Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HIGH CONCENTRATION FORMULATION
Document Type and Number:
WIPO Patent Application WO/2016/207778
Kind Code:
A1
Abstract:
The present disclosure provides high concentration formulations of cortexolone-17α-propionate suitable for treating alopecia.

Inventors:
LONGO LUIGI MARIA (IT)
Application Number:
PCT/IB2016/053662
Publication Date:
December 29, 2016
Filing Date:
June 20, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CASSIOPEA S P A (IT)
International Classes:
A61K9/06; A61K9/00; A61K9/08; A61K31/573; A61K47/10; A61P17/14
Domestic Patent References:
WO2009019138A22009-02-12
WO2003014141A12003-02-20
WO2009019138A22009-02-12
Foreign References:
US8143240B22012-03-27
US8865690B22014-10-21
US8785427B22014-07-22
Other References:
CELASCO GIUSEPPE ET AL: "Biological profile of cortexolone 17alpha-propionate (CB-03-01), a new topical and peripherally selective androgen antagonist", ARZNEIMITTEL FORSCHUNG. DRUG RESEARCH, ECV EDITIO CANTOR VERLAG, AULENDORF, DE, vol. 54, no. 12, 1 January 2004 (2004-01-01), pages 881 - 886, XP008108154, ISSN: 0004-4172
V. TRIFU ET AL: "Cortexolone 17[alpha]-propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. A pilot randomized, double-blind comparative study vs. placebo and tretinoin 0.05% cream", BRITISH JOURNAL OF DERMATOLOGY, vol. 165, no. 1, 2 June 2011 (2011-06-02), pages 177 - 183, XP055135191, ISSN: 0007-0963, DOI: 10.1111/j.1365-2133.2011.10332.x
CELASCO ET AL., ARZNEIM.-FORSCH./DRUG RES, vol. 54, no. 12, 2004, pages 881 - 886
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method of treating alopecia in a subject in need thereof, comprising topically administering to the subject a pharmaceutical formulation comprising at least 2.1 weight percent cortexolone-17a-propionate and one or more pharmaceutically acceptable solvents, wherein the cortexolone-17a-propionate is fully solubilized in the formulation.

2. The method of claim 1, wherein the pharmaceutical formulation comprises from at least

2.1 weight percent to about 20 weight percent cortexolone-17a-propionate.

3. The method of claim 1 or claim 2, wherein the pharmaceutical composition comprises from about 2.5 weight percent to 15 weight percent cortexolone-17a-propionate.

4. The method of claim 3, wherein the pharmaceutical composition comprises about 15 weight percent cortexolone-17a-propionate.

5. The method of claim 3, wherein the pharmaceutical formulation comprises about 10 weight percent cortexolone-17a-propionate.

6. The method of claim 3, wherein the pharmaceutical formulation comprises about 8 weight percent cortexolone-17a-propionate.

7. The method of claim 3, wherein the pharmaceutical formulation comprises about 7.5 weight percent cortexolone-17a-propionate.

8. The method of claim 1, wherein the pharmaceutical formulation comprises from at least

2.1 weight percent to about 5.5 weight percent cortexolone-17a-propionate.

9. The method of claim 8, wherein the pharmaceutical formulation comprises about 3 weight percent cortexolone-17a-propionate.

10. The method of claim 8, wherein the pharmaceutical formulation comprises about 4 weight percent cortexolone-17a-propionate.

11. The method of claim 8, wherein the pharmaceutical formulation comprises about 5 weight percent cortexolone-17a-propionate.

12. The method of claim 8, wherein the pharmaceutical formulation comprises about 5.5 weight percent cortexolone-17a-propionate.

13. The method of claim 8, wherein the pharmaceutical formulation comprises 5 weight percent cortexolone-17a-propionate.

14. The method of any one of claims 1-13, wherein the pharmaceutical formulation is a liquid or semi-solid formulation.

15. The method of any one of claims 1-14, wherein the pharmaceutical formulation is a solution, a suspension, an emulsion, a microemulsion, a cream, a gel, a foam, or an ointment.

16. The method of any one of claims 1-15, wherein the pharmaceutical formulation is anhydrous and contains less than about 5 weight percent water.

17. The method of any one of claims 1-16, wherein the pharmaceutical formulation is a solution.

18. The method of any one of claims 1-17, wherein the formulation provides a mean Cmax of cortexolone-17a-propionate of about 0.5 to about 3 ng/ml after topical application of a single dose.

19. The method of claim 18, where in the mean Cmax of cortexolone-17a-propionate after administration of a single topical dose is about 0.5 to about 1.5 ng/ml

20. The method of claim 19, wherein the mean Cmax of cortexolone-17a-propionate after administration of a single topical dose is 1.04 ± 0.41 ng/ml.

21. The method of any one of claims 1-17, wherein the formulation provides a mean Cmax of cortexolone-17a-propionate of less than about 3 ng/ml after topical application of a single dose comprising about 50 mg of cortexolone-17a-propionate.

22. The method of any one of claims 1-17, wherein the formulation provides a mean Tmax of cortexolone-17a-propionate of less than about 20 hours after administration of a single topical dose.

23. The method of claim 22, wherein the formulation provides a mean Tmax of cortexolone- 17a-propionate of less than about 15 hours after administration of a single topical dose.

24. The method of claim 23, wherein the formulation provides a mean Tmax of cortexolone- 17a-propionate of less than about 12 hours after administration of a single topical dose.

25. The method of any one of claims 1-17, wherein the formulation provides a mean Tmax of cortexolone-17a-propionate of about 6.22 ± 5.17 hours after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

26. The method of any one of claims 1-17, wherein the formulation provides a mean AUC0-t of less than about 25 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

The method of claim 26, wherein the formulation provides a mean AUC0-t of less than about 20 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

28. The method of any one of claims 1-17, wherein the formulation provides a mean AUC0-t of about 15.69 ± 4.3 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

The method of any one of claims 1-17, wherein the formulation provides a mean steady state Cmax of cortexolone-17a-propionate of about 3.82 ± 1.34 ng/ml after topica administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

The method of any one of claims 1-17, wherein the formulation provides a mean steady state Tmax of cortexolone-17a-propionate of about 4.38 ± 1.96 hours after topica administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

The method of any one of claims 1-17, wherein the formulation provides a mean steady- state AUCo-t of about 37.37 ± 12.36 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

32. The method of any one of claims 1-17, wherein the alopecia is androgenetic alopecia, alopecia areata telogen effluvium, anagen effluvium, traction alopecia, or a combination of any of the foregoing. The method of claim 32, wherein the alopecia areata is selected from the group consisting of diffuse alopecia areata, alopecia areata monolocularis, alopecia areata multilocularis, ophiasis, alopecia totalis, and alopecia universalis.

The method of any one of claims 1-32, wherein the alopecia is androgenetic alopecia

35. The method of any one of claims 1-34, wherein the formulation further comprises at least one antioxidant, at least an emulsifier, or a combination of the foregoing.

36. The method of any one of claims 1-35, wherein the formulation is administered topically once or twice daily.

37. The method of any one of claims 1-14 or 18-36, wherein the formulation is a liquid.

38. The method of any one of claims 1-37, wherein from about 0.2 to about 2.0 ml of the formulation are administered during each application.

A topical pharmaceutical formulation comprising cortexolone-17a-propionate at a concentration of at least 2.1 weight percent, and one or more pharmaceutically acceptable solvents, wherein the cortexolone-17a-propionate is fully solubilized in the formulation.

40. The topical pharmaceutical formulation of claim 39, wherein the formulation comprises cortexolone-17a-propionate at a concentration from at least 2.1 weight percent to about 20 weight percent.

The topical pharmaceutical formulation of claim 39 or claim 40, wherein the pharmaceutical formulation comprises from at least 2.1 weight percent to about 17 weight percent cortexolone-17a-propionate.

42. The topical pharmaceutical formulation of any one of claims 39-41, wherein the pharmaceutical composition comprises from about 2.5 weight percent to 15 weight percent cortexolone-17a-propionate.

43. The topical pharmaceutical formulation of claim 42, wherein the pharmaceutical composition comprises about 15 weight percent cortexolone-17a-propionate.

44. The topical pharmaceutical formulation of claim 42, wherein the pharmaceutical formulation comprises about 10 weight percent cortexolone-17a-propionate.

45. The topical pharmaceutical formulation of claim 42, wherein the pharmaceutical formulation comprises about 8 weight percent cortexolone-17a-propionate.

46. The topical pharmaceutical formulation of claim 42, wherein the pharmaceutical formulation comprises about 7.5 weight percent cortexolone-17a-propionate.

47. The topical pharmaceutical formulation of claim 39, wherein the formulation comprises cortexolone-17a-propionate at a concentration from at least 2.1 weight percent to about 5.5 weight percent.

48. The topical pharmaceutical formulation of claim 47, wherein the pharmaceutical formulation comprises about 3 weight percent cortexolone-17a-propionate.

49. The topical pharmaceutical formulation of claim 47, wherein the pharmaceutical formulation comprises about 4 weight percent cortexolone-17a-propionate.

50. The topical pharmaceutical formulation of claim 47, wherein the pharmaceutical formulation comprises about 5 weight percent cortexolone-17a-propionate.

51. The topical pharmaceutical formulation of claim 47, wherein the pharmaceutical formulation comprises about 5.5 weight percent cortexolone-17a-propionate.

52. The topical pharmaceutical formulation of claim 47, wherein the pharmaceutical formulation comprises 5 weight percent cortexolone-17a-propionate.

53. The topical pharmaceutical formulation of any one of claims 39-52, wherein the pharmaceutical formulation is a liquid or semi-solid formulation.

54. The topical pharmaceutical formulation of claim 53, wherein the liquid or semi-solid, formulation is a solution, a suspension, an emulsion, a microemulsion, a cream, a gel, a foam, or an ointment.

55. The topical pharmaceutical formulation of any one of claims 39-54, wherein the pharmaceutical formulation is anhydrous and comprises less than about 5 percent water by weight.

56. The topical pharmaceutical formulation of claim 55, wherein the pharmaceutical formulation is anhydrous and comprises less than about 3 percent water by weight.

57. The topical pharmaceutical formulation of claim 55, wherein the pharmaceutical formulation is a solution.

58. The topical pharmaceutical formulation of any one of claims 39-57, wherein the formulation provides a mean steady-state Cmax of less than about 7.0 ng/ml of cortexolone- 17a-propionate upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate.

59. The topical pharmaceutical formulation of any one of claims 39-57, wherein the formulation provides a mean steady-state Tmax of cortexolone-17a-propionate of less than about 8.0 hours, upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate.

60. The topical pharmaceutical formulation of any one of claims 39-57, wherein the formulation provides an AUCX of cortexolone-17a-propionate of less than about 64.1 (ng*h)/ml, upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate

61. The topical pharmaceutical formulation of any one of claims 39-60, wherein the formulation is for use in the treatment of alopecia.

62. The topical pharmaceutical formulation of any one of claims 39-61, wherein the one or more pharmaceutically acceptable solvents are selected from the group consisting of a polyol, a polyol ether, and a C1-C7 alcohol.

63. The topical pharmaceutical formulation of claim 62, wherein the C C7 alcohol is ethanol, isopropanol, or methanol.

64. The topical pharmaceutical formulation of claim 63, wherein the C C7 alcohol is ethanol.

65. The topical pharmaceutical formulation of claim 64, wherein the ethanol is 96°.

66. The topical pharmaceutical formulation of claim 65, wherein the ethanol is absolute ethanol.

67. The topical pharmaceutical formulation of any one of claims 62-67, wherein the polyol is selected from the group consisting of ethylene glycol, propylene glycol, glycerol, and hexanetriol.

68. The topical pharmaceutical formulation of claim 67, wherein the polyol is propylene glycol.

69. The topical pharmaceutical formulation of any one of claims 62-68, wherein the polyol ether is selected from the group consisting of polypropylene glycol, polyethylene glycol, polyethylene-polypropylene triblock copolymers, dipropylene glycol, and diethylene glycol monoethyl ether.

70. The topical pharmaceutical formulation of claim 69, wherein the polyol ether is diethylene glycol monoethyl ether.

71. The topical pharmaceutical formulation of claim 62, wherein the polyol is propylene glycol, the polyol ether is diethylene glycol monoethyl ether, and the C1-C7 alcohol is ethanol.

72. The topical pharmaceutical formulation of claim 71, wherein the formulation is anhydrous and contains less than 5% water by weight or less than 3% water by weight.

73. The topical pharmaceutical formulation of any one of claims 39-72, wherein the formulation further comprises an emulsifier.

74. The topical pharmaceutical formulation of any one of claims 39-73, wherein the formulation further comprises an antioxidant.

75. The topical pharmaceutical formulation of claim 74, wherein the antioxidant is selected from the group consisting of butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), ascorbyl palmitate, ascorbic acid, alpha tocopherol, and propyl gallate.

76. The topical pharmaceutical formulation of claim 75, wherein the antioxidant ascorbyl palmitate.

77. The topical pharmaceutical formulation of claim 73, wherein the emulsifier is selected from the group consisting of PEG-15 hydroxystearate (also known as polyoxyl-15- hydroxystearate), PEG-30 stearate, PEG-40 laurate, PEG-40 oleate, polysorbate 20, polysorbate 60, polysorbate 80, PEG-20 cetostearyl ether, polyoxyl 25 cetostearyl, cetomacrogol 1000, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate propylene glycol esters of fatty acids, polyglycerol esters of fatty acids, polyoxyl 5 castor oil, polyoxyl 15 castor oil, polyoxyl 35 castor oil, polyoxyl 40 hydrogenated castor oil, caprylocapryl polyoxyil-8 glycerides; caprylocaproyl polyoxylglycerides, lauroyl polyoxylglycerides, oleoyl polyoxylglycerides, and combinations of any of the foregoing.

78. The topical pharmaceutical formulation of claim 77, wherein the emulsifier is polysorbate 80.

79. A method of treating alopecia, the method comprising topically administering to a subject in need thereof, the topical pharmaceutical formulation as claimed in any one of claims 39- 78, wherein twice a day topical administration of the formulation for at least six months achieves hair growth comparable to hair growth observed upon administration of oral finasteride for the same period of time.

80. The method of claim 79, wherein the method provides a reduced incidence of adverse effects selected from the group consisting of decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate.

81. A method of treating alopecia, the method comprising topically administering to a subject in need thereof the topical pharmaceutical formulation of any one of claims 39-78, wherein the topical administration of the formulation for at least six months achieves a mean value of the change from baseline in non-vellus Target Area Hair Count (TAHC) of about 12.7 in comparison with vehicle.

82. A method of treating alopecia, the method comprising topically administering to a subject in need thereof the topical pharmaceutical formulation of any one of claims 39-78, wherein the topical administration of the formulation for at least six months achieves a weighted average Hair Growth Assessment (HGA) score of about 0.30 in comparison with vehicle.

83. A method of treating alopecia, the method comprising topically administering to a subject in need thereof the topical pharmaceutical formulation of any one of claims 39-78, wherein the topical administration of the formulation for at least six months achieves a weighted average Investigator's Global Assessment (IGA) score of about 0.43 in comparison with vehicle.

84. A method of treating alopecia, the method comprising topically administering to a subject in need thereof the topical pharmaceutical formulation of any one of claims 39-78, wherein the topical administration of the formulation for at least six months is free from systemic antiandrogenic side-effects. A method of treating alopecia, the method comprising topically administering to a subject in need thereof the topical pharmaceutical formulation of any one of claims 39-78, wherein the topical administration of the formulation provides a) a mean change from baseline in Target Area Hair Count equal to or higher than 9 hairs/cm2 after 6 months of daily or BID application; or b) a mean change from baseline in Target Area Hair Count equal to or higher than 10 hairs/cm2 after 6 months of daily or BID application; or c) a mean change from baseline in Target Area Hair Count equal to or higher than 11 hairs/cm2 after 6 months of daily or BID application; or d) a mean change from baseline in Target Area Hair Count equal to or higher than 12 hairs/cm2 after 6 months of daily or BID application; or e) a weighted average HGA score equal to or higher than 0.20 after 6 months of daily or BID application; or

f) a weighted average HGA score equal to or higher than 0.30 after 6 months of daily or BID application; or

g) a weighted average HGA score equal to or higher than 0.40 after 6 months of daily or BID application; or

h) a weighted average IGA score equal to or higher than 0.10 after 6 months of daily or BID application; or

i) a weighted average IGA score equal to or higher than 0.20 after 6 months of daily or BID application; or

j) a weighted average IGA score equal to or higher than 0.30 after 6 months of daily or BID application; or

k) a favorable (positive) HGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

I) a favorable (positive) HGA score in at least about 20 % of subjects after 6 months of daily or BID application; or m) a favorable (positive) HGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

n) a favorable (positive) IGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

o) a favorable (positive) IGA score in at least about 20 % of subjects after 6 months of daily or BID application; or

P) a favorable (positive) IGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

q) a favorable (positive) IGA score in at least about 40 % of subjects after 6 months of daily or BID application.

86. The topical pharmaceutical formulation according to any one of claims 33-78, wherein the formulation is for use in the treatment of alopecia.

87. The topical pharmaceutical formulation for use according to claim 86, wherein the alopecia is androgenetic alopecia, alopecia areata, telogen effluvium, anagen effluvium, traction alopecia, or a combination of any of the foregoing.

88. The topical pharmaceutical formulation for use according to claim 87, wherein the alopecia is androgenetic alopecia.

89. The topical pharmaceutical formulation for use according to claim 87, wherein the alopecia areata is selected from the group consisting of diffuse alopecia areata, alopecia areata monolocularis, alopecia areata multilocularis, ophiasis, alopecia totalis, and alopecia universalis.

90. The topical pharmaceutical formulation for use according to any one of claims 86-89,

wherein the formulation is administered once or twice daily.

91. The topical pharmaceutical formulation for use according to any one of claims 86-90, wherein twice a day topical administration of the formulation for at least six months achieves hair growth comparable to hair growth observed upon administration of oral finasteride for the same period of time.

92. The topical pharmaceutical formulation for use according to claim 91, wherein the topical administration provides a reduced incidence of adverse effects selected from the group consisting of decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate.

93. The topical pharmaceutical formulation for use according to any one of claims 86-

90, wherein the topical administration of the formulation for at least six months achieves a mean value of the change from baseline in non-vellus Target Area Hair Count (TAHC) of about 12.7 in comparison with vehicle.

94. The topical pharmaceutical formulation for use according to any one of claims 86-90,

wherein the topical administration of the formulation for at least six months achieves a weighted average Hair Growth Assessment (HGA) score of about 0.30 in comparison with vehicle.

95. The topical pharmaceutical formulation of any one of claims 86-90, wherein the topical administration of the formulation for at least six months achieves a weighted average Investigator's Global Assessment (IGA) score of about 0.43 in comparison with vehicle.

96. The topical pharmaceutical formulation for use according to any one of claims 86-90, wherein the topical administration of the formulation for at least six months is free from systemic antiandrogenic side-effects. The topical pharmaceutical formulation for use according to any one of claims 86-90, wherein the topical administration of the formulation provides

a) a mean change from baseline in Target Area Hair Count equal to or higher than 9 hairs/cm2 after 6 months of daily or BID application; or b) a mean change from baseline in Target Area Hair Count equal to or higher than 10 hairs/cm2 after 6 months of daily or BID application; or c) a mean change from baseline in Target Area Hair Count equal to or higher than 11 hairs/cm2 after 6 months of daily or BID application; or d) a mean change from baseline in Target Area Hair Count equal to or higher than 12 hairs/cm2 after 6 months of daily or BID application; or e) a weighted average HGA score equal to or higher than 0.20 after 6 months of daily or BID application; or

f) a weighted average HGA score equal to or higher than 0.30 after 6 months of daily or BID application; or

g) a weighted average HGA score equal to or higher than 0.40 after 6 months of daily or BID application; or

h) a weighted average IGA score equal to or higher than 0.10 after 6 months of daily or BID application; or

i) a weighted average IGA score equal to or higher than 0.20 after 6 months of daily or BID application; or

j) a weighted average IGA score equal to or higher than 0.30 after 6 months of daily or BID application; or

k) a favorable (positive) HGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

I) a favorable (positive) HGA score in at least about 20 % of subjects after 6 months of daily or BID application; or a favorable (positive) HGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

a favorable (positive) IGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

a favorable (positive) IGA score in at least about 20 % of subjects after 6 months of daily or BID application; or

a favorable (positive) IGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

a favorable (positive) IGA score in at least about 40 % of subjects after 6 months of daily or BID application.

Description:
HIGH CONCENTRATION FORMU LATION

BACKGROU ND

[0001] Alopecia is a group of disorders with multiple and varying etiologies that results in hair loss from the body. The most common form of alopecia is androgenetic alopecia ("AGA"). AGA is also commonly called alopecia androgenetica, male pattern baldness, or male-pattern or female-pattern hair loss. It has been reported that AGA affects roughly 50% of men over 40. This form of alopecia may eventually affect up to 80% of white men by the age of 70 and about half of all women. Hair loss is often a cause of great concern to a patient for cosmetic and psychological reasons, but it can also be an important sign of systemic disease.

[0002] AGA is a hereditary hair follicle disease that is also androgen-dependent.

Dihydrotestosterone, in particular, plays a major role in the development and progression of AGA. Hair loss with AGA is progressive such that patients with disease experience a reduction in the normal 4:1 terminal-to-vellus hair ratio over a period of time. During this process, terminal hairs are converted to indeterminate hairs and finally into vellus hairs. Men present with hair thinning in the temporal areas, which advances to the crown (vertex) area as the AGA progresses. Women usually have more diffuse thinning on the crown area, and less commonly present with a male-type pattern.

[0003] In vivo, AGA is characterized by increased levels and activity of 5a-reductase isoenzymes. These enzymes convert testosterone (T) into its active metabolite dihydrotestosterone (DHT). High concentrations of DHT at the hair-follicle level, due to increased binding capacity of the hormone to hair-follicle androgen receptors, shorten the hair cycle and gradually miniaturize scalp follicles. Following miniaturization of the follicles, fibrous tracts remain. These DHT-dependent effects are considered, in most cases, reversible, such that AGA may be susceptible to medical treatment with drugs able to reduce DHT production, or to antagonize DHT/T interaction with hair-follicle androgen receptors. [0004] Current medical management of AGA comprises surgical and pharmacological treatment options. The most common form of surgical intervention for AGA is hair transplantation. This procedure has been performed successfully for the past four decades. Hair transplantation involves harvesting intact hair follicles from within a safe donor area (SDA) of a patient's scalp by either follicular unit strip surgery (FUSS) or follicular unit extraction (FUE). Refinements to these procedures over the last decade have led to markedly improved hair survival and more natural appearing results.

[0005] Although cosmetic results after surgery are often satisfactory, surgery can only be performed when a sufficient quantity of donor plugs (or follicles) are available to cover the balding area(s). Additionally, hair transplantation is generally reserved for patients who have experienced massive hair loss and for patients in whom AGA is not still evolving. In younger patients, for example, hair transplantation is generally not recommended because androgens, particularly DHT, can act on the newly transplanted follicles resulting in same thinning and eventual hair loss that affected the originally present follicles. For this reason, at the initial onset and in the initial phases of the AGA, pharmacological intervention is preferred.

[0006] Currently, only two medicinal products are approved for treatment of AGA: minoxidil (sold commercially as ROGAINE ® and its generic forms) and finasteride (sold commercially as PROPECIA ® ). Minoxidil, which is formulated as a topical drug product, is currently available at two strength levels - 2% (topical solution) and 5% (as a topical solution or foam). To exert its effect minoxidil needs to be transformed into its active metabolite, minoxidil sulfate, by the enzyme sulfotransferase. This enzyme is present in the outer root sheath of anagen follicles.

[0007] Although the exact mechanism by which minoxidil promotes hair growth is still unclear, it is believed that minoxidil sulfate opens ATP-sensitive potassium channels in cell membranes resulting in vasodilation. Vasodilation, however, does not appear to be responsible for minoxidil-induced hair growth. Other possible effects of minoxidil on hair follicles include: a) increased expression of vascular endothelial growth factor (VEGF) mRNA in the dermal papilla, which indicates that the drug induces angiogenesis in the dermal papilla; b) activation of cytoprotective prostaglandin synthase-1, a cytoprotective enzyme that stimulates hair growth; and c) increased expression of hepatocyte growth factor (HGF), which is an hair growth promoter.

[0008] In various clinical trials, minoxidil was found to be effective in significantly improving total hair count and non-vellus hair count after 6 to 12 months of treatment, in comparison to placebo. Moreover, minoxidil preparations are well tolerated, with only mild side effects. Despite these beneficial effects, minoxidil does not reduce dihydrotestosterone (DHT) or the enzyme responsible for its accumulation around the hair follicle, 5-alpha reductase - the primary mediator of male pattern baldness in genetically susceptible individuals. As a result, when treatment is stopped, DHT shrinks and ultimately destroys genetically predisposed hair follicles.

[0009] Finasteride, on the other hand, inhibits 5-alpha reductase type II, which is responsible for transforming testosterone into DHT at the follicle. Finasteride is administered orally in a lmg tablet formulation. A single oral administration of finasteride 1 mg decreases serum DHT as well as scalp DHT up to 70 % compared to baseline. In several published clinical trials, finasteride 1 mg was found to be effective in significantly improving the total hair count in comparison to placebo after 6 months of treatment. The significant increase in total hair counts, in comparison to placebo, in patients under treatment with finasteride 1 mg were maintained in long term treatments (up to 60 months).

[0010] Even though finasteride effectively stops hair loss and improves new hair growth, there are possible adverse effects associated with its use. Foremost, finasteride is contraindicated in women due to suspected teratogenic effects. Thus, women who are or may become pregnant, regardless of whether they suffer from AGA, are strongly cautioned to avoid contact with broken or crushed tablets. Additionally, because finasteride is distributed systemically, it not only reduces DHT levels at the hair follicle, but in the plasma as well. This systemic activity is responsible for finasteride's main side effects, which include decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate. Other less common side effects include breast swelling, palpitations, pain in testicles, persistent decrease in sex drive after discontinuation, infertility, and depression.

[0011] In view of the deficiencies and drawbacks associated with finasteride and minoxidil therapies, it is apparent that there is a need in the art for new methods and active agents for treating alopecia, and in particular, AGA.

BRI EF SU MMARY

[0012] Cortexolone-17a-propionate (17a-propionyloxy-21-hydroxy-pregna-4-ene-3,20- dione) is a known topical antiandrogen that displaces androgenic hormones from binding with their receptors. It is known to be suitable for treating acne, alopecia, and other diseases of skin and cutaneous appendages. See, e.g., U.S. Patent Nos. 8,143,240 and 8,865,690. The compound is also known to exist in several distinct crystalline polymorphs, each having unique properties. See, e.g., U.S. Patent No. 8,785,427. Each of these patents is incorporated herein by reference in its entirety.

[0013] Although certain formulations of cortexolone-17a-propionate have been disclosed in the art, these formulations were limited to a concentration of less than 2% by weight due to inherent solubility limitations of the active and concerns that the administration of higher concentrations could lead to greater and unwanted systemic exposure to the active and/or its metabolites - particularly after repeated administration.

[0014] Despite these concerns, it has now been unexpectedly discovered that it is possible to prepare pharmaceutical formulations comprising cortexolone-17a-propionate as the therapeutic agent for topical administration at concentrations of greater than 2% by weight. These pharmaceutical formulations have favorable characteristics for the treatment of alopecia and provide optimal topical delivery of the therapeutic agent as evidenced by the pharmacokinetic profiles described herein. I n particular, these pharmacokinetic profiles show that the formulations described herein advantageously maximize topical delivery of cortexolone-17a-propionate in the skin and/or in the scalp while simultaneously minimizing systemic exposure to cortexolone-17a-propionate or any of its metabolites. As a result of such drug disposition, an advantageous clinical result can be realized after daily topical administration for an appropriate amount of time, such as a few days, weeks, or months. Moreover, it has also been discovered that the pharmaceutical formulations described herein possess favorable stability profiles, allowing for finished product storage at room temperature for at least two years.

[0015] In certain embodiments the present disclosure provides a method of treating alopecia in a patient in need thereof, comprising topically administering to the patient a pharmaceutical formulation comprising at least 2.1 weight percent cortexolone-17a- propionate and one or more pharmaceutically acceptable solvents.

[0016] In certain embodiments, the pharmaceutical formulation comprises from about 2.1 weight percent to about 20 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises from about 2.1 weight percent to about 17 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises from about 2.5 weight percent to about 17 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical composition comprises from about 2.5 weight percent to 15 weight percent cortexolone-17a-propionate. In other embodiment, the pharmaceutical composition comprises from about 3 weight percent to about 15 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 6 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 7 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 7.5 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 8 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 9 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 10 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 11 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 12 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 13 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 14 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 15 weight percent cortexolone-17a-propionate.

[0017] In certain embodiments, the pharmaceutical formulation comprises from about 2.1 weight percent to about 5.5 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 2.5 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 3 weight percent cortexolone-17a-propionate. In other embodiments, the pharmaceutical formulation comprises about 4 weight percent cortexolone-17a-propionate. In still other embodiments, the pharmaceutical formulation comprises about 5 weight percent cortexolone-17a-propionate. In still further embodiments, the pharmaceutical formulation comprises about 5.5 weight percent cortexolone-17a-propionate. In yet another embodiment, the pharmaceutical formulation comprises 5 weight percent cortexolone-17a-propionate.

[0018] In certain embodiments, the pharmaceutical formulation is a liquid or semi-solid formulation. In a further embodiment, the pharmaceutical formulation is a solution, a suspension, an emulsion, a microemulsion, a cream, a gel, a foam, or an ointment.

[0019] In certain embodiments, the pharmaceutical formulation is anhydrous and contains less than about 5 weight percent water. In further embodiments, the pharmaceutical formulation is a solution.

[0020] In certain embodiments, the formulation provides a mean C max of cortexolone-17a- propionate of about 0.5 to about 3 ng/ml after topical application of a single dose. In certain embodiments, the mean C max of cortexolone-17a-propionate after administration of a single topical dose is about 0.5 to about 1.5 ng/ml. In other embodiments, the mean C max of cortexolone-17a-propionate after administration of a single topical dose is 1.04 ± 0.41 ng/ml. [0021] In still other embodiments, the formulation provides a mean C max of cortexolone-

17a-propionate of less than about 3 ng/ml after topical application of a single dose comprising about 50 mg of cortexolone-17a-propionate.

[0022] In some embodiments, the formulation provides a mean T max of cortexolone-17a- propionate of less than about 20 hours after administration of a single topical dose. In other embodiments, the formulation provides a mean T max of cortexolone-17a-propionate of less than about 15 hours after administration of a single topical dose. In yet another embodiment, the formulation provides a mean T max of cortexolone-17a-propionate of less than about 12 hours after administration of a single topical dose. And in yet another embodiment, the formulation provides a mean T max of cortexolone-17a-propionate of about 6.22 ± 5.17 hours after topical administration of a single dose comprising about 50 mg of cortexolone-17a- propionate.

[0023] In certain embodiments, the formulation provides a mean AUC 0 - t of less than about

25 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate. In other embodiments, the formulation provides a mean AUC 0 - t of less than about 20 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate. And in still other embodiments, the formulation provides a mean AUC 0 - t of about 15.69 ± 4.3 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

[0024] In some embodiments, the formulation provides a mean steady-state C max of cortexolone-17a-propionate of about 3.82 ± 1.34 ng/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

[0025] In other embodiments, the formulation provides a mean steady-state T max of cortexolone-17a-propionate of about 4.38 ± 1.96 hours after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate.

[0026] In some embodiments, the formulation provides a mean steady-state AUC 0 - t of about 37.37 ± 12.36 (ng*h)/ml after topical administration of a single dose comprising about 50 mg of cortexolone-17a-propionate. [0027] In some embodiments, the formulations of the invention provide a mean change from baseline in Target Area Hair Count equal to or higher than 8 hairs/cm 2 after 6 months of daily or BID application.

[0028] In some embodiments, the alopecia is androgenetic alopecia, alopecia areata, telogen effluvium, anagen effluvium, traction alopecia, or a combination of any of the foregoing. In certain embodiments, the alopecia areata is selected from the group consisting of diffuse alopecia areata, alopecia areata monolocularis, alopecia areata multilocularis, ophiasis, alopecia totalis, and alopecia universalis.

[0029] In some embodiments, the alopecia is androgenetic alopecia.

[0030] In some embodiments, the formulation further comprises at least one antioxidant, at least an emulsifier, or a combination of the foregoing.

[0031] In some embodiments, the formulation is administered once or twice daily.

[0032] In some embodiments, the formulation is a liquid.

[0033] In some embodiments, from about 0.2 to about 2.0 ml of the formulation are administered during each application.

[0034] In a further embodiment, the present disclosure provides a topical pharmaceutical formulation comprising cortexolone-17a-propionate at a concentration of at least 2.1 weight percent, and one or more pharmaceutically acceptable solvents.

[0035] In certain embodiments, the formulation comprises from about 2.1 weight percent to about 20 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises from about 2.1 weight percent to about 17 weight percent cortexolone- 17a-propionate. In other embodiments, the formulation comprises from about 2.5 weight percent to about 17 weight percent cortexolone-17a-propionate. In other embodiments, the composition comprises from about 2.5 weight percent to 15 weight percent cortexolone-17a- propionate. In other embodiment, the composition comprises from about 3 weight percent to about 15 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 6 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 7 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 7.5 weight percent cortexolone-17a- propionate. In other embodiments, the formulation comprises about 8 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 9 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 10 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 11 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 12 weight percent cortexolone-17a- propionate. In other embodiments, the formulation comprises about 13 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 14 weight percent cortexolone-17a-propionate. In other embodiments, the formulation comprises about 15 weight percent cortexolone-17a-propionate.

[0036] In certain embodiments, the formulation comprises cortexolone-17a-propionate at a concentration from about 2.1 weight percent to about 5.5 weight percent. In other embodiments, the formulation comprises about 2.5 weight percent cortexolone-17a- propionate. In other embodiments, the topical pharmaceutical formulation comprises about 3 weight percent cortexolone-17a-propionate. In another embodiment, the pharmaceutical formulation comprises about 4 weight percent cortexolone-17a-propionate. In still another embodiment, the pharmaceutical formulation comprises about 5 weight percent cortexolone- 17a-propionate. In yet a further embodiment, the pharmaceutical formulation comprises about 5.5 weight percent cortexolone-17a-propionate. And in yet another embodiment, the pharmaceutical formulation comprises 5 weight percent cortexolone-17a-propionate.

[0037] In certain embodiments, the pharmaceutical formulation is a liquid or semi-solid formulation. In some embodiments, the liquid or semi-solid, formulation is a solution, a suspension, an emulsion, a microemulsion, a cream, a gel, a foam, or an ointment.

[0038] In some embodiments, the pharmaceutical formulation is anhydrous and comprises less than about 5 percent water by weight. In other embodiments, the pharmaceutical formulation is anhydrous and comprises less than about 3 percent water by weight.

[0039] In some embodiments, the topical pharmaceutical formulation is a solution. [0040] In some embodiments, the formulation provides a mean steady-state C max of less than about 7.0 ng/ml of cortexolone-17a-propionate upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate.

[0041] In other embodiments, the formulation provides a mean steady-state T max of cortexolone-17a-propionate of less than about 8.0 hours, upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate.

[0042] In certain embodiments, the formulation provides an AUC X of cortexolone-17a- propionate of less than about 64.1 (ng*h)/ml, upon the application of an amount of the formulation including about 50 mg of cortexolone-17a-propionate

[0043] In certain embodiments, the formulation is for use in the treatment of alopecia.

[0044] In certain embodiments, the one or more pharmaceutically acceptable solvents are selected from the group consisting of a polyol, a polyol ether, and a C1-C7 alcohol. In some embodiments, the C C 7 alcohol is ethanol, isopropanol, or methanol. In other embodiments, the C C 7 alcohol is ethanol. In certain embodiments, the ethanol is 96°. In some embodiments, the ethanol is absolute ethanol.

[0045] In some embodiments, the polyol is selected from the group consisting of ethylene glycol, propylene glycol, glycerol, and hexanetriol. In particular embodiments, the polyol is propylene glycol.

[0046] In certain embodiments, the polyol ether is selected from the group consisting of polypropylene glycol, polyethylene glycol, polyethylene-polypropylene triblock copolymers, dipropylene glycol, and diethylene glycol monoethyl ether. In particular embodiments, the polyol ether is diethylene glycol monoethyl ether.

[0047] In some embodiments, polyol is propylene glycol, the polyol ether is diethylene glycol monoethyl ether, and the C C 7 alcohol is ethanol.

[0048] In certain embodiments, formulation is anhydrous and contains less than 5% water by weight or less than 3% water by weight.

[0049] In other embodiments, the formulation further comprises an emulsifier. [0050] In certain embodiments, the formulation further comprises an antioxidant. In some embodiments, the antioxidant is selected from the group consisting of butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), ascorbyl palmitate, ascorbic acid, alpha tocopherol, and propyl gallate. In certain embodiments, the antioxidant ascorbyl palmitate.

[0051] In certain embodiments, the emulsifier is selected from the group consisting of

PEG-15 hydroxystearate (also known as polyoxyl-15-hydroxystearate), PEG-30 stearate, PEG-40 laurate, PEG-40 oleate, polysorbate 20, polysorbate 60, polysorbate 80, PEG-20 cetostearyl ether, polyoxyl 25 cetostearyl, cetomacrogol 1000, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate propylene glycol esters of fatty acids, polyglycerol esters of fatty acids, polyoxyl 5 castor oil, polyoxyl 15 castor oil, polyoxyl 35 castor oil, polyoxyl 40 hydrogenated castor oil, caprylocapryl polyoxyil-8 glycerides; caprylocaproyl polyoxylglycerides, lauroyi polyoxylglycerides, oleoyi polyoxylglycerides, and combinations of any of the foregoing. In certain embodiments, the emulsifier is polysorbate 80.

[0052] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulation as described herein, wherein twice a day topical administration of the formulation for at least six months achieves hair growth comparable to hair growth observed upon administration of oral finasteride for the same period of time. In one aspect, the method provides a reduced incidence of adverse effects selected from the group consisting of decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate.

[0053] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulation as described herein, wherein the topical administration of the formulation for at least six months achieves a mean value of the change from baseline in non- vellus Target Area Hair Count (TAHC) of about 12.7 in comparison with vehicle.

[0054] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulation as described herein, wherein the topical administration of the formulation for at least six months achieves a weighted average Hair Growth Assessment (HGA) score of about 0.30 in comparison with vehicle.

[0055] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulations as described herein, wherein the topical administration of the formulation for at least six months achieves a weighted average Investigator's Global Assessment (IGA) score of about 0.43 in comparison with vehicle.

[0056] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulation as described herein, wherein the topical administration of the formulation for at least six months is free from systemic antiandrogenic side-effects.

[0057] In another embodiment, the present description provides a method of treating alopecia, the method comprising topically administering to a subject in need thereof a topical pharmaceutical formulation as described herein, wherein the topical administration of the formulation provides

[0058] a mean change from baseline in Target Area Hair Count equal to or higher than 9 hairs/cm 2 after 6 months of daily or BID application; or

[0059] a mean change from baseline in Target Area Hair Count equal to or higher than 10 hairs/cm 2 after 6 months of daily or BID application; or

[0060] a mean change from baseline in Target Area Hair Count equal to or higher than 11 hairs/cm 2 after 6 months of daily or BID application; or

[0061] a mean change from baseline in Target Area Hair Count equal to or higher than 12 hairs/cm 2 after 6 months of daily or BID application; or

[0062] a weighted average HGA score equal to or higher than 0.20 after 6 months of daily or BID application; or

[0063] a weighted average HGA score equal to or higher than 0.30 after 6 months of daily or BID application; or [0064] a weighted average HGA score equal to or higher than 0.40 after 6 months of daily or BID application; or

[0065] a weighted average IGA score equal to or higher than 0.10 after 6 months of daily or

BID application; or

[0066] a weighted average IGA score equal to or higher than 0.20 after 6 months of daily or

BID application; or

[0067] a weighted average IGA score equal to or higher than 0.30 after 6 months of daily or

BID application; or

[0068] a favorable (positive) HGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

[0069] a favorable (positive) HGA score in at least about 20 % of subjects after 6 months of daily or BID application; or

[0070] a favorable (positive) HGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

[0071] a favorable (positive) IGA score in at least about 10 % of subjects after 6 months of daily or BID application; or

[0072] a favorable (positive) IGA score in at least about 20 % of subjects after 6 months of daily or BID application; or

[0073] a favorable (positive) IGA score in at least about 30 % of subjects after 6 months of daily or BID application; or

[0074] a favorable (positive) IGA score in at least about 40 % of subjects after 6 months of daily or BID application.

BRIEF DESCRI PTION OF THE DRAWINGS

[0075] The foregoing summary, as well as the following detailed description of the

embodiments, will be better understood when read in conjunction with the appended figures. For the purpose of illustration, the figures may describe the use of specific embodiments. It should be understood, however, that the compounds, formulations and compositions described herein are not limited to the precise embodiments discussed or described in the figures.

[0076] Figure 1 depicts the frequency distribution of Hair Growth Assessment (HGA),

performed by study subjects, in two treatment groups after 6 months of treatment in the Phase 2 clinical study in males with androgenic alopecia (AGA) described in Example 9.

[0077] Figure 2 depicts the frequency distribution of Investigator's Global Assessment

(IGA) for hair growth in two treatment groups after 6 months of treatment in the Phase 2 clinical study in males with AGA described in Example 9.

DETAI LED DESCRI PTION

[0078] The formulations described herein constitute a major improvement to the currently available therapies of alopecia, and specifically AGA, and would allow the physicians to have an effective, valuable and safe treatment of alopecia, and in particular AGA.

[0079] It has been found that the pharmaceutical formulations for topical administration described herein provide an optimal topical delivery of the therapeutic agent cortexolone-17a- propionate and, when used in the treatment of alopecia, and in particular AGA, are able to achieve similar results, in terms of efficacy, as commercially available finasteride 1 mg tablets (PROPECIA ® ) per os once daily, with minimal adverse effects. PROPECIA ® has been approved for the treatment of AGA. A comparison of the effect of a formulation described herein and that of PROPECIA ® is provided in Example 11 based on the information available on the US Food and Drug Administration website and provided in the PROPECIA ® package insert.

[0080] The pharmaceutical formulations described herein minimize systemic exposure and thus are safe. The results of the study described in Example 9 of the experimental section demonstrate that cortexolone-17a-propionate solution 5% was well tolerated locally. The incidence of local tolerability signs was low and the incidence of "treatment-emergent" local tolerability signs was generally similar among cortexolone-17a-propionate solution 5% and vehicle treatment groups. Most local tolerability signs were minimal/trace to mild in severity. Scaling and pruritus were most frequently observed "treatment-emergent" signs, with fewer subjects having erythema, folliculitis, and hyperpigmentation. In addition, the incidence of adverse events (AEs) was similar among the two treatment groups, with most events typically mild in severity and not related to the test article. Importantly, no AEs that could be correlated to systemic antiandrogenic activity of cortexolone-17a-propionate (e.g. decrease of libido, erectile dysfunction and ejaculation disorders) occurred during the 6-month treatment. The study of the Example 9 also confirms that the formulations described herein, when topically applied on the scalp, are optimal for achieving a local delivery of the therapeutic agent, meanwhile avoiding its excessive systemic distribution, which may cause the occurrence of antiandrogenic effects in treated subjects. The low levels of circulating cortexolone-17a- propionate, following the application of cortexolone-17a-propionate on the scalp, is also evident from the PK parameters exemplified in Example 6. At least this property differentiates the formulations described herein from finasteride 1 mg tablets (PROPECIA ® ): as reported in the PROPECIA ® package insert, the drug has many side effects, such as decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate. These adverse events of PROPECIA ® , which are reported in > 1% of the patients, are caused by the systemic antiandrogenic activity exerted by the therapeutic agent. Differently from PROPECIA ® , the formulations described herein are able to maximize the delivery of the therapeutic agent to the target site (i.e. the hair follicle), minimizing its systemic adsorption, which translates into a pharmacological profile characterized by efficacy in promoting hair regrowth (due to local activity on hair follicles), with absence of significant adverse events, including those due to systemic antiandrogenic effects.

1] It is evident, based on the studies described in the experimental section, that the formulations described herein represent a great improvement with respect to the currently available systemic antiandrogenic drug, PROPECIA ® , for the treatment of AGA. An exemplary formulation of the invention, disclosed in Example 4b, when tested in a phase II clinical trial in subjects affected by AGA (with topical applications twice daily on the scalp of the affected area for 6 months), demonstrated to be effective in stimulating the hair growth, as assessed by the two co-primary endpoints (the mean change from baseline in non-vellus TAHC and the subject self-assessment HGA questionnaire), which had a larger magnitude of improvement with respect to the vehicle. As discussed in Example 11, topical administration of this exemplary formulation of Example 4b provided a mean change in non-vellus TAHC at 6 months which was almost identical to that of PROPECIA ® at 6 months (12.7 for formulation of Example 4b vs. 12.2 calculated from the published results of two phase I II clinical studies for Propecia ® ), without systemic antiandrogenic effects that are known to be responsible of the main side effects of PROPECIA ® , such as decreased libido, erectile dysfunction (impotence) and ejaculation disorders.

[0082] The articles "a," "an," and "the" are used herein to refer to one or to more than one

(i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.

[0083] As used herein, the term "about" means ± 10% of the specified value, unless otherwise indicated.

[0084] As used herein, the phrases "active agent" and "therapeutic agent" are interchangeable and refer to cortexolone-17a-propionate.

[0085] The term "alopecia" as used herein refers to, collectively, or individually as specified, androgenetic alopecia (AGA), alopecia areata (including diffuse alopecia areata, alopecia areata monolocularis, alopecia areata multilocularis, ophiasis, alopecia totalis, and alopecia universalis), telogen effluvium, anagen effluvium, and traction alopecia.

[0086] As used herein, the term "anhydrous" means substantially free of water, i.e. having less than about 5 weight percent water, and, in certain embodiments as specified herein, less than about 3 weight percent water.

[0087] As used herein, the term "antioxidant" includes those pharmaceutically acceptable antioxidants known to those of ordinary skill in the art. Examples include, but are not limited to, butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), ascorbyl palmitate, ascorbic acid, alpha tocopherol (also known as Vitamin E), propyl gallate, and the like.

[0088] As used herein, the phrase "area under the curve" or "AUC" refers to the area under the curve defined by changes in the plasma concentration of the active agent (or one of its metabolites, as specified) over time following the application of a dose of the active agent itself or a formulation comprising the same. "AUC 0 -∞" is the area under the concentration- time curve extrapolated from t 0 to infinity following the application of a dose. "AUC 0 - t " is the area under the concentration-time curve from time zero to time t following the application of a dose, wherein t is the last time point with a measurable concentration. AUC X refers to mean steady-state AUC.

[0089] As used herein, the phrase "C1-C7 alcohol" refers to an alcohol having up to 7 carbons that is suitable for use in a topical pharmaceutical formulation. Examples of such C1-C7 alcohols include, but are not limited to, methanol, ethanol, isopropanol, n-butanol, n-propanol, benzyl alcohol, and the like. Without wishing to be bound by any particular theory, it is believed that C1-C7 alcohols, and particularly short chain C1-C7 alcohols, like ethyl, propyl or isopropyl alcohols, exert a solubilizing activity on cortexolone-17a-propionate. It is further believed that such C1-C7 alcohols contribute to the spreadability of the formulations described herein.

[0090] As used herein, the term "C max " refers to the maximum concentration of an active agent, or a metabolite thereof, on a graph of the plasma concentration of the active agent (or its metabolite) vs. time. C maxx refers to the maximum concentration of an active agent, or a degradation product thereof, on a graph of the plasma concentration of the active agent (or its degradation product or metabolite) vs. time when the steady state level has been reached.

[0091] As used herein "cortexolone" (also known as "11-Deoxycortisol" or "Reichstein's substance") refers to the compound havin the structure:

[0092] As used herein "cortexolone-17a-propionate" refers to the compound 17a- propionyloxy-21-hydroxy-pregna-4-ene-3,20-dione, which is equivalent to the chemical structure:

[0093] As used herein, the term "metabolite" refers to those compounds that result from the in vivo metabolism or degradation of cortexolone-17a-propionate. Exemplary known metabolites include, but are not limited to, cortexolone and tetrahydrocortexolone.

[0094] As used herein, the term "degradation product" refers to those compounds that result from the in vitro degradation of cortexolone-17a-propionate. Exemplary known cortexolone-17a-propionate degradation products include, but are not limited to, cortexolone- 21-propionate and cortexolone.

[0095] As used herein the term "ester" refers to esterified organic solvents including, but not limited to, ethyl acetate and ethyl lactate.

[0096] As used herein the phrase "natural oil" refers to those oils isolable from natural sources. Exemplary natural oils include, but are not limited to, almond oil, olive oil, cottonseed oil, safflower oil, and the like

[0097] As used herein, the term "polyol" refers to organic molecules containing two or more hydroxyl groups. Exemplary polyols include, but are not limited to, ethylene glycol, propylene glycol, glycerol, hexanetriol, and the like.

[0098] As used herein, the phrase "polyol ether" refers to a polyol ether suitable for use in a topical pharmaceutical formulation. Exemplary polyol ethers include, but are not limited to, polypropylene glycol, polyethylene glycol, polyethylene-polypropylene triblock copolymers, dipropylene glycol, diethylene glycol monoethyl ether, and the like.

[0099] As used herein, the phrase "penetration enhancer," refers to those pharmaceutically acceptable compounds that increase penetration of the active agent. Exemplary penetration enhancers include, but are not limited to, polyoxyethylene alkyl ethers, polyoxyl glycerides, dimethyl sulfoxide, pyrrolidone, N-methyl-2-pyrrolidone, diethylene glycol monoethyl ether (TRANSCUTOL ® ), dimethyl isosorbide, diethyl sebacate, azone, menthol, nerol, camphor, methyl salicylate, Tween 80, SDS, benzalkonium chloride, polyoxyl 40 hydrogenated castor oil (CREMOPHOR ® RH40, KOLLIPHOR ® RH40), didecyldimethylammonium bromide (DDAB), didecyltrimethylammonium bromide (DTAB), fatty acids esters such as isopropyl myristate, isopropyl palmitate and the like, fatty acids such as oleic acid, palmitic acid, linoleic acid, and salts thereof, fatty alcohols such as oleyl alcohol, myristyl alcohol, stearyl alcohol and the like, medium-chain triglycerides, and combinations of any of the foregoing.

[0100] As used herein, the term "solvent" means one or a mixture of more than one pharmaceutically acceptable solvents suitable for topical application, including without limitation, the scalp, that is used to solubilize cortexolone-17a-propionate in a formulation described herein.

[0101] As used herein "tetrahydrocortexolone" refers to the compound having the

Chemical Abstract Service (CAS) Registry Number 68-60-0.

[0102] As used herein, the term "T max " refers to the time at which maximum plasma concentration of an active agent (or metabolite thereof) is reached after application of the active agent. The term T max refers to the time when the maximum concentration of an active agent, or metabolite thereof, on a graph of the plasma concentration of the active agent (or its metabolite) vs. time when the steady state level has been reached

[0103] The terms "treat," "treating," and "treatment" refer to any indicia of success in the treatment or amelioration of an injury, disease, or condition, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, disease, or condition more tolerable to the patient; slowing in the rate of degeneration or decline; or improving a patient's physical or mental well-being. The treatment or amelioration of symptoms can be based on objective or subject parameters, including the results of a physical examination, neuropsychiatric examinations, or psychiatric evaluation.

[0104] The term "preventing" refers to keeping from happening, existing, or alternatively delaying the onset or recurrence of a disease, disorder, or condition to which such term applies, or of one or more symptoms associated with a disease, disorder, or condition. The term "preventing" also refers to reducing the incidence of a disease, disorder or condition. The term "prevention" refers to the act of preventing.

[0105] As used herein, the phrase "weight percent" is intended to encompass and disclose embodiments wherein the weight percent is weight percent by volume (w/v) and percentage of weight by total weight (w/w) for a given value. By way of example, an embodiment comprising 10 weight percent of element "X" discloses embodiments comprising both 10 weight percent "X" (w/v) and 10 weight percent "X" (w/w). Similarly, and again by way of example, an embodiment comprising 10 weight percent "X," 20 weight percent "Y," and 60 weight percent "Z," discloses embodiments comprising: a) 10 weight percent "X" (w/v), 20 weight percent "Y" (w/v), and 60 weight percent "Z" (w/v); and b) 10 weight percent "X" (w/w), 20 weight percent "Y" (w/w), and 60 weight percent "Z" (w/w). The foregoing notwithstanding, in certain embodiments, a value will be marked specifically "(w/w)" or "(w/v)." In those instances, the value should be interpreted as disclosing only the labeled value, i.e. only (w/w) or only (w/v) - but not both.

[0106] As used herein, the terms "comprises," "comprising," "having," "including,"

"containing," and the like are open-ended terms meaning "including, but not limited to." To the extent a given embodiment disclosed herein "comprises" certain elements, it should be understood that present disclosure also specifically contemplates and discloses embodiments that "consist essentially of" those elements and that "consist of" those elements.

[0107] As used herein the terms "consists essentially of," "consisting essentially of," and the like are to be construed as a semi-closed terms, meaning that no other ingredients which materially affect the basic and novel characteristics of an embodiment are included.

[0108] As used herein, the terms "consists of," "consisting of," and the like are to be construed as closed terms, such that an embodiment "consisting of" a particular set of elements excludes any element, step, or ingredient not specified in the embodiment.

[0109] As used herein, the terms "Target Area Hair Count" or "TAHC" refer to the change, from baseline, in the number of non-vellus hairs in a target area of the scalp. The target area can be, for example, 1 cm 2 or a circle of a diameter of 1 inch (5.1 cm 2 ). [0110] As used herein, the terms "Hair Growth Assessment" or "HGA" refer to a score given by the subject comparing the baseline standardized global photo of the subject's scalp with a "real time" standardized global photo.

[0111] As used herein, the terms "Investigator's Global Assessment" or "IGA" refer to a score given by an evaluator comparing the baseline standardized global photo of the subject's scalp with a "real time" standardized global photo.

[0112] As used herein, the term "Bis In Die" or "BID" means "twice a day".

[0113] As used herein, the term "allocation group" refers to a group of people participating in a clinical trial that are randomly allocated to either the group receiving the treatment under investigation or to a group receiving standard treatment (or placebo treatment) as the control.

[0114] As used herein, the term "ethanol," means ethyl alcohol, i.e. CH3CH2OH, and includes pure (absolute) ethanol and 96° ethanol, the latter being ethanol containing water in an amount typically ranging from about 4% to about 5.1% by volume.

[0115] The present disclosure provides fully solubilized pharmaceutical formulations of cortexolone-17a-propionate comprising a solvent and at least 2.1 weight percent cortexolone- 17a-propionate up to about 20 weight percent cortexolone-17a-propionate, including all intermediate values there between. In particular embodiments, the formulation can comprise at least 2.1 weight percent up to about 17 weight percent cortexolone-17a-propionate, at least 2.5 weight percent up to about 17 weight percent cortexolone-17a-propionate, at least 2.5 weight percent up to about 15 weight percent cortexolone-17a-propionate, or at least 3 weight percent up to about 15 weight percent cortexolone-17a-propionate. In other embodiments, the formulation can comprise about 6, about 7, about 7.5, about 8, about 9, about 10, about 11, about 12, about 13, about 14, or about 15 weight percent of cortexolone- 17a-propionate.

[0116] The present disclosure also provides fully solubilized pharmaceutical formulations of cortexolone-17a-propionate comprising a solvent and at least 2.1 weight percent cortexolone-17a-propionate up to about 5.5 weight percent cortexolone-17a-propionate, including all intermediate values there between. In particular embodiments, the formulation can comprise at least 2.2 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.3 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.4 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.5 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.6 up to about 5.5 weight percent cortexolone-17a- propionate, at least 2.7 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.8 up to about 5.5 weight percent cortexolone-17a-propionate, at least 2.9 up to about 5.5 weight percent cortexolone-17a-propionate, or at least 3.0 up to about 5.5 weight percent cortexolone-17a-propionate. In other embodiments, the formulation can comprise about 2.3, about 2.5, about 3, about 3.5, about 4, about 4.5, about 5, or about 5.5 weight percent cortexolone-17a-propionate. In a particular embodiment, the formulation can comprise about 5 weight percent of cortexolone-17a-propionate. In each of these embodiments, the cortexolone-17a-propionate is fully solubilized in the formulation. As used herein, "fully solubilized" means at least 95 weight percent, at least 98 weight percent, at least 99 weight percent, or at least 99.9 weight percent of the cortexolone-17a-propionate is solubilized in the formulation.

[0117] Cortexolone-17a-propionate solubility exceeding 2 weight percent has not been previously reported and was difficult to achieve. For Example, WO 2009/019138 discloses formulations having up to 2 weight percent cortexolone-17a-propionate, but does not teach or suggest whether higher concentration formulations can be prepared or how such formulation should be prepared.

[0118] Similarly, Celasco, et al., Arzneim.-Forsch./Drug Res. 54, No. 12, 881-886 (2004), teaches suspending a 5 mg sample of cortexolone-17a-propionate in 0.2 ml suspending vehicle. But like WO 2009/019138, Celasco is silent on whether fully solubilized high- concentration formulations can or should be prepared.

[0119] In certain embodiments, the formulation is anhydrous and includes less than about

5 weight percent water. In further embodiments, the anhydrous formulation includes less than about 3 weight percent water. [0120] In certain embodiments, the formulation can be a liquid (including, for example, a solution or a suspension or an emulsion or a microemulsion) or can be semi-solid (including, for example, a cream, a gel, or an ointment, or a foam). Regardless of the form of the formulation (solution, gel, emulsion, etc.), the formulation has a suitable consistency to be spread on the scalp and/or on the skin.

[0121] In one embodiment, the formulation can be a liquid formulation.

[0122] In another embodiment, the formulation can be anhydrous. In another embodiment, the formulation can be a solution. In some embodiments, the solution is anhydrous having less than about 5 weight percent water. In other embodiments, the anhydrous solution has less than about 3 weight percent water.

[0123] In one embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration wherein the formulation comprises cortexolone-17a- propionate in an amount of at least 2.1 weight percent and one or more pharmaceutically acceptable solvents.

[0124] In another embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration wherein the formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 20 weight percent and one or more pharmaceutically acceptable solvents. Suitable ranges and amounts of cortexolone-17a-propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0125] In another embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration wherein the formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 5.5 weight percent and one or more pharmaceutically acceptable solvents. Suitable ranges and amounts of cortexolone-17a-propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0126] In still another embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration for use in the treatment and/or prevention of alopecia, wherein the formulation comprises cortexolone-17a-propionate in an amount of at least 2.1 weight percent and one or more pharmaceutically acceptable solvents.

[0127] In another embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration for use in the treatment and/or prevention of alopecia, wherein the formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 20 weight percent and one or more pharmaceutically acceptable solvents. Suitable ranges and amounts of cortexolone-17a- propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0128] In yet another embodiment, the present disclosure provides a pharmaceutical formulation suitable for topical administration for use in the treatment and/or prevention of alopecia, wherein the formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 5.5 weight percent and one or more pharmaceutically acceptable solvents. Suitable ranges and amounts of cortexolone-17a- propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0129] In another embodiment, the present disclosure provides a method for treating and/or preventing alopecia in a mammal in need thereof, the method comprising topically administering a effective amount of a pharmaceutical formulation suitable for topical administration, wherein the formulation comprises cortexolone-17a-propionate in an amount of at least 2.1 weight percent and one or more pharmaceutically acceptable solvents.

[0130] A further aspect of the invention is a method for treating and/or preventing alopecia in a mammal in need thereof, said method comprising the topical administration of a therapeutic amount of a pharmaceutical formulation suitable for topical administration, wherein the formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 20 weight percent and one or more pharmaceutically acceptable solvents. Suitable ranges and amounts of cortexolone-17a-propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0131] A further aspect of the present invention is a method for treating and/or preventing alopecia in a mammal in need thereof, said method comprising the topical administration of a therapeutic amount of a pharmaceutical formulation suitable for topical administration, wherein said formulation comprises cortexolone-17a-propionate in an amount ranging from about 2.1 weight percent to about 5.5 weight percent and one or more physiologically acceptable solvents. Suitable ranges and amounts of cortexolone-17a-propionate in this aspect of the invention are described above in connection with topical pharmaceutical formulations comprising cortexolone-17a-propionate.

[0132] In certain embodiments, the mammal is a human.

[0133] In particular embodiments, topical administration comprises application of the formulation to the skin and/or the scalp.

[0134] In particular embodiments, the alopecia is androgenetic alopecia (AGA), alopecia areata (including diffuse alopecia areata, alopecia areata monolocularis, alopecia areata multilocularis, ophiasis, alopecia totalis and alopecia universalis), telogen effluvium, anagen effluvium, and traction alopecia. In particular embodiments, the alopecia is AGA.

[0135] In certain embodiments, the pharmaceutical formulation can be in form of a solution, a gel, a fluid ointment, a suspension, a microemulsion, or a foam.

[0136] In addition to the solvent and the specified amount of cortexolone-17a-propionate, the formulation described herein can also optionally contain at least one penetration enhancer, and optionally at least one pharmaceutically acceptable excipient.

[0137] In certain embodiments, the formulation can further include at least one antioxidant, at least one emulsifier, or a combination of the foregoing.

The Solvent

[0138] In certain embodiments, the solvent can be anhydrous including less than about 5 weight percent water. In other embodiments, the solvent can be anhydrous and include less than about 3 weight percent water. [0139] In certain embodiments, the solvent can be selected from the group comprising or the group consisting of: water, a C1-C7 alcohol, a polyol ether, a polyol, a natural oil, an ester, tricaprylin (2,3-di(octanoyloxy)propyl octanoate), a medium-chain triglyceride, caprylocaproyl polyoxyl-8 glycerides, and combinations of any of the foregoing.

[0140] In some embodiments, the formulation can comprise at least about 50 weight percent solvent. In other embodiments, the formulation can comprise at least about 60 weight percent solvent. In other embodiments, the formulation can comprise at least about 70 weight percent solvent. In other embodiments, the formulation can comprise at least about 80 weight percent solvent. In other embodiments, the formulation can comprise at least about 85 weight percent solvent, at least about 90 weight percent solvent, at least about 91 weight percent solvent, at least about 92 weight percent solvent; at least about 93 weight percent solvent; at least about 94 weight percent solvent; or at least about 95 weight percent solvent.

[0141] In certain embodiments, the solvent can comprise a mixture of a C1-C7 alcohol, a polyol ether, a polyol. In such embodiments, the formulation can comprise from about 10 to about 50 weight percent of the polyol ether; from about 5 weight percent to about 55 weight percent of the polyol; and about 5 to about 50 weight percent of the C1-C7 alcohol. In a particular embodiment, the mixture of the C1-C7 alcohol, the polyol ether, and the polyol can be present at about a 1:1:1 ratio on a w/w/w basis. In certain embodiments, each of the C1-C7 alcohol, the polyol ether, and the polyol are present at about 30 weight percent.

[0142] In particular embodiments, the formulation can comprise from about 15 to about

45 weight percent of the polyol ether; from about 20 to about 40 weight percent of the polyol ether; from about 25 to about 35 weight percent of the polyol ether; or from about 30 to about 35 weight percent of the polyol ether. In particular embodiments, the formulation can comprise about 30 weight percent of the polyol ether. In other embodiments, the formulation can comprise about 32 weight percent of the polyol ether.

[0143] In particular embodiments, the polyol ether is selected from the group comprising or the group consisting of: polyethylene glycol, polypropylene glycol, polyethylene- polypropylene triblock copolymers, dipropylene glycol, diethylene glycol monoethyl ether (TRANSCUTOL ® ), and combinations thereof.

[0144] When the polyol ether is a polyethylene glycol, it can be selected from the group comprising or the group consisting of: polyethylene glycol 200, polyethylene glycol 300, polyethylene glycol 400, polyethylene glycol 540, and polyethylene glycol 600. In particular embodiments, the polyol ether can be polyethylene glycol 200. In other embodiments, the polyol ether can be polyethylene glycol 400. In still other embodiments, the polyol ether can be diethylene glycol monoethyl ether.

[0145] In particular embodiments, the polyol can be selected from the group comprising or the group consisting of: propylene glycol, ethylene glycol, glycerol, hexanetriol, and combinations thereof. In particular embodiments, the polyol can be glycerol. In other embodiments, the polyol can be propylene glycol.

[0146] In particular embodiments, the polyol can be present in an amount ranging from about 5 weight percent to about 55 weight percent of the total formulation; from about 10 weight percent to about 50 weight percent of the total formulation; from about 20 weight percent to about 45 weight percent of the total formulation; and in certain embodiments, from about 25 weight percent to about 40 weight percent. In a particular embodiment, the polyol comprises about 30 weight percent of the total formulation. In a further embodiment, the polyol comprising about 30 weight percent of the total formulation can be propylene glycol.

[0147] In some embodiments, the formulation can comprise from about 5 to about 50 weight percent of the C1-C7 alcohol. In particular embodiments, the formulation can comprise from about 15 to about 45 weight percent of the Ci-C 7 alcohol; from about 20 to about 40 weight percent of the C C 7 alcohol; or from about 25 to about 35 weight percent of the C C 7 alcohol. In particular embodiments, the formulation can comprise about 30 weight percent of the C C 7 alcohol. In still further embodiments, the formulation can comprise the Ci-C 7 alcohol in an amount ranging from about 10 weight percent to about 40 weight percent, or from about 15 weight percent to about 35 weight percent. [0148] In particular embodiments, the C1-C7 alcohol is selected from the group comprising or the group consisting of: methanol, ethanol, isopropanol, n-butanol, n-propanol, and benzyl alcohol. In particular embodiments, the C -C7 alcohol is ethanol. In some embodiments, the ethanol comprises about 30 weight percent of the total formulation. In other embodiments, the C1-C7 alcohol is isopropanol.

[0149] In some embodiments, the C1-C7 alcohol can contain a small water fraction, generally in the range of from about 4 percent to about 5.1 percent by volume. In embodiments wherein the formulation is anhydrous, the C1-C7 alcohol having from about 4 percent to about 5.1 percent water by volume can be present in the formulation in an amount such that the water content in the finished formulation itself is less than about 5 weight percent or less than about 3 weight percent.

[0150] In certain embodiments, the solvent can comprise a polyol, a polyol ether, and a Ci-

C 7 alcohol. In particular embodiments, the solvent can comprise ethanol as the C1-C7 alcohol, diethylene glycol monomethyl ether as the polyol ether, and propylene glycol as the polyol. These solvents can be present in any acceptable ratio, but typically are present such that each is about 25 to about 35 weight percent of the formulation.

Penetration Enhancers

[0151] In certain embodiments, in addition to the solvent, the formulations disclosed herein can include one or more penetration enhancers. Without wishing to be bound by a particular theory, penetration enhancers are believed to beneficially affect delivery of the therapeutic agent into the skin (including the scalp) and/or the hair follicles. Advantageously, and in some embodiments, the solvent or some component(s) of the solvent act as a penetration enhancer. For example, in embodiments wherein the formulation comprises ethanol and/or diethylene glycol monoethyl ether, these solvents can also act to enhance penetration of the active agent into the skin (including the scalp) and/or the follicles. The presence of ethanol and/or diethylene glycol monoethyl ether notwithstanding, the formulations described herein can optionally further include additional penetration enhancers. [0152] Examples of further penetration enhancer that can be incorporated into formulations described herein include, but are not limited to, polyoxyethylene alkyl ethers, polyoxyl glycerides, dimethyl sulfoxide, pyrrolidone, N-methyl-2-pyrrolidone, diethylene glycol monoethyl ether (TRANSCUTOL ® ), dimethyl isosorbide, diethyl sebacate, azone, menthol, nerol, camphor, methyl salicylate, Tween 80, SDS, benzalkonium chloride, polyoxyl 40 hydrogenated castor oil (CREMOPHOR ® RH40, KOLLIPHOR ® RH40), didecyldimethylammonium bromide (DDAB), didecyltrimethylammonium bromide (DTAB), fatty acids esters such as isopropyl myristate, isopropyl palmitate and the like, fatty acids such as oleic acid, palmitic acid, linoleic acid, and salts thereof, fatty alcohols such as oleyl alcohol, myristyl alcohol, stearyl alcohol and the like, medium-chain triglycerides, and combinations of any of the foregoing.

[0153] In one embodiment, the penetration enhancer is dimethyl isosorbide. In another embodiment, the penetration enhancer is a mixture comprising dimethyl isosorbide and diethylene glycol monoethyl ether. In another embodiment, the at least one penetration enhancer is dimethyl sulfoxide.

[0154] In certain embodiments, the penetration enhancer can be present in an amount ranging from about 1 weight percent to about 50 weight percent with respect to the total weight of the formulation. In other embodiments, the penetration enhancer can be present from about 2 weight percent to about 40 weight percent with respect to the total weight of the formulation; or from about 5 weight percent to about 35 weight percent with respect to the total weight of the formulation. According to some embodiments, the skin penetration enhancer can be present in the formulation described herein in an amount of about 1% weight percent, about 2 weight percent, about 5 weight percent, about 10 weight percent, about 15 weight percent, about 20 weight percent, about 25 weight percent, about 30 weight percent, about 35 weight percent, about 40 weight percent, about 45 weight percent, or about 50 weight percent with respect to the total weight of the formulation. Other Ingredients

[0155] Because oxidation can result in degradation of the active agent, in some embodiments, the formulation can further include up to about 1 weight percent of an antioxidant. The antioxidant can be selected from the group comprising or the group consisting of: BHT, BHA, ascorbyl palmitate, ascorbic acid, alpha tocopherol (also known as Vitamin E), propyl gallate, and combinations of any of the foregoing.

[0156] In particular embodiments, the formulation can include up to about 0.5 weight percent of an antioxidant or include about 0.5 weight percent antioxidant. In particular embodiments, the antioxidant can be ascorbyl palmitate. In other embodiments, the antioxidant can be BHA. In still other embodiments, the antioxidant can be BHT. In still further embodiments, formulation can include about 0.5 weight percent ascorbyl palmitate.

[0157] In some embodiments, the formulation can further include an emulsifier. Without wishing to be bound to any particular theory, it is believed that emulsifiers, when present, assist or facilitate dissolution of any solid substances, such as the active agent, in the formulation. In other embodiments, however, the emulsifier can be present to facilitate incorporation of two non-miscible liquids into each other (e.g. an emulsion or microemulsion).

[0158] In other embodiments, and without wishing to be bound by any particular theory, an emulsifier can increase product spreadability. For example, and without wishing to be bound to a particular theory, when the formulation is a liquid solution, the presence of a suitable amount of an emulsifier is believed to decrease the surface tension between the formulation and the lipid environment of the superficial layer of the skin and/or scalp. This makes it easier to spread the formulation and is believed to assist penetration of the therapeutic agent into skin (including the scalp) and/or hair follicles.

[0159] In certain embodiments, the emulsifier can be selected from the group comprising or the group consisting of: polyethylene glycol (PEG)-fatty acid monoesters such as PEG-15 hydroxystearate (also known as polyoxyl-15-hydroxystearate), PEG-30 stearate, PEG-40 laurate, PEG-40 oleate and the like; polyoxyethylene sorbitan fatty acid esters such as polysorbate 20, polysorbate 60, polysorbate 80 and the like; polyoxyethylene alkyl ethers such as PEG-20 cetostearyl ether, polyoxyl 25 cetostearyl, cetomacrogol 1000 and the like; sorbitan fatty acid esters such as sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, and the like; propylene glycol esters of fatty acids; polyglycerol esters of fatty acids; polyoxyethylene castor oil derivatives such as polyoxyl 5 castor oil, polyoxyl 15 castor oil, polyoxyl 35 castor oil, polyoxyl 40 hydrogenated castor oil and the like; caprylocapryl polyoxyil- 8 glycerides; polyoxylglycerides such as caprylocaproyl polyoxylglycerides, lauroyl polyoxylglycerides, oleoyi polyoxylglycerides, and the like; along with combinations of any of the foreoging.

[0160] In particular embodiments, the formulation can include the emulsifier in an amount of up to about 0.5 weight percent of the total formulation. In some embodiments, the emulsifier can be present in an amount ranging from 0.05 to 0.2 weight percent. In still further embodiments, the formulation can include the emulsifier in an amount up to about 0.1 weight percent or it can include about 0.1 weight percent emulsifier. In particular embodiments, the emulsifier can be polyoxyl 40 hydrogenated castor oil. In other embodiments, the emulsifier can be polyoxyl-15-hydroxystearate. In particular embodiments, the emulsifier can be polysorbate 80. In still further embodiments, the emulsifier is present in the formulation at about 0.1 weight percent.

Pharmacokinetic Parameters

[0161] It has been surprisingly found that the formulations described herein provide acceptable pharmacokinetics profiles of both cortexolone-17a-propionate and/or its metabolites and that the formulations described herein appear to provide excellent local delivery of the therapeutic agent while minimizing systemic exposure. This is a beneficial result because, without wishing to be bound by any particular theory, it is believed that the active agent is most effective when delivered to the hair follicles directly, rather than through the systemic circulation. Thus, it is an advantage of formulations of the present disclosure to be able to deliver the active agent with sufficient penetration to reach the follicles, but without so much penetration as to be widely systemically available after topical application. [0162] For example, in certain embodiments, the formulations described herein provide a mean steady-state C max of cortexolone-17a-propionate of about 2 ng/ml to about 6 ng/ml; of about 2.5 ng/ml to about 5.5 ng/ml; or of about 3 ng/ml to about 5 ng/ml. In certain embodiments the mean steady-state C max of cortexolone-17a-propionate can be about 4 ng/ml, or about 3.8 ng/ml. In other embodiments, the mean steady-state C max can be less than about 6 ng/ml, less than about 5 ng/ml, less than about 4 ng/ml, less than about 3 ng/ml, or less than about 2 ng/ml.

[0163] In certain embodiments, the formulation described herein provides a mean steady- state T max of cortexolone-17a -propionate of from about 2 to about 7 hours; from about 2.5 to about 6.5 hours; from about 3 to about 6 hours; from about 3.5 to about 5 hours; or from about 4 to about 5 hours. In particular embodiments, the mean steady-state T max of cortexolone-17a-propionate can be about 4.5 or 4.4 hours. In certain embodiments, the mean steady-state T max of cortexolone-17a -propionate can be less than about 8 hours, less than about 7 hours, less than about 6 hours, less than about 5 hours, or less than about 4 hours.

[0164] The present formulation also provides a desirable AUC X of from about 20 (ng*h)/ml to about 55 (ng*h)/ml; of from about 25 (ng*h)/ml to about 50 (ng*h)/ml; of from about 25 (ng*h)/ml to about 45 (ng*h)/ml; or from about 30 (ng*h)/ml to about 40 (ng*h)/ml. In certain embodiments, the mean AUC X can be about 35 (ng*h)/ml or about 37 (ng*h)/ml. In other embodiments, the AUC X can be less than about 55 (ng*h)/ml, less than about 45 (ng*h)/ml, less than about 40 (ng*h)/ml, or less than about 35 (ng*h)/ml.

[0165] The present formulation also provides a desirable excretion profile for cortexolone-

17a-propionate. For example, after initial topical application of the formulation, in certain embodiments, less than about 0.5 percent of the cortexolone-17a-propionate administered can be detected in a patient's urine. In other embodiments, less than about 0.4, less than about 0.35, less than about 0.3, or less than about 0.25, less than about 0.2, or less than about 0.15 percent of the cortexolone-17a-propionate can be detected in a given patient's urine. In certain embodiments, no cortexolone-17a-propionate is detectable in a given patient's urine. [0166] In certain embodiments, after steady state has been achieved, less than about 700 μg of the cortexolone-17a-propionate administered can be detected in a patient's urine. In other embodiments, less than about 650, less than about 600, less than about 500, less than about 400, less than about 300, or less than about 250 μg of the cortexolone-17a-propionate can be detected in a given patient's urine. In certain embodiments, even after steady state has been achieved, no cortexolone-17a-propionate can be detected in a given patient's urine.

[0167] In certain embodiments, cortexolone can also be detected in a patient's urine. For example, in certain embodiments after initial topical application of the formulation less than about 0.5 μg of cortexolone can be detected in a patient's urine. In other embodiments, less than about 0.4, less than about 0.35, less than about 0.3, or less than about 0.25, less than about 0.2, or less than about 0.15 μg of cortexolone can be detected in a given patient's urine. In certain embodiments, no cortexolone can be detected in a given patient's urine.

[0168] After steady state has been achieved, and in certain embodiments, less than about

2 μg of cortexolone can be detected in a patient's urine. In other embodiments, less than about 1.75, less than about 1.5, less than about 1.25, less than about 1, less than about 0.75, or less than about 0.5 μg of the cortexolone can be detected in a given patient's urine. In certain embodiments, even after steady state has been achieved, no cortexolone can be detected in a given patient's urine.

[0169] In certain embodiments, tetrahydrocortexolone can also be detected in a patient's urine. For example, in certain embodiments after initial topical application of the formulation less than about 150 μg of tetrahydrocortexolone can be detected in a patient's urine. In other embodiments, less than about 125, less than about 100, less than about 75, less than about 65, less than about 55, or less than about 50 μg of tetrahydrocortexolone can be detected in a given patient's urine. In certain embodiments, no tetrahydrocortexolone can be detected in a given patient's urine.

[0170] After steady state has been achieved, and in certain embodiments, less than about

400 μg of tetrahydrocortexolone can be detected in a patient's urine. In other embodiments, less than about 350, less than about 325, less than about 300, less than about 225, less than about 150, or less than about 230 μg of the tetrahydrocortexolone can be detected in a given patient's urine. In certain embodiments, even after steady state has been achieved, no tetrahydrocortexolone can be detected in a given patient's urine.

Modes of Administration

[0171] The formulations described herein can be administered according to varying schedules. In one embodiment, the mode of administration of the formulations can be continuous. For example, the formulations can be applied topically once a day, twice daily, three times a day, four times a day, or more, as specified by a physician. In particular embodiments, a formulation described herein can be applied topically once a day or twice daily. According to a particular embodiment, the formulation described herein can be applied topically once a day. According to another embodiment, the formulation described herein can be applied topically twice daily.

[0172] In certain embodiments, a dosing regimen can be tapered. That is, the formulation can be applied once a day for a first period of time, twice daily thereafter for a second period of time, three times a day thereafter for a third period of time, and so on. In a particular embodiment, the formulation can be applied once a day on the first day, and twice daily thereafter, with the appropriate duration of treatment determined by a subject's physician.

[0173] In certain embodiments, the formulation can be applied over the course of a period of days, weeks, or months. For example, the formulation can be applied one, two, three, four, or five times a day for up to: 1, 2, 3, 4, 5, 6, or 7 days; about 2 weeks, about 3 weeks, or about 4 weeks; about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about a year (i.e. about 12 months), about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months or about 24 months. According to a particular embodiment, the formulation can be applied once on the first day and twice daily thereafter for about 4 weeks. [0174] In another embodiments, the formulation can be topically applied to the scalp once a day for 1 month, once a day for 2 months, once a day for 3 months, once a day for 4 months, once a day 5 months, once a day for 6 months, once a day for 8 months, once a day for 12 months, once a day for 14 months, once a day for 16 months, once a day for 18 months, once a day for 20 months, once a day for 22 months, or once a day for 24 months. In certain embodiments, the formulation can be topically applied on the scalp once daily for 6 months. In certain embodiments, the formulation can be topically applied on the scalp once daily for 12 months.

[0175] In other embodiments, the formulation can be topically applied on the scalp twice daily for about 1 month, for about 2 months; for about 3 months; for about 4 months; for about 5 months; for about 6 months, for about 8 months, for about 12 months, for about 14 months, for about 16 months, for about 18 months, for about 20 months, for about 22 months, or for about 24 months. In certain embodiments, the formulation can be topically applied on the scalp twice daily for 6 months. In certain embodiments, the formulation can be topically applied on the scalp twice daily for 12 months.

[0176] In other embodiments, the formulation can be applied more than twice daily (i.e.

TID, QID, etc) for about 1 month, for about 2 months; for about 3 months; for about 4 months; for about 5 months; for about 6 months, for about 8 months, for about 12 months, for about 14 months, for about 16 months, for about 18 months, for about 20 months, for about 22 months, or for about 24 months. In certain embodiments, the formulation can be topically applied on the scalp more than twice daily (i.e. TID, QID, etc) for 6 months. In certain embodiments, the formulation can be topically applied on the scalp more than twice daily (i.e. TID, QID, etc) for 12 months.

[0177] In another embodiment, the mode of administration of the formulations can be cyclic. For example, the formulations can first be applied in a continuous way as described above for a desired period of time, the application is then discontinued for a period of time, such as a few days, and then the application of the formulations is started again as described above. The treatment period can include one or more cycles which can be the same or different. In certain embodiments, the treatment period can be as follows: a) the formulation is topically applied on the scalp for a period of time, such as 4 months, by continuous administration, b) the application is discontinued for a few days, such as 2-5 days, and 3) the topical application is continued for an additional period of time, such as 6 months.

[0178] The amount of cortexolone-17a-propionate that can be applied to a patient in need thereof can vary. In some embodiments, about 400 mg of cortexolone-17a-propionate can be applied, about 375 mg of cortexolone-17a-propionate can be applied, about 350 mg of cortexolone-17a-propionate can be applied, about 325 mg of cortexolone-17a-propionate can be applied, about 300 mg of cortexolone-17a-propionate can be applied, about 275 mg of cortexolone-17a-propionate can be applied, about 250 mg of cortexolone-17a-propionate can be applied, or about 225 mg of cortexolone-17a-propionate can be applied. In some embodiments, about 200 mg of cortexolone-17a-propionate can be applied, about 175 mg of cortexolone-17a-propionate can be applied, about 150 mg of cortexolone-17a-propionate can be applied, about 125 mg of cortexolone-17a-propionate can be applied, about 100 mg of cortexolone-17a-propionate can be applied, about 75 mg of cortexolone-17a-propionate can be applied, about 50 mg of cortexolone-17a-propionate can be applied, about 25 mg of cortexolone-17a-propionate can be applied, about 12.5 mg of cortexolone-17a-propionate can be applied, or about 6.25 mg of cortexolone-17a-propionate can be applied. Determination of the appropriate amount of the formulation that should be administered to a given patient in a single application is within the skill of the ordinarily skilled physician.

[0179] In particular embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 400 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 350 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 300 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 250 mg. [0180] In particular embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 200 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 170 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 150 mg. In particular embodiments, the amount of cortexolone-17a-propionate in a single application can range from about 20 mg to about 100 mg. In particular embodiments, the amount of cortexolone-17a-propionate in a single application can be about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, or about 100 mg. In another embodiment, the amount of cortexolone-17a- propionate in a single application can be about 25 mg. In another embodiment, the amount of cortexolone-17a-propionate in a single application can be about 30 mg. In yet another embodiment, the amount of cortexolone-17a-propionate in a single application can be about 50 mg. In yet another embodiment, the amount of cortexolone-17a-propionate in a single application can be about 75 mg. In yet another embodiment, the amount of cortexolone-17a- propionate in a single application can be about 80 mg. In yet another embodiment, the amount of cortexolone-17a-propionate in a single application can be about 100 mg. In other embodiments, the amount of cortexolone-17a-propionate in a single application can be about 110 mg, about 111 mg, about 112 mg, about 113 mg, about 114 mg, about 115 mg, about 116 mg, about 117 mg, about 118 mg, about 119 mg, or about 200 mg. In yet another embodiment, the amount of cortexolone-17a-propionate in a single application can be about 150 mg.

[0181] By way of example only, about 50 mg of cortexolone-17a-propionate could be administered in 1 ml of an embodiment of a formulation disclosed herein, wherein the formulation comprises about 5 weight percent cortexolone-17a-propionate.

[0182] In some embodiments, the formulation can be self-administered by the patient once a day or twice daily. In particular embodiments, when the formulation is in liquid form having an active agent concentration of about 5 weight percent, the formulation can be self- administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0183] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 2.5 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0184] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 3 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about

1.5 ml, and in further embodiments at about 1ml.

[0185] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 7.5 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0186] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 8 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about

1.5 ml, and in further embodiments at about 1ml.

[0187] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 10 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0188] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 15 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0189] In other embodiments, when the formulation is in liquid form having an active agent concentration of about 20 weight percent, the formulation can be self-administered once a day or twice daily at a dose ranging from about 0.2 to about 2.0 ml, from about 0.5 to about 1.5 ml, and in further embodiments at about 1ml.

[0190] The formulation described herein can be applied to any body surface in need of treatment, such as the scalp, face (e.g., the eyebrow, eyelashes, upper lip, lower lip, chin, cheeks, beard area, or mustache area), arms, armpits, legs, chest, abdomen, or any combination of the foregoing. In certain embodiments, treatment is not delivered to the face. In other embodiments, the formulation can be applied to the scalp.

Efficacy Parameters

[0191] It has been surprisingly found that the pharmaceutical formulations described herein are able to maximize the delivery of the therapeutic agent cortexolone-17a-propionate to the target site (i.e. the hair follicle), minimizing its systemic adsorption. Thus, it is an advantage of the pharmaceutical formulations of the present disclosure to be able to deliver the active agent with sufficient penetration to reach the follicles, but without so much penetration as to be widely systemically available after topical application. This translates into a pharmacological profile characterized by efficacy in promoting hair regrowth (local activity on hair follicles), with absence of significant adverse events, including those due to systemic antiandrogenic effects (low systemic adsorption of the therapeutic agent).

[0192] In some embodiments, the formulations described herein provide a mean change from baseline in Target Area Hair Count equal to or higher than 8 hairs/cm 2 after 6 months of daily or BID application.

[0193] In other embodiments, the formulations described herein provide a mean change from baseline in Target Area Hair Count equal to or higher than 9 hairs/cm 2 after 6 months of daily or BID application.

[0194] In other embodiments, the formulations described herein provide a mean change from baseline in Target Area Hair Count equal to or higher than 10 hairs/cm 2 after 6 months of daily or BID application. [0195] In other embodiments, the formulations described herein provide a mean change from baseline in Target Area Hair Count equal to or higher than 11 hairs/cm 2 after 6 months of daily or BID application.

[0196] In other embodiments, the formulations described herein provide a mean change from baseline in Target Area Hair Count equal to or higher than 12 hairs/cm 2 after 6 months of daily or BID application.

[0197] In some embodiments, the formulations described herein provide a weighted average HGA score equal to or higher than 0.20 after 6 months of daily or BID application.

[0198] In other embodiments, the formulations described herein provide a weighted average HGA score equal to or higher than 0.30 after 6 months of daily or BID application.

[0199] In some embodiments, the formulations described herein provide a weighted average HGA score equal to or higher than 0.40 after 6 months of daily or BID application.

[0200] In some embodiments, the formulations described herein provide a weighted average IGA score equal to or higher than 0.10 after 6 months of daily or BID application.

[0201] In other embodiments, the formulations described herein provide a weighted average IGA score equal to or higher than 0.20 after 6 months of daily or BID application.

[0202] In other embodiments, the formulations described herein provide a weighted average IGA score equal to or higher than 0.30 after 6 months of daily or BID application.

[0203] In some embodiments, the formulations described herein provide a favorable

(positive) HGA score in at least about 10 % of subjects after 6 months of daily or BID application.

[0204] In other embodiments, the formulations described herein provide a favorable

(positive) HGA score in at least about 20 % of subjects after 6 months of daily or BID application.

[0205] In other embodiments, the formulations described herein provide a favorable

(positive) HGA score in at least about 30 % of subjects after 6 months of daily or BID application. [0206] In some embodiments, the formulations described herein provide a favorable

(positive) IGA score in at least about 10 % of subjects after 6 months of daily or BID application.

[0207] In other embodiments, the formulations described herein provide a favorable

(positive) IGA score in at least about 20 % of subjects after 6 months of daily or BID application.

[0208] In other embodiments, the formulations described herein provide a favorable

(positive) IGA score in at least about 30 % of subjects after 6 months of daily or BID application.

[0209] In other embodiments, the formulations described herein provide a favorable

(positive) IGA score in at least about 40 % of subjects after 6 months of daily or BID application.

[0210] In some embodiments, the formulations described herein are effective in stimulating the hair re-growth, providing the TAHC, HGA scores and IGA scores above disclosed, without exerting a systemic antiandrogenic activity. Such formulations are devoid of side effects attributable to systemic antiandrogenic effects. Said side effects include, but are not limited to, decreased libido, erectile dysfunction (impotence), ejaculation disorders, and decreased volume of ejaculate.

Storage Stability

[0211] Storage stability is an important metric for pharmaceutical products. In general, greater stability means that a given formulation is both easier to transport and store, increasing the likelihood that it will be stocked by pharmacies and that patients will not have to be concerned with special storage instructions. The formulations described herein have a desirable stability profile allowing for storage of the final formulation for at least about 3 months, at least about 6 months, at least about 9 months, at least about 12 months, at least about 15 months, at least about 18 months, at least about 21 months, or at least about two years - each at room or refrigerated temperatures.

[0212] For example, one of the main degradation pathways of cortexolone-17a-propionate is transesterification to cortexolone-21-propionate (17a-hydroxy-21-propionyloxy-pregna-4- ene-3,20-dione):

[0213] It has now been surprisingly discovered that by maintaining the formulations disclosed herein at a pH of less than about 6, and in certain embodiments, less than about 5, between about 4 and about 5, or at about 4, this degradation process can be dramatically slowed. In particular embodiments, the pH can be 4. The appropriate pH can be obtained via addition of a suitable amount of a pH modifier.

[0214] Acceptable pH modifiers include those pharmaceutically acceptable organic and inorganic acids known to those of ordinary skill in the art. Examples of such acids, include, but are not limited to l-hydroxy-2-naphthoic acid; 2,2-dichloroacetic acid; 2- hydroxyethanesulfonic acid; 2-oxoglutaric acid; 4-acetamidobenzoic acid; 4-aminosalicylic acid; acetic acid; adipic acid; L-ascorbic acid; L-aspartic acid; benzenesulfonic acid; benzoic acid; (+)- camphoric acid; (+)-camphor-10-sulfonic acid; capric acid (decanoic acid); caproic acid (hexanoic acid); caprylic acid (octanoic acid); carbonic acid; cinnamic acid; citric acid; cyclamic acid; dodecylsulfuric acid; ethane-l,2-disulfonic acid; ethanesulfonic acid; formic acid; fumaric acid; galactaric acid; gentisic acid; D-glucoheptonic acid; D-gluconic acid; D-glucuronic acid; glutamic acid; glutaric acid; glycerophosphoric acid; glycolic acid; hippuric acid; hydrobromic acid; hydrochloric acid; isobutyric acid; lactic acid; lactobionic acid; lauric acid; maleic acid; L- malic acid; malonic acid; mandelic acid; methanesulfonic acid; naphthalene-l,5-disulfonic acid; naphthalene-2-sulfonic acid; nicotinic acid; nitric acid; oleic acid; oxalic acid; palmitic acid; pamoic acid; phosphoric acid; proprionic acid; L-pyroglutamic acid; salicylic acid; sebacic acid; stearic acid; succinic acid; sulfuric acid; L-tartaric acid; thiocyanic acid; p-toluenesulfonic acid; and undecylenic acid. In particular embodiments, the pH modifying agent can be citric acid. [0215] In certain embodiments, the formulations described herein can, at room or refrigerated temperatures, have less than about 5 weight percent cortexolone-21-propionate or other degradation product after storage for a period of about 24 months.

[0216] While pH can be important to stability, it has also been discovered that addition of an antioxidant to the formulation can assist in maintaining storage stability. The antioxidant can be present in addition to a pH modifier. But in some embodiments, the pH modifier can be substantially or completely absent. The antioxidant can be present in the amounts specified elsewhere herein.

[0217] It has surprisingly found that a formulation containing cortexolone-17a-propionate as disclosed herein provides acceptable pharmacokinetics profiles of both cortexolone 17a- propionate and/or its metabolites, providing a therapeutically effective amount of the therapeutic agent to the scalp and/or skin while minimizing systemic exposure. Without wishing to be bound to a particular theory, it is believed that the formulations disclosed herein deliver the therapeutic agent to the derma, with deeper penetration minimized by the nature of the formulations disclosed herein. This effect is evidenced in Franz cell data reported in Examples 6 and 7, below.

[0218] Thanks to such a favorable pharmacokinetic profile, with high therapeutic levels of the therapeutic agent at the target site and low levels in the systemic circulation, the formulation as disclosed herein is efficacious for treating alopecia, without significant adverse events.

[0219] In addition, the formulation herein disclosed has an optimal stability profile allowing a storage of the final product for at least two years (upon storage at refrigerated or room temperature, as defined by Pharmaceutical Guidelines).

EXAMPLES

[0220] The formulations described herein are now further detailed with reference to the following examples. These examples are provided for the purpose of illustration only and the embodiments described herein should in no way be construed as being limited to these examples. Rather, the embodiments should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.

[0221] Example 1: Evaluation of Cortexolone-17a-propionate Solubility.

[0222] 5 g of cortexolone-17a-propionate was added to 100 ml of each of the solvents or solvent mixtures shown in Table 1, below, in an effort to produce a 5% w/v solution of cortexolone-17a-propionate. In the examples in Table 1, ethanol refers to ethanol 96°.

[0223] Table 1

[0224] As can be seen from the data in Table 1, water is an unsuitable solvent for cortexolone-17a-propionate when used alone or in a binary mixture. TRANSCUTOL ® increased solubility of the therapeutic agent in each mixture in which it was used. Ethanol was likewise found to readily solubilize cortexolone-17a-propionate, but cannot be used alone due to possible burning after topical application as well its potential for abuse.

[0225] Example 2: Evaluation of pH on Cortexolone-17a-propionate Stability.

[0226] In a suitable container, under stirring, 50 g of cortexolone-17a-propionate was dissolved in a mixture of TRANSCUTOL ® (461 g) and ethanol 96° (200 g). After complete dissolution of the cortexolone-17a-propionate, water (283 g) was added slowly. Finally, polysorbate 80 (5 g) and Alpha tocopherols (Vitamin E) (1 g) were added to the formulation. The natural pH of this formulation was measured using a standard pH electrode to be about 5.1.

[0227] The formulation was then split into three equal batches, each weighing about 300 g.

Citric acid was added to the first batch to lower the pH to about 4. Sodium citrate was added the second batch to increase the pH to about 6. The third batch, having a natural pH of about 5.1, was used as a control. The three batches were then subjected to a short term stability study at 30 °C, with the results shown in Table 2.

[0228] Table 2

[0229] The above data clearly shows that production of cortexolone-21-propionate is significantly lowered at a pH of about 4.

[0230] Example 3: Antioxidant Evaluation

[0231] To a formulation containing 5 weight percent cortexolone-17a-propionate in a mixture of TRANSCUTOL ® , propylene glycol, and ethanol (96 °) in ratio of about 1:1:1 by weight, and further containing polysorbate 80 at 0.1 weight percent, the following antioxidants were added: Alpha tocopherols (Vitamin E) 0.3 weight percent; butylated hydroxyanisole (BHA) 0.01 weight percent, or ascorbyl palmitate 0.5 weight percent. The three formulations were then studied under short term stability conditions. The results are shown in Table 3.

[0232] Table 3

[0233] Ascorbyl palmitate provided the least amount of total degradation products.

[0234] Example 4a: Anhydrous 5% w/w Solution

[0235] A 5 weight percent (w/w) solution of cortexolone-17a-propionate having the components shown in Table 4, below, was prepared by solubilizing the therapeutic agent in the mixture of solvents followed by the addition of the antioxidant (ascorbyl palmitate) and the emulsifier (polysorbate 80).

[0236] Table 4

[0237] This formulation provided the stability profile (40 °C/75% RH ) shown in Table 5.

[0238] Table 5

*Percentages of cortexolone-21-propionate and total impurities calculated as noted in Examples 2 and 3.

[0239] Example 4b: Anhydrous 5% w/v Solution

[0240] A 5% (w/v) solution of cortexolone-17a-propionate having the components shown in Table 6, below, was prepared by solubilizing the therapeutic agent in the mixture of solvents followed by the addition of the antioxidant (ascorbyl palmitate) and the emulsifier

(polysorbate 80).

[0241] Table 6

[0242] Example 5: Aqueous 5% w/w Solution

[0243] A formulation of cortexolone-17a-propionate having the components shown in

Table 7, below, was prepared by solubilizing the therapeutic agent in the mixture of solvents followed by the addition of the antioxidant and the emulsifier.

[0244] Table 7

[0245] The stability of this formulation at 40 °C/75% RH is shown in Table 8. [0246] Table 8

[0247] Based on the impurity profile, the formulation prepared in Example 4a is more stable than the aqueous formulation of Example 5.

[0248] Example 6: Clinical Evaluation

[0249] The formulation described in Example 4b was studied in a clinical trial to evaluate its pharmacokinetic profile, safety, and tolerability. According to the study protocol, 1 ml of the formulation (corresponding to 50 mg of cortexolone-17a-propionate) was applied once a day on study day 1 and then twice daily thereafter on study days 2-28 to the areas of the scalp suffering from alopecia. The formulation provided the pharmacokinetic profile shown in Tables 9 and 10 and the excretion data shown in Table 11. Statistical analyses were performed using SAS ® version 9.1.3, service pack 4 for Windows and Phoenix WinNonLin 6.3, Pharsight Corporation, USA.

[0250] Table 9 - PK parameters after single dose administration

[0251] Table 10 - PK parameters after repeated dose administration (steady state)

[0252] Table 11 - Excretion parameters at 1 st and 55 th application

[0253] Example 7: Franz Cell Diffusion Test 1

[0254] In an in vitro study, the skin penetration potential of cortexolone-17a-propionate was compared to cyproterone acetate using a standard Franz cell. According to the protocol, 300 mg of each test formulations was applied as saturated solution with a concentration ranging from 0.7 to 1 weight (w/w) to 2.54 cm 2 of exposed skin area. The receptor chamber volume ranged from 4.75 ml to 6.4 ml of phosphate buffered saline/fetal calf serum (2:1) containing penicillin/streptomycin according to standard protocol, and was maintained at a temperature of 32 °C +/- 1 °C. Stirring was maintained at 300 rpm.

[0255] The experiments were conducted for 48 h in triplicate. 100 μΙ fractions were withdrawn at 5-7 time points during the 48h test period. After withdrawing a 100 μΙ fraction for analysis, it was replaced with fresh receiver fluid in an equal volume. At the end of the experiment, the skin was taken from the Franz cells and the skin's stratum corneum was removed by tape stripping (20x) with transparent adhesive tape (Kores, Spain). [0256] The strippings were analyzed by dissolution in a mixture of propylene glycol: oleyl alcohol 9:1. The resulting mixture was analyzed for concentration of cyproterone acetate and cortexolone-17a-propionate, with the results provided in Table 12, below.

[0257] Table 12

[0258] As can be seen from the data, cortexolone-17a-propionate permeates skin about 3 times as much as cyproterone acetate and has a permeation rate that is about 10 times greater than cyproterone acetate.

[0259] Example 8: Franz Cell Diffusion Test 2

[0260] Concentration of the active agent in the receiver fluid was measured next using a system largely identical to the system in Example 7. 3 human donor skin samples were used and the permeation, expressed as Cumulative Permeated amount ^g/cm 2 ), is shown in Table 13. This experiment compared the anhydrous solution of Example 4a and the aqueous solutions of Example 5. Each contained the same amount of cortexolone-17a-propionate (5%).

[0261] Table 13

[0262] As is evident from the data in Table 13, water in the formulation can reduce cortexolone-17a-propionate's penetration into the skin.

[0263] Example 9: Phase 2 clinical study in males with androgenetic alopecia (AGA)

[0264] The formulation described in Example 4b was studied in a multicenter, randomized, double-blind, phase 2 controlled study. In the study, the formulation of Example 4b (cortexolone-17a-propionate solution 5%) was compared to vehicle solution as placebo. Both the formulation containing the active and the vehicle formulation were applied twice-daily for 26 weeks in males with androgenetic alopecia (AGA). The study was compliant with Good Clinical Practices (GCP) for Clinical Research Studies.

[0265] Objective

[0266] The primary objective of this study was to compare the safety and efficacy of topical application of cortexolone-17a-propionate 5% solution (having the composition of Example 4b) (twice a day) and the vehicle solution (twice a day) in males with AGA.

[0267] Study subjects

[0268] The subjects were 18 to 50 years of age and had mild to moderate AGA at the temple and vertex regions of the scalp, with a modified Hamilton-Norwood Scale rating of III vertex to V (lllv, IV, V), and ongoing hair loss. [0269] Treatment

[0270] Subjects received either cortexolone-17a-propionate 5% solution or vehicle control solution depending on the allocation group and applied the provided formulation to the balding areas of the scalp (vertex and temple) twice a day for 6 months.

[0271] Visits schedule

[0272] Subjects had visits at baseline (visit 2), 1 st month (visit 3), 2 nd month (visit 4), 4 th month (visit 5) and 6 th month (visit 6). Subject screening (visit 1) took place within 2 weeks of baseline visit (Visit 2). In all the study visits, all measurements for efficacy and safety endpoints were performed, with the exception of visits 2 and 3, where only local tolerability assessment (LTA) and adverse events (AEs) evaluation were performed.

[0273] Study measurements

[0274] In Table 14, the study measurements for hair loss classification, efficacy endpoints and safety endpoints are reported.

[0275] Table 14. Study measurements

Satisfaction Scale (HGSS).

1. HGA - Scalp hair growth was compared from baseline using the following 7-point scale: greatly decreased (-3), moderately decreased (-2), slightly decreased (-1), no change (0), slightly increased (1), moderately increased (2), and greatly increased (3).

2. HGI - Hair growth was compared from baseline by three questions on a health outcome questionnaire: [1] "Since the start of treatment, when 1 look at my thinning area, 1 can see...(scalp)", [2] "Since the start of treatment, my hair now covers...(scalp)", and [3] "Since the start of treatment, the appearance (thickness/quality/amount) of the thinning area on my head is..."; were scored using the following 7-point scale: much less (-3), moderately less (-2), slightly less (-1), the same amount (0), slightly more (1), moderately more (2), much more (3).

3. HGSS - Hair appearance/growth was compared from baseline by five questions: How satisfied do you feel about: [1] The overall appearance of your hair; [2] The appearance of the thinning area(s) within treatment areas on your head; [3] The amount of scalp that can be seen in the treatment areas;

[4] The amount of hair in the treatment areas; [5] The growth of hair in the treatment areas; were scored using the following 7-point scale: very dissatisfied (-3), dissatisfied (-2), somewhat dissatisfied (-1), neutral/neither satisfied nor dissatisfied (0), somewhat satisfied (1), satisfied (2), very satisfied (3).

• Investigator Global Assessment (IGA) measuring change in scalp hair growth. The evaluator used a standardized global photo (see above) of the subject's scalp taken at baseline and compared it with the clinic visit's live-assessment of the subject's scalp hair growth using a 7-point scale: greatly decreased (-3), moderately decreased (-2), slightly decreased (-1), no change (0), slightly increased (1), moderately increased (2), and greatly increased (3).

Safety Local and systemic adverse events (AEs) were assessed at each visit.

Local tolerability assessment (LTA) of erythema, scaling, pruritus, and burning/stinging were graded on discrete 5-point scale: none (0), minimal (1), mild (2), moderate (3), and severe (4) and performed at Baseline, and Months 1, 2, 4, and 6. In addition, the investigator assessed reactions known to be associated with topical application of steroids including skin atrophy, telangiectasia, folliculitis,

hypopigmentation, and hyperpigmentation graded on discrete 5-point scale: none (0), trace (1), mild (2), moderate (3), and severe (4).

[0276] In Table 15, the study endpoints, categorized as efficacy or safety endpoints, are reported.

[0277] Table 15. Study endpoints.

[0278] The Intent-To-Treat (ITT) population included all randomized subjects, that received at least one application of the study drug, and was the primary population used for safety assessment. The per-protocol (PP) population was the subset of the ITT population completing the study and without major protocol deviations, and was considered as the primary population for statistical analysis of efficacy endpoints. At the end of the study, the ITT was subdivided in the study groups as follows: 31 subjects in cortexolone-17a-propionate solution 5 % arm and 33 subjects in vehicle solution arm. At the end of the study, the PP population was subdivided in the study groups as follows: 23 subjects in cortexolone-17a-propionate solution 5 % arm and 25 subjects in vehicle solution arm. [0279] Changes from Baseline in Non-Vellus Target Area Hair Count (TAHC) at Month 6

(primary efficacy endpoint)

[0280] Non-vellus Target Area Hair Count (TAHC) was calculated using digital image analysis from standardized macro photographs collected at month 2, 4 and 6. Table 16 reports the change from baseline in non-vellus TAHC at month 6 for the PP population: the values refer to the change, from baseline, in the number of non-vellus hairs in a 1 cm 2 area of the scalp after 6 months of treatment.

[0281] Table 16. Change from Baseline in Non-Vellus TAHC at Month 6.

[0282] As is demonstrated in the data above, cortexolone-17a-propionate solution (5%) had larger changes from baseline in non-vellus TAHC compared to vehicle at month 6. Cortexolone-17a-propionate solution 5% (according to Example 4b) had a larger mean change from baseline in non-vellus TAHC (12.7) with respect to vehicle, which had a mean change from Baseline in non-vellus TAHC of 2.9.

[0283] Hair Growth Assessment at month 6

[0284] Subjects used the baseline standardized global photo of their scalp and compared it, side by side, with a "real time" standardized global photo at month 6 to provide a comparative assessment for HGA. Subjects evaluated hair growth using a 7-point scale. Figure 1 depicts HGA frequency distribution for the two treatment groups at month 6. In the figure, negative HGA scores (-3, -2 and -1) have been grouped into a global HGA score named "unfavorable"; score 0 is named "no change," and positive scores (+1, +2 and +3) have been grouped into a global HGA score named "favorable". HGA scores for the PP population at month 6 are reported in Table 17.

[0285] Table 17. HGA scores for PP population at month 6.

[0286] The proportion of subjects who rated scalp hair growth as favorable was directionally larger in cortexolone-17a-propionate solution 5% (39%) compared to vehicle (16%). Of those subjects with favorable hair growth, the proportion of subjects who rated hair growth as greatly increased (+3), moderately increased (+2), and slightly increased (+1) was 4%, 13%, and 22%, respectively for cortexolone-17a-propionate solution 5%, and 0%, 8%, and 8% respectively, for vehicle. The weighted average of HGA at month 6 for both the treatment groups (PP populations) was calculated: cortexolone-17a-propionate solution 5% had a higher HGA weighted average at month 6 (0.30) compared to placebo (-0.24).

[0287] This data demonstrates that subjects in the cortexolone-17a-propionate solution

5% group had a larger magnitude of improvement compared to vehicle solution.

[0288] Hair Growth Index (HGI) at Month 6

[0289] Subjects used the baseline standardized global photo of their scalp and compared it, side by side, with a "real time" standardized global photo at month 6 to provide a comparative assessment for HGI. Hair growth was compared from baseline by three questions on a health outcome questionnaire. The proportion of subjects who rated scalp hair growth as favorable was larger in cortexolone-17a-propionate solution 5% (Ql: 39%, Q2: 35%, and Q3: 43%) compared to vehicle (Ql: 16%, Q2: 12%, and Q3: 20%).

[0290] Hair Growth Satisfaction Scale (HGSS) at Month 6

[0291] Subjects used the baseline standardized global photo of their scalp and compared it, side by side, with a "real time" standardized global photo at month 6 to provide a comparative assessment for HGSS. Hair appearance/growth was compared from baseline using five questions. The proportion of subjects who were satisfied with scalp hair growth was larger in cortexolone-17a-propionate solution 5% compared to vehicle solution. For questions #1-5 (Q1-Q5), the proportion of subjects who rated scalp hair growth as favorable (scores of +1, +2 and +3) was higher for cortexolone-17a-propionate solution 5% (Ql: 38%, Q2-Q5: 30%) compared to vehicle solution (Q1/Q3: 8%, Q2/Q4/Q5: 20%). For cortexolone-17a-propionate 5% solution, no subjects were 'very dissatisfied' (score of -3) for any of the HGSS questions at Month 6, whereas, for vehicle, there were some subjects very dissatisfied in all the HGSS questions at month 6.

[0292] Investigator's Global Assessment (IGA) at Month 6

[0293] At month 6, investigators used a standardized global photo of the subject's scalp taken at baseline and compared it with a live-assessment of the subject's scalp hair growth using a 7-point scale. Figure 2 depicts the frequency distribution of IGA for two treatment groups (one for cortexolone-17a-propionate solution and one for vehicle solution) at month 6. In Figure 2, negative IGA scores (-3, -2 and -1) are grouped into a global IGA score named "unfavorable"; score 0 is named "no change" and positive scores (+1, +2 and +3) are grouped into a global IGA score named "favorable". IGA scores for the PP population at month 6 are reported in Table 18. [0294] Table 18. IGA scores for PP population at month 6.

[0295] The proportion of subjects who had a favorable IGA score (scores of +1, +2 and +3) was higher in the cortexolone-17a-propionate solution 5% group (43%) than in the vehicle group (36%), and the proportion of subjects who had an unfavorable IGA score (scores of -1, -2 and -3) was lower for cortexolone-17a-propionate (9%) compared to the vehicle (20%). The weighted average of IGA at month 6 for both the treatment groups (PP populations) was calculated: cortexolone-17a-propionate solution 5% had a higher IGA weighted average at month 6 (0.43) compared to placebo (0.12).

[0296] This data demonstrates that subjects in the cortexolone-17a-propionate solution

5% group had a larger magnitude of improvement compared to the vehicle solution.

[0297] Local Tolerability and Adverse Events Assessment

[0298] Local Tolerability Assessment analysis was performed on all subjects that applied the test article at least once (ITT population). The incidence of each sign in the LTA was low throughout the study and generally similar among the treatment groups. Of the local tolerability signs reported, most signs were minimal to mild in severity, with (in descending order of incidence) pruritus, scaling, erythema, folliculitis, hyperpigmentation, and hypopigmentation present after treatment. [0299] The incidence of Adverse Events (AEs) was similar among the treatment groups, with most events typically mild in severity and not related to the test article. In the cortexolone-17a-propionate solution 5% group, AEs occurred in 18/31 subjects (58.1%), while, in the vehicle group, AEs occurred in 17/33 subjects (51.5%). The majority of AEs were mild. 4 AEs in vehicle group were rated as severe, with only one of them (headache) possibly related to the test article. No severe AE in cortexolone-17a-propionate group occurred.

[0300] Importantly, no adverse events which could be due to a systemic antiandrogenic effect (e.g. decreased libido, erectile dysfunction, and/or ejaculation disorder) occurred in any of the patients treated with cortexolone-17a-propionate solution 5% for 6 months.

[0301] Conclusions

[0302] The study was a Phase II POC study in a limited number of subjects, and was not powered to show statistically significant differences among study groups; nevertheless, the study demonstrated a superior improvement in hair growth of cortexolone 17a-propionate solution 5% compared to vehicle: at month 6, in the PP population, cortexolone 17a- propionate (12.7) had superior change from Baseline in non-vellus Target Area Hair Count (TAHC) compared to vehicle (2.9). The result of subject Hair Growth Assessment questionnaire (the other primary efficacy endpoint of the study) was consistent with the quantitative TAHC measurements, and the proportion of subjects who rated scalp hair growth as favorable was larger in cortexolone 17a-propionate (39%) compared to vehicle (16%); of those subjects who rated scalp hair growth as favorable, cortexolone 17a-propionate tended to have a larger magnitude of improvement compared to vehicle. The results of the secondary endpoints were generally consistent with that of the primary endpoints.

[0303] Example 10: 15% (w/v) solution

[0304] A 15% (w/v) solution of cortexolone-17a-propionate having the components shown in Table 19, below, was prepared by solubilizing the therapeutic agent in the mixture of solvents followed by the addition of the antioxidant (ascorbyl palmitate) and the emulsifier (polysorbate 80). [0305] Table 19

[0306] Example 11: Comparison of the effects of the formulation of Example 4b and the commercially available finasteride 1 mg tablet (PROPECIA ® )

[0307] Based on the results of finasteride 1 mg tablets (PROPECIA ® ) described in the two

Phase II I clinical studies published online by the United States Food and Drug Administration (the FDA), it can be concluded that the mean change from baseline in non-vellus TAHC at month 6 for cortexolone-17a-propionate solution 5% (according to Example 4b), as reported in Example 9, is very similar to the mean change from baseline in non-vellus TAHC at month 6 observed in 2 phase II I clinical trials on finasteride 1 mg tablets (tradename Propecia ® ) per os once daily. Table 20 below reports the change from baseline in TAHC at month 6 in the two finasteride studies (data of active finasteride groups) as described in the NDA for PROPECIA ® published online by the FDA:

[0308] Table 20. Mean change from baseline in TAHC at month 6 in the 2 finasteride phase III clinical studies (data of active finasteride groups are reported) - original data.

[0309] In the two finasteride studies, the target area was a 1-inch diameter circle, which corresponds to 5.1 cm 2 . I n order to compare the changes from baseline in TAHC at month 6 obtained in the finasteride studies and the change from baseline in TAHC at month 6 obtained with cortexolone-17a-propionate in the study of the present Example 9 (where the target area was 1 cm 2 ), the original finasteride data have been recalculated to take into account the differences between the total surfaces of the target areas (i.e., the values were divided by 5.1) as shown in Table 21.

[0310] Table 21. Mean change from baseline in TAHC at month 6 in the 2 finasteride phase III clinical studies (data of active finasteride groups are reported), recalculated over a target area of 1 cm 2 .

[0311] Accordingly, the changes from baseline in TAHC at month 6 for finasteride were as follows: 13.6 in study 087, 11.5 in study 089 and 12.2 for the two studies combined. It is apparent that these values are almost identical to the change from baseline in TAHC at month 6, in the PP population, for cortexolone-17a-propionate solution 5% prepared according to Example 4b (12.7). As discussed above, at month 6, the efficacy, measured as hair growth, of finasteride 1 mg tablets given per os once daily and of cortexolone-17a-propionate solution 5% topically administered twice daily on the scalp of the patients are comparable.

[0312] The phraseology or terminology herein is for the purpose of description and not of limitation. As such, the terminology and/or phraseology of the present specification should be interpreted by the skilled artisan in light of the teachings and guidance herein. [0313] The breadth and scope of the present invention should not be limited by any of the above-described exemplary embodiments, but should be defined only in accordance with the following claims and their equivalents.

[0314] All patents, patent applications, and other references noted or referenced in this application are hereby incorporated by reference in their entirety.