 1 hour. Hydrochloric acid (0.1 N, 10 ml) was added to quench the reaction and then the organics were extracted with ethyl acetate (10 mL x 3). The combined organic phase was washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1 -> 10:1) to afford the title compound as a white solid (160 mg, Yield: 54%). Mp = 81-82.1 °C. R f = 0.2 (6:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.50 (t, J = 7.2 Hz, 1 H), 7.20-7.15 (m, 2H), 5.55 (s, 1 H), 5.05 (s, 1 H), 1.84-1.70 (m, 1 H), 1.54-1.44 (m, 4H), 1.35-1.28 (m, 2H), 0.95(s, 3H) , 0.94 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 260 (M + H + ).

Example 218: 4-(3,3-dimethylcyclohex-1-enecarbonyl)-2- fluorobenzonitrile

To a stirred solution of compound the product of Example 217 (130.0 mg, 0.50 mmol) in dichloromethane (6 mL) at 0 °C was portionwise added Dess-Martin periodinane (383.2 mg, 0.90 mmol). The mixture was warmed to room temperature and stirred for 1.5 hours. Additional dichloromethane (20 mL) was added and the organic phase was washed with aqueous saturated sodium bicarbonate and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to afford the title compound as a white solid (100 mg, Yield: 78%). Mp = 78.5- 79.1 °C; R = 0.6 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.69 (t, J = 8.0 Hz, 1 H), 7.46-7.40 (m, 2H), 6.24 (s, 2H), 2.36-2.33 (m, 2H), 1.76-1.73 (m, 2H), 1.66-1.51 (m, 2H), 1.07 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 258 (M + H + ).

Example 219: (4-chlorophenyl)(3,3-dimethylcyclohex-1-enyl)methanol To a solution of 4-chlorophenylmagnesium bromide (22.97 mL, 22.79 mmol) in dry tetrahydrofuran (5 mL) at -78 °C under argon was slowly added the product of Example 153d (700 mg, 5.06 mmol). The reaction was stirred at -78 °C for 2 hours. The reaction was quenched by the addition of saturated aqueous ammonium chloride (20 mL) and after warming to room temperature the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 70:1 -> 40:1 ) to afford the title compound as a colorless oil (650 mg, Yield: 51%). R f = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.31-7.26 (m, 4 H), 5.60 (s, 1 H), 5.04 (s, 1 H), 1.84-1.78 (m, 2 H), 1.67-1.63 (m, 1 H), 1.61-1.53 (m, 2 H), 1.42-1.34 (m, 2 H), 1.01 (s, 3 H), 1.00 (s, 3 H) ppm.

Example 220: (4-chlorophenyl)(3,3-dimethylcyclohex-1-enyl)methanone

To a solution of the product of Example 219 (60 mg, 0.24 mmol) in dichloromethane (1.5 mL) at 0 °C was added sodium bicarbonate (20.2 mg, 0.24 mmol) and Dess-Martin periodinane (202.9 mg, 0.48 mmol). The mixture was warmed to room temperature and stirred for 1.5 hours. Saturated aqueous sodium bicarbonate (10 mL) was added and then the reaction mixture was extracted with dichloromethane. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a white solid (43 mg, Yield: 72%). Mp = 82-84 °C; R = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.58 (d, J = 8.4 Hz, 2 H), 7.40 (d, J = 8.4 Hz, 2 H), 6.21 (s, 1 H), 2.37-2.33 (m, 2 H), 1.75-1.72 (m, 2 H), 1.53-1.50 (m, 2 H), 1 .06 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 249 (M + H + ).

Example 221: (±)-(4aR,9R,9aS)-7-fluoro-1 ,1,4a-trimethyl-6-

(trifluoromethoxy) 2,3,4,4a,9,9a-hexahydro-1H-fluoren-9-ol

To a stirred solution of the product of Example 213 (23 mg, 0.07 mmol) in methanol (2 mL) at room temperature was added sodium borohydride (22 mg, 0.56 mmol). The reaction was stirred at room temperature for 15 min and then water (20 mL) was added. The mixture was extracted with ethyl acetate (30 mL x 3) and the combined organic phase was washed with brine (20 mL x 2), dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 200:1 -> 25: 1) gave the title compound as a white solid

(21 mg, Yield: 90%). Mp = 91-93 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 +D 2 0) δ 7.21 (d, J = 9.2 Hz, 1 H), 7.01 (d, J = 6.8 Hz, 1 H), 4.98 (d, J = 9.2 Hz, 1 H), 1.71-1.31 (m, 5H), 1.18 (s, 3H), 1.16 (s, 3H), 1.01-0.93 (m, 1 H) ppm; Mass spectrum (ESI +ve) m/z 315 (M - H 2 0 + H + ).

Example 222: (±)-(4aR,9aR)-6-Fluoro-1 ,1 -dimethyl-2,3,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one

A mixture of the product of Example 210 (29 mg, 0.13 mmol) in methansulfonic acid (3 mL) was stirred at 50 °C for 5 hours. Saturated aqueous sodium bicarbonate (20 mL) was added slowly and then the mixture was extracted with ethyl acetate (30 mL x 3). The organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 50:1) gave the title compound as a white solid (16 mg, Yield: 55%). Mp = 49-51 °C; R f = 0.6 (20:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDC ): δ 7.69 (dd, J = 8.4, 5.6 Hz, 1 H), 7.08 (dd, J = 8.4, 2.0 Hz, 1 H), 7.03 (dt, J = 8.4, 2.0 Hz, 1 H), 3.42-3.36 (m, 1 H), 2.51 (d, J = 6.8 Hz, 1 H), 2.05-1.96 (m, 1 H), 1.65-1.35 (m, 5 H), 1.15 (s, 3 H), 1.01 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 233 (M + H + ). Example 223: (2-Deutero-6,6-dimethylcyclohex-1-enyl)(4-fluorophenyl) methanone

To a stirred solution of the product of Example 211 (130 mg, 0.55 mmol) in dry dichloromethane (5 mL) at 0 °C was added sodium bicarbonate (46 mg, 0.55 mmol) and Dess-Martin periodinane (466 mg, 1.10 mmol). The mixture was stirred at room temperature for 2 hours and then the mixture was concentrated under reduced pressure. The residue was purified by flash chromatography (elue nt: petroleum ether/ethyl acetate = 100:1) to give a colorless oil (90 mg) that was further purified by preparative thin layer chromatography to give the title compound as a colorless oil (79 mg, Yield: 62%). Rf = 0.5 (50:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI 3 ): δ 7.75 (dd, J = 8.8, 5.6 Hz, 2 H), 7.08 (t, J = 8.8 Hz, 2 H), 2.20 (t, J = 6.4 Hz, 2 H), 1.74-1.71 (m, 2 H), 1.59-1.55 (m, 2 H), 1.26 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 234 (M + H + ).

Example 224: (±)-(4aS,9aR)-4a-Deutero-6-fluoro- ,1 -dimethyl-2,3,4,4a- tetrahydro-1 H-f luoren-9(9aH)-one

A stirred solution of the product of Example 223 (66 mg, 0.28 mmol) in dry methanesulfonic acid (2 ml_) was stirred at 50 °C for 7 hours. Water (20 mL) was added and then the mixture was extracted with ethyl acetate (50 mL). The organic phase was washed with water (30 mL x 2) and brine (30 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 50:1) to afford the title compound as a white solid (38 mg, Yield: 58%). Mp = 54.3-55.4 °C; R/ = 0.4 (50:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI3): δ 7.69 (dd, J = 8.4, 5.6 Hz, 1 H), 7.07 (dd, J = 8.4, 2.4 Hz, 1 H), 7.02 (dt, J = 8.8, 2.4 Hz, 1 H), 2.50 (s, 1 H), 2.03-1.98 (m, 1 H), 1.62-1.35 (m, 5 H), 1.15 (s, 3 H), 1.01 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 234 (M + H + ).

Example 225: (2-Deutero-6,6-dimethylcyclohex-1-enyl)(3,4- difluorophenyl) methanone

To a stirred solution of the product of Example 212 (85 mg, 0.34 mmol) in dry dichloromethane ( 4 mL) at 0 °C was added sodium bicarbonate (29 mg, 0.34 mmol) and Dess-Martin periodinane (288 mg, 0.68 mmol). The mixture was stirred at room temperature for 2 h. TLC showed the reaction was completed. The mixture solution was concentrated to load onto a silica gel column and purified by flash chromatography (eluent: petroleum ether/ethyl acetate = 100:1 ) to give a colorless oil (61 mg) which was further purified by preparative thin layer chromatography to afford the title compound as a colorless oil (53 mg, Yield: 63%). = 0.5 (50:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI 3 ): δ 7.59-7.53 (m, 1 H), 7.52-7.48 (m, 1 H), 7.22-7.16 (m, 1 H), 2.22 (t, J = 6.4 Hz, 1 H), 1.75-1.69 (m, 2 H), 1.59-1.55 (m, 2 H), 1.25 (s, 6 H) ppm; Mass spectrum (ESI +ve) m/z 252 (M + H + ).

Example 226: (±)-(4aS,9aS)-6-Chloro-4,4-dimethy 1-2,3 ,4,4a-tetrahydro-1 H- fluoren-9(9aH)-one

The product of Example 220 (250 mg, 1.0 mmol) was dissolved in methansulfonic acid (8 mL) and the reaction solution was stirred at 65 °C overnight. After the reaction was cooled to room temperature was added H20 (30 mL) was added and the mixture was extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to afford the title compound as a yellow solid (63 mg, Yield 25%). Mp = 67-69 °C; R, = 0.35 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.69 (d, J = 8.0 Hz, 1 H), 7.50 (d, J = 1.6 Hz, 1 H), 7.37-7.35 (m, 1 H), 3.15 (d, J = 10.8 Hz, 1 H), 2.70- 2.66 (m, 1 H), 2.47-2.42 (m, 1 H), 1.56-1.49 (m, 2 H), 1.44-1.33 (m, 3 H), 1.17 (d, J = 4.0 Hz, 3 H), 0.13 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 249 (M + H + ). Example 227: (±)-(4aS,9a/?)-4a-Deutero-6,7-difluoro-1 ,1-dlmethyl-2,3,4,4a- tetrahydro-1 H-fluoren-9(9a W)-one

A stirred solution of the product of Example 225 (43 mg, 0.17 mmol) in dry methanesulfonic acid (2 mL) was stirred at 50 °C for 2.5 hours. Water (20 mL) was added and the mixture was extracted with ethyl acetate (50 mL). The organic layer was washed with water (30 mL χ 2), brine (30 mL * 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (eluent petroleum ether/ethyl acetate = 50:1 ) to give a white solid (28 mg) that was further purified by Prep-HPLC to give the title compound as a white solid ( 5.6 mg, Yield: 36%). Mp = 58.4-59.5 °C; R, = 0.4 (50:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI3): δ 7.47 (dd, J = 8.8, 7.6 Hz, 1 H), 7.20 (dd, J = 9.2, 6.4 Hz, 1 H), 2.49 (s, 1 H), 2.05-1.98 (m, 1 H), 1.63-1.46 (m, 2 H), 1.38-1.34 (m, 3 H), 1.15 (s, 3 H), 1.03 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 252 (M + H + ).

Example 228: (3,4-Difluorophenyl)(6,6-dimethylcyclohex-1-enyl)methanol To a suspension of magnesium (120 mg, 4.92 mmol) in dry tetrahydrofuran (4 mL) under argon was added a solution of 4-bromo-1 ,2- difluorobenzene ( 50 mg, 0.98 mmol) in dry tetrahydrofuran (2 mL) and iodine (5 mg). The mixture was gradually warmed to 50 °C and then additional 4- bromo-1 ,2-difluorobenzene (800 mg, 3.94 mmol) in dry THF (8 mL) was added slowly and after the addition was complete the mixture was stirred at reflux for 1 hour. The solution was cooled to -78 °C and a solution of the product of Example 209d (170 mg, 1.23 mmol) in dry tetrahydrofuran (2 mL) was added. The reaction was warmed and stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride (10 mL) followed by water (20 mL) was added and the mixture was extracted with ethyl acetate (20 ml x 3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gei column chromatography (eiuent: petroleum ether/ethyl acetate = 100:1) gave a white solid (57 mg) that was further purified by prep-HPLC to afford the title compound as a white solid (40 mg, Yield: 13%). Mp = 97-98 °C; R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 HNMR (400 MHz, CDCI 3 ): δ 7.26-7.17 (m, 1 H), 7.11-7.07 (m, 2 H), 5.64 (t, J = 4.0 Hz, 1 H), 5.29 (d, J = 4.8 Hz, 1 H), 2.06-2.01 (m, 2 H), 1.68 (d, J = 4.4 Hz, 1 H), 1.65-1.58 (m, 2 H), 1.54-1.47 (m, 2H), 1.18 (s, 3 H), 0.97 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 235 (M - H 2 0 + H + ).

Example 229: (+)-(4aS,9S,9aS)-6-chloro-4,4-dimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 226 (65 mg, 0.26 mmol) in methanol (2.5 mL) 0 °C was added sodium borohydride (29.7 mg, 0.78 mmol) in portions. The reaction was warmed to room temperature and stirred for 2 hours. The reaction was quenched by the addition of water (5 mL) and then it was concentrated under reduced pressure. The residue was extracted with ethyl acetate and the organic phase was dried over anhydrous sodium sulfate and then it was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 60:1) to afford the title compound as a white solid (38 mg, Yield: 58%). R f = 0.2 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.48 (s, 1 H), 7.32-7.30 (m, 1 H), 7.21-7.19 (m, 1 H), 4.95 (t, J = 6.8 Hz, 1 H), 2.78 (d, J - 5.6 Hz, 1 H), 2.72-2.65 (m, 1 H), 1.71 (d, J = 8.0 Hz, 1 H), 1.64-1.59 (m, 1 H), 1.55-1.50 (m, 2 H), 1.43-1.29 (m, 2 H), 1.17 (s, 3 H), 1.15-1.08 (m, 4 H) ppm; Mass spectrum (ESI +ve) m/z 233 (M - H 2 0 + H + ).

Example 230: (6,6-dimethylcyclohex-1-enyl)(4-methoxyphenyl)methanol

To a solution of 4-methoxyphenylmagnesium bromide ( 17.6 ml_, 8.8 mmol) under argon at -78 °C was slowly added the product of Example 209d (270 mg, 1.95 mmol). The reaction was warmed to room temperature and stirred for 2 hours. The mixture was quenched by the addition of saturated aqueous ammonium chloride (25 ml_). The reaction mixture was extracted with ethyl acetate and the organic phase was dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 40:1 -> 20:1) to afford the title compound as a colorless oil (350 mg, Yield: 73%). R f = 0.2 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI 3 ) δ 7.31-7.26 (m, 2 H), 6.87-6.84 (m, 2 H), 5.75 (t, J = 3.8 Hz, 1 H), 5.29 (d, J = 3.6 Hz, 1 H), 3.80 (s, 3 H), 2.08-2.04 (m, 2 H), 1.66-1.59 (m, 3 H), 1.50-1.45 (m, 2 H), 1.18 (s, 3 H), 0.88 (s, 3 H) ppm; Mass spectrum (ESI +ve) m/z 229 (M - H 2 0 + H + ).

Example 231 : (3,3-dimethylcyclohex-1-enyl)(4-methoxyphenyl)methanol

To a solution of the product of Example 153d (500 mg, 3.6 mmol) in tetrahydrofuran (10 mL) at -78 °C was added (4-methoxyphenyl)magnesium bromide solution (10.8 mL, 0.5 M solution in . tetrahydrofuran). The reaction mixture was stirred for 1 hour before it was slowly warmed to 0 °C and saturated aqueous ammonium chloride was then added to quench the reaction. The mixture was extracted with ethyl acetate (20 mL x 3) and the combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure. The residue was purified by flash column chromatography to afford the title compound as a colorless oil (786 mg, Yield: 89%). R = 0.3 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI3) δ 7.25 (dd, J = 2.8, 8.8 Hz, 2H), 6.89-6.85 (m, 2H), 5.60 (s, 1 H), 5.00 (d, J = 3.2 Hz, 1 H), 3.80 (s, 3H), 1.82-1.76 (m, 2H), 1.72-1.64 (m, 1 H), 1.58-1.54 (m, 2H), 1.43-1.36 (m, 2H), 1.02 (s, 3H), 1.01 (s, 3H) ppm.

Example 232: (3,3-dimethylcyclohex-1-enyl)(4-methoxyphenyl) methanone

The solution of the product of Example 231 (150 mg, 0.61 mmol) in dichloromethane (10 mL) was added Dess-Martin periodinane (517 mg, 1.22 mmol) and sodium bicarbonate (51.24 mg, 0.61 mmol). The mixture was stirred at room temperature for 30 minutes. The reaction mixture was quenched with 2N hydrochloric acid (5 mL) and then the mixture was extracted with dichloromethane (20 mL x 3) and the combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a colorless oil (100 mg, Yield: 67%). R f = 0.8 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.69 (dd, J = 2.0, 6.8 Hz, 2H), 6.92 (dd, J = 2.0, 7.2 Hz, 2H), 6.17 (s, 1 H), 3.87 (s, 3H), 2.37-2.33 (s, 2H), 1.75-1.71 (m, 2H), 1.53-1.50 (m, 2H), 1.07 (s, 6H) ppm.

Example 233: (±)-(4aS,9aS)-6-methoxy-4,4-dimethyl-2,3,4,4a-tetrahydro- 1 H-fluoren-9(9aH)-one

A solution of the product of Example 232 (100 mg, 0.41 mmol) in methansulfonic acid (4.0 mL) was stirred at 80 °C overnight. The reaction solution was poured onto ice and the resulting mixture was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash column chromatography to afford the title compound as a white solid (160 mg, Yield: 64%). Hf = 0.7 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.70 (d, J = 8.4 Hz, 1 H), 6.96 (d, J = 2.4 Hz, 1 H), 6.90 (dd, J = 2.0, 8.4 Hz, 1 H), 3.89 (s, 3H), 3.11 (d, J = 7.2 Hz, 1 H), 2.67 (t, J = 7.6 Hz, 1 H), 2.45-2.40 (m, 1 H)„ 1.56-1.47 (m, 2H), 1.43-1.32 (m, 3H), 1.17 (s, 3H), 0.14 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 245 (M + H + ). Example 234: (3,3-dimethylcyclohex-1-enyl)(o-tolyl)methanol

To a stirred solution of o-tolylmagnesium bromide (1.0 M in tetrahydrofuran, 2.18 mL, 2.18 mmol) cooled down to -78 °C was added slowly a solution of the product of Example 153d (200 mg, 1.45 mmol) in dry terahydrofuran (10 mL). The reaction mixture was warmed gradually to room temperature and stirred for 1.5 hours. Saturated aqueous ammonium chloride (10 mL) was added slowly to quench the reaction. Water (20 mL) was added and then the mixture was extracted with ethyl acetate (50 mL x 3). The combined organic phase was washed with brine (50 mL x 2), dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1) gave the title compound as a colorless oil (333 mg, Yield: 100%). R f = 0.4 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.45 (d, J = 7.8 Hz, 1 H), 7.23-7.1 1 (m, 3H), 5.54 (s, 1 H), 5.23 (d, J = 2.8 Hz, 1 H), 2.29 (s, 3H), 1.89-1.82 (m, 1 H), 1.72-1.68 (m, 2H), 1.65-1.57 (m, 2H), 1.45-1.33 (m, 2H), 1 .00 (s, 3H), 0.98 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 213 (M - H 2 0 + H + ).

Example 235: (3,3-dimethylcyclohex-1-enyl)(o-tolyl)methanone

A stirred solution of the product of Example 234 (339 mg, 1.47 mmol) in dichloromethane (3 mL) cooled to 0 °C was added Dess-Martin periodinane (1.25 g, 2.94 mmol). After 30 minutes the reaction was warmed to room temperature and stirred for 1 hour. Saturated aqueous sodium bicarbonate (20 mL) was added slowly and then the reaction mixture was extracted with dichloromethane (30 mL x 3). The combined organic phase was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 50:1 ) gave the title compound as a colorless oil (241 mg, Yield: 71 %), R f = 0.5 (20:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI 3 ) δ 7.33-7.29 (m, 1 H), 7.21- 7.17 (m, 3H), 6.15 (s, 1 H), 2.37-2.32 (m, 2H), 2.26 (s, 3H), 1.75-1.69 (m, 2H), 1.50-1.47 (m, 2H), 1.00 (s, 6H) ppm; Mass spectrum (ESI +ve) m/z 229 (M + H + ).

Example 236: (+)-(4aS,9aS)-4,4,8-trimethyl-2,3 l 4,4a-tetrahydro-1 H- fluoren-9(9aH)-one

A solution of the product of Example 235 (233 mg, 1.02 mmol) in methansulfonic acid (3 mL) was stirred at 50 °C overnight. Saturated aqueous sodium bicarbonate (20 mL) was added slowly and then the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (30 mL x 2), dried over anhydrous sodium sulfate , and then concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 80:1) to afford the title compound as a colorless colorless oil (25 mg, Yield: 11%). R f = 0.5 (50:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCb) δ 7.38 (t, J = 7.4 Hz, 1 H), 7.30 (d, J = 7.6 Hz, 1 H), 7.10 (d, J = 7.2 Hz, 1 H), 3.1 1 (d, J = 7.2 Hz, 1 H), 2.66-2.63 (m, 1 H), 2.63 (s, 3H), 2.44 (d, J = 13.2 Hz, 1 H), 1.55-1.47 (m, 2H), 1.42-1.32 (m, 3H), 1.15 (s, 3H), 0.10 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 229 (M + H + ).

Example 237: (±)-(4aS,9aS)-4,4-dimethyl-9-oxo-2, 3,4,4a, 9, 9a-hexahydro- IW-fluorene-6-carbonitrile Example 237a: (±)-(4aS,9aS)-6-hydroxy-4,4-dimethyl-2,3,4,4a-tetrahydro- 1W-fluoren-9(9aW)-one To a solution of the product of Example 233 (1.1 g, 0.45 mmol) in dichloromethane (2.0 mL) at -78 °C was added boron tribromide solution (2.25 mL, 1.0 M solution in dichloromethane). The reaction mixture was stirred at -78 °C for 30 minutes and then slowly warmed to room temperature. After 12 hours the dark solution was poured onto ice and the mixture was extracted with dichloromethane (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by flash chromatography afforded the title compound as a yellow solid (45 mg, 43% Yield), Rf = 0.2 (10:1 petroleum ether/ethyl acetate); Mass spectrum (ESI +ve) m/z 231 (M + H + ).

Example 237b: (±)-(4aS,9aS)-4,4-dimethyl-9-oxo-2, 3,4,4a, 9,9a-hexahydro- 1 W-fluoren-6-yl trifluoromethanesulfonate

To a solution of the product of Example 237a (40 mg, 0.17 mmol) and triethylamine (26 mg) in dichloromethane (5 mL) at -20 °C was added trifluoromethansulfonic anhydride (73 mg, 0.26 mmol). The reaction mixture was stirred for 30 minutes and then was siowiy warmed to room temperature. The organic phase was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound as a yellow oil (70 mg, Yield: 100% yield). Rf = 0.8 (10:1 petroleum ether/ethyl acetate); Mass spectrum (ESI +ve) m/z 363 (M + H + ). Example 237: (±)-(4aS,9aS)-4,4-dimethyl-9-oxo-2,3,4,4a,9,9a-hexahydro- 1 W-fluorene-6-carbonitrile

A mixture of the product of Example 237b (70 mg, 0.19 mmol) dicyanozinc (30 mg, 0.25 mmol) and palladium tetrakistriphenylphosphine (23 mg, 0.02 mmol) in dimethylformamide/water (4 mL /1 mL) was stirred at 100 °C overnight under argon. Saturated aqueous ammonium chloride was added to the mixture, then it was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (10 mg, Yield: 17%). = 0.5 (10:1 petroleum ether/ethyl acetate); H NMR (400 MHz, CDCI3) δ 7.85 (d, J = 7.6 Hz, 1 H), 7.81 (s, 1 H), 7.67 (dd, J = 1.2 Hz 8.0 Hz, 1 H), 3.23 (d, J = 7.2 Hz, 1 H), 2.71 (t, = 7.2 Hz, 1H), 2.46 (dd, J = 2.8 Hz 16.0 Hz, 1 H), 1.61-1.51 (m, 2H), 1.46-1.35 (m, 3H), 1.18 (s, 3H), 0.07 (s, 3H) ppm; Mass spectrum (ESI +ve) m/z 240 (M + H + ) ppm.

Example 238: (±)-(4aS,9aS)-6,7-difluoro-4,4-dimethyl-2,3,4,4a-tetrahydro - 1//-fluoren-9(9aW)-one

The solution of the product of Example 185 (50 mg, 0.2 mmol) in methansulfornic acid (2 ml) was stirred at 50 °C overnight. The reaction mixture was quenched with water (5 mL) and then it was extracted with ethyl acetate (20 mL x 3), washed with saturated aqueous sodium bicarbonate (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford the title compound as a white solid (25 mg, Yield: 50%). R f = 0.8 (10:1 petroleum ether/ethyl acetate); 1 H NMR (400 MHz, CDCI. 3 ) δ 7.57-7.53 (m, 1 H), 7.32-7.28 (m, 1 H), 3.13 (d, J = 7.2Hz, 1 H), 2.68 (t, J = 7.2 Hz, 1 H), 2.43 (dd, J = 2.4, 15.6 Hz, 1 H), 1.55-1.49 (m, 2H), 1 .44-1 .33 (m, 3H), 1.15 (s, 3H), 0.12 (s, 3H) ppm.

Example 239: (±)-(4aS,9S,9aS)-6,7-difluoro-4,4-dimethyl-2,3,4,4a,9,9a- hexahydro-1 H-fluoren-9-ol

To a solution of the product of Example 238 (34 mg, 0.136 mmol) in methanol (5 mL) at room temperature was added sodium borohydride (1 1 mg, 0.272 mmol) and the mixture was stirred overnight. The reaction mixture was quenched with water (5 mL) and then it was extracted whit ethyl acetate (20 ' mL x 3) and the combined organic phase was washed with saturated aqueous sodium bicarbonate (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography to give the title compound as a white solid (10 mg, Yield: 30%). R f = 0.3 (10:1 petroleum ether/ethyl acetate); H NMR.(400 MHz, CDCI 3 ) δ 7.31-7.29 (m, 1H), 7.17 (t, J = 8.8 Hz, 1 H), 4.93 (t, J = 6.0 Hz, 1H), 2.75-2.67 (m, 2H), 1.72 (d, J = 7.6 Hz, 1 H), 1.65-1.60 (m, 1 H), 1.52-1.48 (m, 2H), 1.35-1.30 (m, 2H), 1.25-1.19 (m, 1H), 1.14 (s, 3H), 1.12 (s, 3H) ppm.

Biology Examples

In carrying out the procedures of the present invention it is of course to be understood that reference to particular buffers, media, reagents, cells, culture conditions and the like are not intended to be limiting, but are to be read so as to include all related materials that one of ordinary skill in the art would recognize as being of interest or value in the particular context in which that discussion is presented. For example, it is often possible to substitute one buffer system or culture medium for another and still achieve similar, if not identical, results. Those of skill in the art will have sufficient knowledge of such systems and methodologies so as to be able, without undue experimentation, to make such substitutions as will optimally serve their purposes in using the methods and procedures disclosed herein.

The invention is described in more detail in the following non-limiting examples. It is to be understood that these particular methods and examples in no way limit the invention to the embodiments described herein and that other embodiments and uses will no doubt suggest themselves to those skilled in the art.

Reagents

Monoclonal anti-rhodopsin 1 D4 antibody can be purchased from University of British Columbia. Cell lines and culture conditions

Stable cell lines expressing opsin protein were generated using the Flp-ln T-Rex system. The stable cells were grown in DMEM high glucose media supplemented with 10% (vlv) fetal bovine serum, antibiotic/antimycotic solution, 5 μ/ml blasticidin and hygromycin at 37 °C in presence of 5% C0 2 . For all the experiments the cells were allowed to reach confluence and were induced to produce opsin with 1 μg/ml tetracycline after change of media and then compounds were added. The plates were incubated for 48 hours after which the cells were harvested.

SDS-PAGE and western blotting Proteins were separated on SDS-PAGE gels and western blotted as described in (Noorwez et al., J. Biol. Chem. 279,16278-16284 (2004)).

The in vivo efficacy of the compounds of the invention in treating macular degeneration can be demonstrated by various tests well known in the art. For example, human patients are selected based on a diagnosis of macular degeneration (such as where there is a gross diagnosis of this condition or where they have been shown to exhibit build-up of toxic visual cycle products, such as A2E, lipofuscin, or drusen in their eyes. A compound of the invention, such as that of Formula I, is administered to a test group while a placebo, such as PBS or DMSO, is administered to a control group that may be as large or may be somewhat smaller than the test group. The test compound is administered either on a one time basis or on a sequential basis (for example, weekly or daily) or according to some other predetermined schedule.

Administration of the test compound is normally by oral or parenteral means and in an amount effective to retard the development and/or reoccurrence of macular degeneration. An effective dose amount is generally in the range of about 1 to 5,000 mg or in the range of 10 to 2,000 mg/kg. Administration may include multiple doses per day.

Efficacy of the test compound in retarding progression of macular degeneration is generally by measuring increase in visual acuity (for example, using Early Treatment Diabetic RP Study (ETDRS) charts (Lighthouse, Long Island, N.Y.). Other means of following and evaluating efficacy is by measuring/monitoring the autofluorescence or absorption spectra of such indicators as N-retinylidene-phosphatidylethanolamine, dihydro-N- retinylidene-N-retinyl-phosphatidylethanolamine, N-retinylidene-N-retinyl- phosphatidylethanolamine, dihydro-N-retinylidene-N-retinyl-ethanolamine, and/or N-retinylidene-phosphatidylethanolamine in the eye of the patient. Autofluorescence is monitored using different types of instrument, for example, a confocal scanning laser ophthalmoscope.

Accumulation of lipofuscin in the retinal pigment epithelium (RPE) is a common pathological feature observed in various degenerative diseases of the retina. A toxic vitamin A-based fluorophore (A2E) present within lipofuscin granules has been implicated in death of RPE and photoreceptor cells. Such experiments can employ an animal model which manifests accelerated lipofuscin accumulation to evaluate the efficacy of a therapeutic approach based upon reduction of serum vitamin A (retinol). Administration of test compound to mice harboring a null mutation in the Stargardt's disease gene (ABCA4) produces reductions in serum retinol/retinol binding protein and arrested accumulation of A2E and lipofuscin autofluorescence in the RPE.

Test animals are available for use in testing efficacy of a test compound in reducing build-up of toxic pigments, such as lipofuscin. For example, mice have been produced that exhibit increased production of such toxic product. Such mice have been described in the literature (see, for example, Widder et al., U.S. Pub. 2006/0167088) and their value and utility are well known to those in the art. Alternatively, the efficacy of the identified compound may be assayed in an animal model of autosomal dominant retinitis pigmentosa (adRP) that utilizes a transgenic mouse line, huP23H(+):rho(+/+), or huP23H(+):rho(+/-), both on a C57BI/6 background. These mice express the human class II misfolding rhodopsin P23H mutant gene that is under the control of the normal mouse rhodopsin gene transcriptional control elements (Mao et al., Human Gene Therapy, March 2011). The transgene is not inserted at the normal mouse rhodopsin locus, but rather elsewhere in the genome. Such efficacy was shown for compound 3a using this model.

Showing the efficacy of compounds of the invention in protecting against light toxicity is conveniently performed by methods well known in the art (see, for example, Sieving et al, PNAS, Vol. 98, pp 1835-40 (2001)). Biology Example 1

Rhodopsin Purification and Regeneration

P23H cell opsin expression is induced 48 hrs in the presence of DMSO (blank) or various concentrations of test compound (generally 1 to- 40 μΜ). P23H opsin producing cells are washed with PBS and lysed in cold PBS-D for 1 hour. The lysate is cleared by centrifugation and added to 1 D4-coupled sepharose beads and incubated for 1 hour at 4°C. Opsin was eluted from the antibody beads with a competing peptide corresponding to the last 18 amino acids of rhodopsin in the same buffer. The purified opsin is immediately used for rhodopsin regeneration studies using 9-cis retinal as chromophore. Opsin («25 μΜ) is mixed with 10 μΜ 9-c/s retinal and the absorbance is determined over the range of 250-650 nm every two minutes in a Cary 50 spectrophotometer (Varian) until no more rhodopsin is regenerated as measured by the increase in 480-500 nm absorbance. Table 1 contains the results of β-ionone (reference compound 1) and test compounds in which the 480-500 nm absorbance is expressed as a fold increase over the DMSO control. Figure 2 is the spectral results using reference compound 1 according to Biology Example 1. Table 1

Other Embodiments

From the foregoing description, it will be apparent that variations and modifications may be made to the invention described herein to adopt it to various usages and conditions. Such embodiments are also within the scope of the following claims.

The recitation of a listing of elements in any definition of a variable herein includes definitions of that variable as any single element or combination (or subcombination) of listed elements. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

All patents and publications mentioned in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference.