Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FLUORO-CHLORO BISPHENOL ETHER COMPOUNDS AND METHODS FOR THEIR USE
Document Type and Number:
WIPO Patent Application WO/2016/058080
Kind Code:
A1
Abstract:
Compounds having a structure of Formula I: or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are as defined herein, and wherein at least one of R1, R2, R3, R4 or R5 is F, are provided. Uses of such compounds for treatment of various indications, inciuding prostate cancer as well as methods of treatment involving such compounds are also provided.

Inventors:
SADAR MARIANNE DOROTHY (CA)
ANDERSEN RAYMOND JOHN (CA)
FERNANDEZ JAVIER GARCIA (ES)
Application Number:
PCT/CA2015/000533
Publication Date:
April 21, 2016
Filing Date:
October 14, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRITISH COLUMBIA CANCER AGENCY (CA)
UNIV BRITISH COLUMBIA UNIVERSITY INDUSTRY LIAISON OFFICE (CA)
SADAR MARIANNE DOROTHY (CA)
ANDERSEN RAYMOND JOHN (CA)
FERNANDEZ JAVIER GARCIA (ES)
International Classes:
C07C43/23; A61K31/09; A61P5/28; A61P35/00; C07C43/225
Domestic Patent References:
WO2013028791A12013-02-28
WO2013028572A12013-02-28
WO2010000066A12010-01-07
WO2011082487A12011-07-14
Attorney, Agent or Firm:
DEETH WILLIAMS WALL LLP (Suite 400Toronto, Ontario M5H 3S5, CA)
Download PDF:
Claims:
CLAIMS

A compound having a structure of Formula I:

or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:

R1 is F,OHorOY;

R2, R3 R5, R6, R7 and R8 are each independently H or F;

R4isH,F,OHorOY;

R9 and R10 are each independently F or C1-C5 alkyl; and Y is a moiety from Table I;

wherein at least one of R\ R2, R3, R4 or R5 is F.

3. The compound of claims 1 or 2, wherein R1 is F.

4. The compound of claims 1 or 2, wherein R! is OH.

5. The compound of claims 1 or 2, wherein at least one of R1 or R4 is OY.

6. The compound of any one of claims 1-5, wherein each of R6 and R7 is H.

7. The compound of any one of claims 1-6, wherein R8 is H.

8. The compound of any one of claims 1-7, wherein R8 is F.

9. The compound of any one of claims 1-8, wherein each of R4 and R5 is H.

10. The compound of any one of claims 1 -8, wherein R4 is F.

11. The compound of any one of claims 1-8, wherein R4 is OH.

12. The compound of any one of claims 1-11, wherein at least two of R1, R2, R3, R4 and R5 are F.

13. The compound of any one of claims 1-11, wherein at least three of R1, R2, R3, R4 and R5 are F.

14. The compound of any one of claims 1-8, wherein at least four of R1, R2, R3, R4 and R5 are F.

15. The compound of any one of claims 1-8, wherein each of R1, R2, R3, R4 and R5 is F.

16. The compound of any one of claims 1-15, wherein at least one of R9 or R10 is

F.

17. The compound of any one of claims 1-15, wherein each of R9 and R10 is F.

18. The compound of any one of claims 1-15, wherein at least one of R9 or R'° is methyl.

19. The compound of any one of claims 1-15, wherein each of R9 and R10 is methyl.

20. The compound of any one of claims 1-19, wherein Y is or

21. The compound of claim 1, wherein the compound has one of the following structur

compound of claim 1, wherein the compound has the following structure

(1):

or a pharmaceutically acceptable salt or stereoisomer thereof.

The compound of claim 1 , wherein the compound has the following structure

(l a):

harmaceutically acceptable salt thereof.

The compound of claim 1 , wherein the compound has the following structure

(l b):

or a pharmaceutically acceptable salt thereof.

25. Use of the compound of any one of claims 1 to 24, for modulating androgen receptor (AR) activity.

26. The use of claim 25, wherein modulating androgen receptor (AR) activity is in a mammalian cell.

27. The use of any one of claims 25 or 26, wherein modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration.

28. The use of claim 27, wherein the indication is prostate cancer.

29. The use of claim 28, wherein the prostate cancer is castration resistant prostate cancer.

30. The use of claim 28, wherein the prostate cancer is androgen-dependent prostate cancer.

31. A method of modulating androgen receptor (AR) activity, the method comprising administering a compound, or pharmaceutically acceptable salt thereof, of any one of claims 1 to 24 to a subject in need thereof.

32. The method of claim 31, wherein modulating androgen receptor (AR) activity is for the treatment of one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration.

33. The method of claim 32, wherein the indication is prostate cancer.

34. The method of claim 33, wherein the prostate cancer is castration resistant prostate cancer.

35. The method of claim 33, wherein the prostate cancer is androgen-dependent prostate cancer.

36. A pharmaceutical composition comprising a compound of any one of claims 1 to 24 and a pharmaceutically acceptable carrier.

37. A pharmaceutical composition comprising a compound of any one of claims 1 to 24, an additional therapeutic agent and a pharmaceutically acceptable carrier.

38. The pharmaceutical composition of claim 37, wherein the additional therapeutic agent is for treating prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy or age-related macular degeneration.

39. The pharmaceutical composition of claim 38, wherein the additional therapeutic agent is ODM-201 , MDV3100 , TAK 700, TOK 001 ; ARN-509; abiraterone acetate, bicalutamide, nilutamide, flutamide, cyproterone acetate, docetaxel, Bevacizumab (Avastin), OSU-HDAC42, VITAXIN, sunitumib, ZD-4054, Cabazitaxel (XRP-6258), MDX- 010 (Ipilimumab), OGX 427, OGX 01 1 , finasteride, dutasteride, turosteride, bexlosteride, izonsteride, FCE 28260, SKF105,1 1 1 or a related compound thereof.

40. Use of the pharmaceutical composition of any one of claims 36 to 39 for modulating androgen receptor (AR) activity.

41. The use of claim 40, wherein modulating androgen receptor (AR) activity is in a mammalian cell.

42. The use of any one of claims 40 or 41 , wherein modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration.

43. The use of claim 42, wherein the indication is prostate cancer.

44. The use of claim 43, wherein the prostate cancer is castration resistant prostate cancer.

45. The use of claim 43, wherein the prostate cancer is androgen-dependent prostate cancer.

46. A method of modulating androgen receptor (AR) activity, the method comprising administering the pharmaceutical composition of any one of claims 36 to 39 to a subject in need thereof.

47. The method of claim 46, wherein modulating androgen receptor (AR) activity is for the treatment of one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration.

48. The method of claim 47, wherein the indication is prostate cancer.

49. The method of claim 48, wherein the prostate cancer is castration resistant prostate cancer.

50. The method of claim 48, wherein the prostate cancer is androgen-dependent prostate cancer.

Description:
FLUORO-CHLORO BISPHENOL ETHER COMPOUNDS AND METHODS FOR THEIR

USE

CROSS REFERENCE TO RELATED APPLICATIONS

This Application claims the benefit of U.S. Provisional Application No. 62/063,651, filed

October 14, 2014, which is hereby incorporated by reference in its entirety for all purposes.

STATEMENT OF GOVERNMENT INTEREST

This invention was made in part with government support under Grant No. 2R01 CA105304 awarded by the National Cancer Institute. The United States Government has certain rights in this invention.

BACKGROUND

This invention relates to therapeutics, their uses and methods for the treatment of various indications, including various cancers. In particular the invention relates to therapies and methods of treatment for cancers such as prostate cancer, including all stages and androgen dependent, androgen-sensitive and castration-resistant cancers (also referred to as hormone refractory, androgen-independent, androgen deprivation resistant, androgen ablation resistant, androgen depletion-independent, castration-recurrent, and anti-androgen-recurrent).

Androgens mediate their effects through the androgen receptor (AR). Androgens play a role in a wide range of developmental and physiological responses and are involved in male sexual differentiation, maintenance of spermatogenesis, and male gonadotropin regulation (R. K. Ross, G. A. Coetzee, C. L. Pearce, J. K. Reichardt, P. Bretsky, L. N. Kolonel, B. E. Henderson, E. Lander, D. Altshuler & G. Daley, Eur Urol 35, 355-361 (1999); A. A. Thomson, Reproduction 121, 187-195 (2001); N. Tanji, K. Aoki & M. Yokoyama, Arch Androl 47, 1-7 (2001)). Several lines of evidence show that androgens are associated with the development of prostate carcinogenesis. Firstly, androgens induce prostatic carcinogenesis in rodent models (R. L. Noble, Cancer Res 37, 1929-1933 (1977); R. L. Noble, Oncology 34, 138-141 (1977)) and men receiving androgens in the form of anabolic steroids have a higher incidence of prostate cancer (J. T. Roberts & D. M. Essenhigh, Lancet 2, 742 (1986); J. A. Jackson, J. Waxman & A. M. Spiekerman, Arch Intern Med 149, 2365-2366 (1989); P. D. Guinan, W. Sadoughi, H. Alsheik, R. J. Ablin, D. Alrenga & I. M. Bush, Am J Surg 131, 599-600 (1976)). Secondly, prostate cancer does not develop if humans or dogs are castrated before puberty (J. D. Wilson & C. Roehrborn, J Clin Endocrinol Metab 84, 4324-4331 (1999); G. Wilding, Cancer Surv 14, 113-130 (1992)). Castration of adult males causes involution of the prostate and apoptosis of prostatic epithelium while eliciting no effect on other male external genitalia (E. M. Bruckheimer & N. Kyprianou, Cell Tissue Res 301, 153-162 (2000); J. T. Isaacs, Prostate 5, 545-557 (1984)). This dependency on androgens provides the underlying rationale for treating prostate cancer with chemical or surgical castration (androgen ablation).

Androgens also play a role in female cancers. One example is ovarian cancer where elevated levels of androgens are associated with an increased risk of developing ovarian cancer (K. J. Helzlsouer, A. J. Alberg, G. B. Gordon, C. Longcope, T. L. Bush, S. C. Hoffman & G. W. Comstock, JAMA 274, 1926-1930 (1995); R. J. Edmondson, J. . Monaghan & B. R. Davies, Br J Cancer 86, 879-885 (2002)). The AR has been detected in a majority of ovarian cancers (H. A. Risch, J Natl Cancer Inst 90, 1774-1786 (1998); B. R. Rao & B. J. Slotman, Endocr Rev 12, 14-26 (1991); G. M. Clinton & W. Hua, Crit Rev Oncol Hematol 25, 1-9 (1997)), whereas estrogen receptor-alpha (ERa) and the progesterone receptor are detected in less than 50% of ovarian tumors.

The only effective treatment available for advanced prostate cancer is the withdrawal of androgens which are essential for the survival of prostate epithelial cells. Androgen ablation therapy causes a temporary reduction in tumor burden concomitant with a decrease in serum prostate-specific antigen (PSA). Unfortunately prostate cancer can eventually grow again in the absence of testicular androgens (castration-resistant disease) (Huber et al 1987 Scand J. Urol Nephrol. 104, 33-39). Castration-resistant prostate cancer is biochemically characterized before the onset of symptoms by a rising titer of serum PSA (Miller et al 1992 J. Urol. 147, 956-961). Once the disease becomes castration-resistant most patients succumb to their disease within two years.

The AR has distinct functional domains that include the carboxy-terminal ligand-binding domain (LBD), a DNA-binding domain (DBD) comprising two zinc finger motifs, and an N- terminus domain (NTD) that contains one or more transcriptional activation domains. Binding of androgen (ligand) to the LBD of the AR results in its activation such that the receptor can effectively bind to its specific DNA consensus site, termed the androgen response element (ARE), on the promoter and enhancer regions of "normally" androgen regulated genes, such as PSA, to initiate transcription. The AR can be activated in the absence of androgen by stimulation of the cAMP-dependent protein kinase (PKA) pathway, with interleukin-6 (IL-6) and by various growth factors (Culig et al 1994 Cancer Res. 54, 5474-5478; Nazareth et al 1996 J. Biol. Chem. 271, 19900-19907; Sadar 1999 J. Biol. Chem. 274, 7777-7783; Ueda et al 2002 A J. Biol. Chem. 277, 7076-7085; and Ueda et al 2002 B J. Biol. Chem. 277, 38087-38094). The mechanism of ligand-independent transformation of the AR has been shown to involve: 1) increased nuclear AR protein suggesting nuclear translocation; 2) increased AR/ARE complex formation; and 3) the AR-NTD (Sadar 1999 J. Biol. Chem. 274, 7777-7783; Ueda et al 2002 A J. Biol. Chem. 277, 7076-7085; and Ueda et al 2002 B J. Biol. Chem. 277, 38087-38094). The AR may be activated in the absence of testicular androgens by alternative signal transduction pathways in castration-resistant disease, which is consistent with the finding that nuclear AR protein is present in secondary prostate cancer tumors (Kim et al 2002 Am. J. Pathol. 160, 219-226; and van der Kwast et al 1991 Inter. J. Cancer 48, 189-193).

Available inhibitors of the AR include nonsteroidal antiandrogens such as bicalutamide (Casodex™), nilutamide, flutamide, investigational drugs MDV3100 and ARN-509, and the steroidal antiandrogen, cyproterone acetate. These antiandrogens target the LBD of the AR and predominantly fail presumably due to poor affinity and mutations that lead to activation of the AR by these same antiandrogens (Taplin, M.E., Bubley, G.1, Kom Y.J., Small E.J., Uptonm M., Rajeshkumarm B., Balkm S.P., Cancer Res., 59, 2511-2515 (1999)). These antiandrogens would also have no effect on the recently discovered AR splice variants that lack the ligand-binding domain (LBD) to result in a constitutively active receptor which promotes progression of androgen-independent prostate cancer (Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ., Cancer Res 68, 5469-77, 2008; Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, Chen H, Kong X, Melamed J, Tepper CG, Kung HJ, Brodie AM, Edwards J, Qiu Y., Cancer Res. 69, 2305-13, 2009; Hu et al 2009 Cancer Res. 69, 16-22; Sun et al 2010 J Clin Invest. 2010 120, 2715-30). Conventional therapy has concentrated on androgen-dependent activation of the AR through its C-terminal domain. Recent studies developing antagonists to the AR have concentrated on the C-terminus and specifically: 1) the allosteric pocket and AF-2 activity (Estebanez-Perpina et al 2007, PNAS 104, 16074-16079); 2) in silico "drug repurposing" procedure for identification of nonsteroidal antagonists (Bisson et al 2007, PNAS 104, 11927- 11932); and coactivator or corepressor interactions (Chang et al 2005, Mol Endocrinology 19, 2478-2490; Hur et al 2004, PLoS Biol 2, E274; Estebanez-Perpifia et al 2005, JBC 280, 8060-8068; He et al 2004, Mol Cell 16, 425-438).

The AR-NTD is also a target for drug development (e.g., WO 2000/001813), since the NTD contains Activation-Function-1 (AF-1) which is the essential region required for AR transcriptional activity (Jenster et al 1991. Mol Endocrinol. 5, 1396-404). The AR-NTD importantly plays a role in activation of the AR in the absence of androgens (Sadar, M.D. 1999 J. Biol. Chem. 274, 7777-7783; Sadar MD et al 1999 Endocr Relat Cancer. 6, 487-502; Ueda et al 2002 J. Biol. Chem. 277, 7076-7085; Ueda 2002 J. Biol. Chem. 277, 38087-38094; Blaszczyk et al 2004 Clin Cancer Res. 10, 1860-9; Dehm et al 2006 J Biol Chem. 28, 27882-93; Gregory et al 2004 J Biol Chem. 279, 7119-30). The AR-NTD is important in hormonal progression of prostate cancer as shown by application of decoy molecules (Quayle et al 2007, Proc Natl Acad Sci USA. , 1331-1336).

While the crystal structure has been resolved for the AR C-terminus LBD, this has not been the case for the NTD due to its high flexibility and intrinsic disorder in solution (Reid et al 2002 J. Biol. Chem. 277, 20079-20086) thereby hampering virtual docking drug discovery approaches.

Recent advances in the development of compounds that modulate AR include the bis- phenol compounds disclosed in published PCT WO 2010/000066 to the British Columbia Cancer Agency Branch and The University of British Columbia. While such compounds appear promising, there remains a need in the art for additional and/or improved compounds that modulate the AR, and which provide treatment for conditions that benefit from such modulation. BRIEF SUMMARY

In one embodiment, the compounds described herein may be used for in vivo or in vitro research uses (e.g., non-clinical) to investigate the mechanisms of orphan and nuclear receptors (including steroid receptors such as the androgen receptor). Furthermore, in other embodiments, these compounds may be used individually or as part of a kit for in vivo or in vitro research to investigate signal transduction pathways and/or the activation of orphan and nuclear receptors using recombinant proteins, cells maintained in culture, and/or animal models.

Certain embodiments of the present invention are based in part on the surprising discovery that the compounds described herein may be used to modulate AR activity either in vivo or in vitro for both research and therapeutic uses. In particular, experiments conducted in support of the present invention show that, in certain embodiments, the presence of a chlorine moiety, at least one fluorine moiety and no hydroxyl substituent alpha to the chloro imparts unexpectedly improved activity and drug-like properties (e.g., favorable half-life and/or metabolism) compared to other known AR modulators. In certain embodiments, the compounds may be used in an effective amount so that androgen receptor activity may be modulated. In certain embodiments, the AR may be mammalian, for example, human. In particular, in certain embodiments, the compounds may be used to inhibit the AR. In certain embodiments, the compounds' modulatory activity may be used in either an in vivo or an in vitro model for the study of at least one of the following indications: prostate cancer, breast cancer, ovarian cancer, salivary gland carcinoma, endometrial cancer, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty (testoxicosis), spinal and bulbar muscular atrophy (SBMA, Kennedy's disease), and age-related macular degeneration. Furthermore, in certain embodiments, the compounds modulatory activity may be used for the treatment of at least one of the following indications: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. In certain embodiments, the indication for treatment can be prostate cancer. In certain embodiments, the prostate cancer can be castration-resistant prostate cancer. In certain embodiments, the prostate cancer may be androgen-dependent prostate cancer. In other embodiments, the indication is Kennedy's disease. In accordance with one embodiment, there is provided a compound having a structure of Formula I:

or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R l , R 2 , R 3 , R 4 , R 5 , R s , R 7 , R 8 , R 9 and R 10 are as defined herein, and wherein at least one of R 1 , R 2 , R 3 , R 4 or R 5 is F.

In other embodiments, the present disclosure provides the use of a compound of Formula I, for modulating androgen receptor (AR) activity. In certain embodiments, methods for modulating AR, as well as pharmaceutical compositions comprising a compound of Formula I and a pharmaceutically acceptable excipient are also provided.

In addition, in certain embodiments, the present disclosure provides combination therapy treatments for any of the disease states disclosed herein, for example, prostate cancer. In certain embodiments, the disclosed therapies include use of a pharmaceutical composition comprising a compound of Formula I, an additional therapeutic agent and a pharmaceutically acceptable excipient. In certain embodiments, methods and compositions related to combination therapy treatments are also provided.

These and other aspects of the invention will be apparent upon reference to the following detailed description. To this end, various documents may be cited to describe in more detail certain background information, procedures, compounds and/or compositions. Each document so cited is hereby incorporated by reference in its entirety.

DETAILED DESCRIPTION

I. Definitions

In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the invention may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments. Unless the context requires otherwise, throughout the specification and claims which follow, the word "comprise" and variations thereof, such as, "comprises" and "comprising" are to be construed in an open, inclusive sense, that is, as "including, but not limited to." Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed invention.

Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments. Also, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the content clearly dictates otherwise.

The terms below, as used herein, have the following meanings, unless indicated otherwise:

"Alkyl" refers to a straight or branched hydrocarbon chain radical which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms, and which is attached to the rest of the molecule by a single bond. Alkyls comprising any number of carbon atoms from 1 to 12 are included. An alkyl comprising up to 5 carbon atoms is a Cj-Cs alkyl. A Cj-Cs alkyl includes Cs alkyls, C alkyls, C3 alkyls, C2 alkyls and Ci alkyl (i.e., methyl) and includes, for example, and without limitation, saturated C1-C5 alkyl, C2-C5 alkenyl and C2-C5 alkynyl. Non-limiting examples of saturated C1-C5 alkyl include methyl, ethyl, n-propyl, i-propyl, sec-propyl, n-butyl, i-butyl, sec-butyl, t-butyl and n-pentyl. Non-limiting examples of C2-C5 alkenyl include vinyl, allyl, isopropenyl, l-propene-2-yl, 1- butene-l-yl, l-butene-2-yl, l-butene-3-yl, 2-butene-l-yl, 2-butene-2-yl, penteneyl and the like. Non-limiting examples of C2-C5 alkynyl include ethynyl, propynyl, butynyl, pentynyland the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted with one or more fluorine atoms (i.e., a hydrogen atom in the alkyl group may be replaced with fluorine).

"Hydroxy" or "hydroxyl" refers to the -OH radical.

"Fluoro" and "chloro" refer to fluorine (F) and chlorine (CI) substituents, respectively, and also include radioisotopes of the same.

As used herein, the symbol " " (hereinafter may be referred to as "a point of attachment bond") denotes a bond that is a point of attachment between two chemical entities, one of which is depicted as being attached to the point of attachment bond and the other of which is not depicted as being attached to the point of attachment bond. For example, " " indicates that the chemical entity "XY" is bonded to another chemical entity via the point of attachment bond. Furthermore, the specific point of attachment to the non-depicted chemical entity may be specified by inference. For example, the compound CH3-R 3 , wherein R 3 is H or

" infers that when R 3 is "XY", the point of attachment bond is the same bond as the bond by which R 3 is depicted as being bonded to CH3.

II. Compounds and Methods

As noted above, the present disclosure provides a compound having a structure of

Formula I:

I

a pharmaceutically acceptable salt or stereoisomer thereof, wherein:

R 1 is F, OH or OY;

R 2 , R 3 R 5 , R 6 , R 7 and R 8 are each independently H or F; R 4 is H, F, OH or OY; R 9 and R 10 are each independently F or C1-C5 alkyl; and

Y is a moiety from Table I;

wherein at least one of R 1 , R 2 , R 3 , R 4 or R 5 is F.

In another embodiment, the compound has one of the following structures (la), (lb), (Ic) or Id):

In some embodiments of any of the foregoing embodiments, R 1 is F, and in other embodiments R 1 is OH. In yet other embodiments, at least one of R 1 or R 4 is OY.

In some other embodiments of any of the foregoing embodiments, each of R^ and R 7 is H. In some other embodiments of any of the foregoing embodiments, R 8 is H, and in some other embodiments R 8 is F. In certain embodiments, R 8 is H and R 4 is OH. In certain embodiments, R 1 is OH, R 8 is H and R 4 is OH.

In still other embodiments of any of the foregoing embodiments, each of R 4 and R 5 is H. In other embodiments, R 4 is F, and in other embodiments R 4 is OH. In still other embodiments, at least two of R 1 , R 2 , R 3 , R 4 and R 5 are F. In some embodiments, at least three of R 1 , R 2 , R 3 , R 4 and R 5 are F. In yet other embodiments, at least four of R 1 , R 2 , R 3 , R 4 and R 5 are F, and in other embodiments each of R 1 , R 2 , R 3 , R 4 and R 5 is F. In some embodiments of any of the foregoing embodiments, at least one of R 9 or R 10 is F, for example in some embodiments each of R 9 and R 10 is F. In some other embodiments, at least one of R 9 or R 10 is methyl, for example in some embodiments each of R 9 and R 10 is methyl. in embodiments of any of the foregoing embodiments, Y is or

In still other embodiments, the compound of Formula (I) has one of the following structur

a pharmaceutically acceptable salt or stereoisomer thereof.

In some embodiments, the com ound of formula I has the following structure (1):

(1)

a pharmaceutically acceptable salt or stereoisomer thereof.

In other embodiments, the compound of formula 1 has the following structure (la):

(la)

or a pharmaceutically acceptable salt thereof.

In still other embodime the compound of formula 1 has the following structure (lb):

(lb)

or a pharmaceutically acceptable salt thereof.

Each R 9 may independently be C1-C5 alkyl. Each R 9 may independently be d-C4 alkyl.

Each R 9 may independently be C1-C3 alkyl. Each R 9 may independently be C1-C2 alkyl. Each R 9 may independently be methyl. Each R 9 may independently be C2 alkyl. Each R 9 may independently be C3 alkyl. Each R 9 may independently be C 4 alkyl. Each R 9 may independently be C5 alkyl. Each R 9 may independently be F. Each R 10 may independently be C1-C5 alkyl.

Each R 10 may independently be C1-C4 alkyl. Each R 10 may independently be C1-C3 alkyl. Each

R 10 may independently be Ci-C 2 alkyl. Each R 10 may independently be methyl. Each R 10 may independently be C2 alkyl. Each R 10 may independently be C3 alkyl. Each R 10 may independently be C 4 alkyl. Each R 10 may independently be C5 alkyl. Each R 10 may independently be F.

In another embodiment, the present disclosure provides the use of any one of the foregoing compounds of Formula (I) for modulating androgen receptor (AR) activity. For example in some embodiments, modulating androgen receptor (AR) activity is in a mammalian cell.

In other embodiments, modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy (i.e., Kennedy's disease), and age-related macular degeneration. For example in some embodiments, the indication is prostate cancer. In other embodiments, the prostate cancer is castration resistant prostate cancer. While in other embodiments, the prostate cancer is androgen-dependent prostate cancer.

In other embodiments, the present disclosure provides a method of modulating androgen receptor (AR) activity, the method comprising administering any one of the foregoing compounds of Formula (I), or pharmaceutically acceptable salt thereof to a subject in need thereof.

In other further embodiments of the foregoing method, modulating androgen receptor (AR) activity is for the treatment of one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. For example in some embodiments, the prostate cancer is castration resistant prostate cancer. In other embodiments, the prostate cancer is androgen-dependent prostate cancer.

In some other embodiments, the present disclosure provides a pharmaceutical composition comprising any one of the foregoing compounds of Formula (Γ) and a pharmaceutically acceptable carrier.

In yet another embodiment, the present disclosure provides a pharmaceutical composition comprising any one of the foregoing compounds of Formula (I), an additional therapeutic agent and a pharmaceutically acceptable carrier. For example, in some embodiments, the additional therapeutic agent is for treating prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy or age-related macular degeneration.

In other embodiments, the additional therapeutic agent is ODM-201, MDV3100 , Orteronel (TAK-700), TOK 001; ARN-509; abiraterone acetate, bicalutamide, nilutamide, flutamide, cyproterone acetate, docetaxel, Bevacizumab (Avastin), OSU-HDAC42, VITAXIN, sunitumib, ZD-4054, Cabazitaxel (XRP-6258), MDX-010 (Ipihmumab), OGX 427, OGX Oi l, finasteride, dutasteride, turosteride, bexlosteride, izonsteride, FCE 28260, SKF 105,111 or a related compound thereof.

In another embodiment, the present disclosure provides the use of any one of the foregoing pharmaceutical compositions for modulating androgen receptor (AR) activity. For example in some embodiments, modulating androgen receptor (AR) activity is in a mammalian cell.

In other embodiments, modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. For example in some embodiments, the indication is prostate cancer. For example, in some embodiments, the prostate cancer is castration resistant prostate cancer, and in other embodiments the prostate cancer is androgen-dependent prostate cancer.

In yet another embodiment, the present disclosure provides a method of modulating androgen receptor (AR) activity, the method comprising admirdstering any one of the foregoing pharmaceutical compositions to a subject in need thereof. For example in some embodiments, modulating androgen receptor (AR) activity is for the treatment of one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. In still other embodiments, the indication is prostate cancer. For example, in some embodiments, the prostate cancer is castration resistant prostate cancer, while in other embodiments, the prostate cancer is androgen-dependent prostate cancer.

The compounds described herein are meant to include all racemic mixtures and all individual enantiomers or combinations thereof, whether or not they are specifically depicted herein. Alternatively, one or more of the OH groups on the above compounds may be substituted to replace the H with a moiety selected from Table 1 (z.e., to form a OY moiety). In yet other embodiments, the present disclosure provide the use of any of the compounds disclosed herein for modulating androgen receptor (AR) activity. For example, in certain embodiments modulating androgen receptor (AR) activity is in a mammalian cell.

In other examples, modulating androgen receptor (AR) activity is for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. For example, in certain embodiments the indication is prostate cancer, for example, castration resistant prostate cancer. In other examples, the prostate cancer is androgen-dependent prostate cancer.

The present disclosure also provides a method of modulating androgen receptor (AR) activity, the method comprising administering any of the compounds disclosed herein, or pharmaceutically acceptable salt thereof, to a subject in need thereof. For example, in certain specific embodiments modulating androgen receptor (AR) activity is for the treatment of one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration

The present disclosure also provides a pharmaceutical composition comprising any one or more of the compounds disclosed herein and a pharmaceutically acceptable carrier. The pharmaceutical composition may be for treating one or more of the following: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration.

In accordance with another embodiment, there is provided a use of the compounds of Formula (I) as described anywhere herein for preparation of a medicament for modulating androgen receptor (AR).

In accordance with a further embodiment, there is provided a method of screening for androgen receptor modulating compounds, wherein the compounds screened are selected from the compounds as described anywhere herein. The modulatmg of the androgen receptor (AR) activity may be in a mammalian cell. The modulating of the androgen receptor (AR) activity may be in a mammal. The mammal may be a human.

Alternatively, the administering may be to a mammal. The administering may be to a mammal in need thereof and in an effective amount for the treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy (e.g., Kennedy's disease), and age-related macular degeneration.

The mammalian cell may be a human cell. The modulating AR activity may be for inhibiting AR N-terminal domain activity. The modulating AR activity may be for inhibiting AR activity. The modulating may be in vivo. The modulating AR activity may be for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy (e.g., Kennedy's disease), and age-related macular degeneration. The indication may be prostate cancer. The prostate cancer may be castration-resistant prostate cancer. The prostate cancer may be androgen-dependent prostate cancer.

Table 1

Amino Acid. Polyethylene Glycol, and Phosphate Based Moieties

Amino Acid Based Moieties

any naturally occurring amino acid side



Moieties from TABLE 1 may be, for example, and without limitation, subdivided into three groups: 1) amino acid based moieties; 2) polyethylene glycol based moieties; and 3) phosphate based moieties. In the Moieties Table 1 above, the first four moieties are amino acid based moieties, the fifth and sixth are polyethylene glycol based moieties and the remaining moieties are phosphate based moieties.

The amino acid side chains of naturally occurring amino acids (as often denoted herein using "(aa)") are well known to a person of skill in the art and may be found in a variety of text books such as "Molecular Cell Biology" by James Darnell et al. Third Edition, published by Scientific American Books in 1995. Often the naturally occurring amino acids are represented by the formula (NH2)C(COOH)(H)(R), where the chemical groups in brackets are each bonded to the carbon not in brackets. R represents the side chains in this particular formula.

Those skilled in the art will appreciate that the point of covalent attachment of the moiety to the compounds as described herein may be, for example, and without limitation, cleaved under specified conditions. Specified conditions may include, for example, and without limitation, in vivo enzymatic or non-enzymatic means. Cleavage of the moiety may occur, for example, and without limitation, spontaneously, or it may be catalyzed, induced by another agent, or a change in a physical parameter or environmental parameter, for example, an enzyme, light, acid, temperature or pH. The moiety may be, for example, and without limitation, a protecting group that acts to mask a functional group, a group that acts as a substrate for one or more active or passive transport mechanisms, or a group that acts to impart or enhance a property of the compound, for example, solubility, bioavailability or localization.

In other particular embodiments of the compounds as described anywhere herein, the compound is a compound selected from one of the compounds in Table 2.

Table 2

Representative Compounds

Prodrugs are also included within the scope of the present disclosure. For example, in one embodiment the hydrogen atom of one or more hydroxyl groups of any of the compounds of Formula I may be replaced with a moiety from Table 1 {i.e., to form a OY moiety). Non-limiting examples of such prodru s include glycine esters and salts thereof as shown below.

In some embodiments, the compounds as described herein or acceptable salts thereof may be used for systemic treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. In some embodiments, the compounds as described herein or acceptable salts thereof above may be used in the preparation of a medicament or a composition for systemic treatment of an indication described herein. In some embodiments, methods of systemically treating any of the indications described herein are also provided. Some aspects of this invention make use of compositions comprising a compound described herein and a pharmaceutically acceptable excipients or carrier. In some embodiments, the prostate cancer is castration-resistant prostate cancer (also referred to as hormone ref actory, androgen-independent, androgen deprivation resistant, androgen ablation resistant, androgen depletion-independent, castration-recurrent, anti-androgen-recurrent). In some embodiments the prostate cancer is androgen-dependent or androgen-sensitive. Methods of treating any of the indications described herein are also provided. Such methods may include administering a compound as described herein or a composition of a compound as described herein, or an effective amount of a compound as described herein or composition of a compound as described herein to a subject in need thereof. Compounds as described herein include all stereoisomers. Accordingly, the compounds include racemic mixtures, enantiomers and diastereomers of any of the compounds described herein.

Compounds as described herein may be in the free form or in the form of a salt thereof. In some embodiments, compounds as described herein may be in the form of a pharmaceutically acceptable salt, which are known in the art (Berge et al., J. Pharm. Sci. 1977, 66, 1). Pharmaceutically acceptable salt as used herein includes, for example, salts that have the desired pharmacological activity of the parent compound (salts which retain the biological effectiveness and/or properties of the parent compound and which are not biologically and/or otherwise undesirable). Compounds as described herein having one or more functional groups capable of forming a salt may be, for example, formed as a pharmaceutically acceptable salt. Compounds containing one or more basic functional groups may be capable of forming a pharmaceutically acceptable salt with, for example, a pharmaceutically acceptable organic or inorganic acid. Pharmaceutically acceptable salts may be derived from, for example, and without limitation, acetic acid, adipic acid, alginic acid, aspartic acid, ascorbic acid, benzoic acid, benzenesulfonic acid, butyric acid, cinnamic acid, citric acid, camphoric acid, camphorsulfonic acid, cyclopentanepropionic acid, diethylacetic acid, digluconic acid, dodecylsulfonic acid, ethanesulfonic acid, formic acid, fumaric acid, glucoheptanoic acid, gluconic acid, glycerophosphoric acid, glycolic acid, hemisulfonic acid, heptanoic acid, hexanoic acid, hydrochloric acid, hydrobromic acid, hydriodic acid, 2-hydroxyethanesuIfonic acid, isonicotinic acid, lactic acid, malic acid, maleic acid, malonic acid, mandelic acid, methanesulfonic acid, 2- napthalenesulfonic acid, naphthalenedisulphonic acid, p-toluenesulfonic acid, nicotinic acid, nitric acid, oxalic acid, pamoic acid, pectinic acid, 3-phenylpropionic acid, phosphoric acid, picric acid, pimelic acid, pivalic acid, propionic acid, pyruvic acid, salicylic acid, succinic acid, sulfuric acid, sulfamic acid, tartaric acid, thiocyanic acid or undecanoic acid. Compounds containing one or more acidic functional groups may be capable of forming pharmaceutically acceptable salts with a pharmaceutically acceptable base, for example, and without limitation, inorganic bases based on alkaline metals or alkaline earth metals or organic bases such as primary amine compounds, secondary amine compounds, tertiary amine compounds, quaternary amine compounds, substituted amines, naturally occurring substituted amines, cyclic amines or basic ion-exchange resins. Pharmaceutically acceptable salts may be derived from, for example, and without limitation, a hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation such as ammonium, sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese or aluminum, ammonia, benzathine, meglumine, methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2- diemylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, glucamine, methylglucamine, theobromine, purines, piperazine, piperidine, procaine, N-ethylpiperidine, theobromine, tetramethylammonium compounds, tetraethylammonium compounds, pyridine, Ν,Ν-dimethylaniline, N- methylpiperidine, morpholine, N-methylmo holine J N-ethylmorpholine, dicyclohexylamine, dibenzylamine, N,N-dibenzylphenethylamine, 1-ephenamine, Ν,Ν'-m^enzylemylenediamine or polyamine resins. In some embodiments, compounds as described herein may contain both acidic and basic groups and may be in the form of inner salts or zwitterions, for example, and without limitation, betaines. Salts as described herein may be prepared by conventional processes known to a person skilled in the art, for example, and without limitation, by reacting the free form with an organic acid or inorganic acid or base, or by anion exchange or cation exchange from other salts. Those skilled in the art will appreciate that preparation of salts may occur in situ during isolation and purification of the compounds or preparation of salts may occur by separately reacting an isolated and purified compound.

In some embodiments, compounds and all different forms thereof (e.g., free forms, salts, polymorphs, isomeric forms) as described herein may be in the solvent addition form, for example, solvates. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent in physical association the compound or salt thereof. The solvent may be, for example, and without limitation, a pharmaceutically acceptable solvent. For example, hydrates are formed when the solvent is water or alcoholates are formed when the solvent is an alcohol.

In some embodiments, compounds and all different forms thereof (e.g., free forms, salts, solvates, isomeric forms) as described herein may include crystalline and amorphous forms, for example, polymorphs, pseudopolymorphs, conformational polymorphs, amorphous forms, or a combination thereof. Polymorphs include different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability and/or solubility. Those skilled in the art will appreciate that various factors including recrystallization solvent, rate of crystallization and storage temperature may cause a single crystal form to dominate.

In some embodiments, compounds and all different forms thereof {e.g., free forms, salts, solvates, polymorphs) as described herein include isomers such as geometrical isomers, optical isomers based on asymmetric carbon, stereoisomers, tautomers, individual enantiomers, individual diastereomers, racemates, diastereomeric mixtures and combinations thereof, and are not limited by the description of the formula illustrated for the sake of convenience.

In some embodiments, pharmaceutical compositions in accordance with this invention may comprise a salt of such a compound, preferably a pharmaceutically or physiologically acceptable salt. Pharmaceutical preparations will typically comprise one or more carriers, excipients or diluents acceptable for the mode of administration of the preparation, be it by injection, inhalation, topical administration, lavage, or other modes suitable for the selected treatment. Suitable carriers, excipients or diluents are those known in the art for use in such modes of administration.

Suitable pharmaceutical compositions may be formulated by means known in the art and their mode of administration and dose determined by the skilled practitioner. For parenteral administration, a compound may be dissolved in sterile water or saline or a pharmaceutically acceptable vehicle used for administration of non-water soluble compounds such as those used for vitamin K. For enteral administration, the compound may be administered in a tablet, capsule or dissolved in liquid form. The tablet or capsule may be enteric coated, or in a formulation for sustained release. Many suitable formulations are known, including, polymeric or protein microparticles encapsulating a compound to be released, ointments, pastes, gels, hydrogels, or solutions which can be used topically or locally to administer a compound. A sustained release patch or implant may be employed to provide release over a prolonged period of time. Many techniques known to one of skill in the art are described in Remington: the Science & Practice of Pharmacy by Alfonso Gennaro, 20 th ed., Lippencott Williams & Wilkins, (2000). Formulations for parenteral administration may, for example, contain excipients, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for modulatory compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.

Compounds or pharmaceutical compositions in accordance with this invention or for use in this invention may be administered by means of a medical device or appliance such as an implant, graft, prosthesis, stent, etc. Also, implants may be devised which are intended to contain and release such compounds or compositions. An example would be an implant made of a polymeric material adapted to release the compound over a period of time.

An "effective amount" of a pharmaceutical composition according to the invention includes a therapeutically effective amount or a prophylactically effective amount A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as reduced tumor size, increased life span or increased life expectancy. A therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as smaller tumors, increased life span, increased life expectancy or prevention of the progression of prostate cancer to the lethal castration resistant stage. Typically, a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount. It is to be noted that dosage values may vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners. The amount of active compound(s) in the composition may vary according to factors such as the disease state, age, sex, and weight of the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.

In some embodiments, compounds and all different forms thereof as described herein may be used, for example, and without limitation, in combination with other treatment methods for at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. For example, compounds and all their different forms as described herein may be used as neoadjuvant (prior), adjunctive (during), and/or adjuvant (after) therapy with surgery, radiation (brachytherapy or external beam), or other therapies (e.g., HEFU), and in combination with chemotherapies, androgen ablation, antiandrogens or any other therapeutic approach.

With respect to combination therapies, one embodiment of the present disclosure provides a combination of any one or more of a compound of Formula I with one or more currently-used or experimental pharmacological therapies which are or may be utilized to treat any of the above disease states (e.g., castration-resistant prostate cancer or Kennedy's disease). Methods, uses and pharmaceutical compositions comprising the above combination are also provided.

Surprisingly, it has been found that the disclosed compounds, which interfere with the AR principally through binding to the N-terminus of the AR, demonstrate beneficial synergistic therapeutic effects when used in concert with existing approved and in-development agents. That is, the biological impact of using the agents in concert with one another produces a biological and therapeutic effect which is greater than the simple additive effect of each of them separately.

Accordingly, one embodiment comprises the use of the disclosed compounds in combination therapy with one or more currently-used or experimental pharmacological therapies which are utilized for treating the above disease states irrespective of the biological mechanism of action of such pharmacological therapies, including without limitation pharmacological therapies which directly or indirectly inhibit the androgen receptor, pharmacological therapies which are cyto-toxic in nature, and pharmacological therapies which interfere with the biological production or function of androgen (hereinafter, the "Other Therapeutic Agents"). By "combination therapy" is meant the administration of any one or more of a compound of Formula I with one or more of another therapeutic agent to the same patient such that their pharmacological effects are contemporaneous with one another, or if not contemporaneous, that their effects are synergistic with one another even though dosed sequentially rather than contemporaneously.

Such administration includes without limitation dosing of one or more of a compound of Formula I and one or more of the Other Therapeutic Agent(s) as separate agents without any comingling prior to dosing, as well as formulations which include one or more Other Androgen- Blocking Therapeutic Agents mixed with one or more compound of Formula I as a pre-mixed formulation. Adininistration of the compound(s) of Formula I in combination with Other Therapeutic Agents for treatment of the above disease states also includes dosing by any dosing method including without limitation, intravenous delivery, oral delivery, intra-peritoneal delivery, intra-muscular delivery, or intra-tumoral delivery.

In another aspect of the present disclosure, the one or more of the Otitter Therapeutic

Agent may be administered to the patient before administration of the compound(s) of Formula I. In another embodiment, the compound(s) of Formula I may be co-administered with one or more of the Other Therapeutic Agents. In yet another aspect, the one or more Other Therapeutic Agent may be administered to the patient after administration of the compound(s) of Formula I. It is fully vvdthin the scope of the disclosure that the ratio of the doses of compound(s) of Formula I to that of the one or more Other Therapeutic Agents may or may not equal to one and may be varied accordingly to achieve the optimal therapeutic benefit.

For greater clarity the compound(s) of Formula I that are combined with the one or more Other Therapeutic Agents for improved treatment of the above disease states may comprise, but are not limited to any compound having a structure of Formula I, including those compounds shown in Table 2.

The Other Therapeutic Agents include without limitation any pharmacological agent which is currently approved by the FDA in the U.S. (or elsewhere by any other regulatory body) for use as pharmacological treatment of any of the above disease states, or which is currently being used experimentally as part of a clinical trial program that relates to the above disease states. Non-limiting examples of the Other Pharmacological Agents comprise, without limitation: the chemical entity known as ODM-201 (also known as BAY1841788) and related compounds, which appears to bind to AR and blocks its cellular function and is currently in clinical development as a treatment for cprostate cancer); the chemical entity known as MDV3100 (4-(3-(4-cyano-3-(trifluoromemyl)phenyl)-5,5-dimemyl-4-oxo-2 -tlu

l-yl)-2-fluoro-N-methylbenzamide) and related compounds, which appears to be a blocker of the AR LBD and is currently in development as a treatment for prostate cancer; the chemical entity known as Orteronel (TAK-700) and related compounds which appears to block androgen synthesis, the chemical entity known as TOK 001 (Galeterone) and related compounds which appears to be a blocker of the AR LBD, and a CYP17 lyase inhibitor, and also appears to decrease overall androgen receptor levels in prostate cancer cells (TOK 001 is currently in development as a treatment for prostate cancer); the chemical entity known as AR -509 (4-[7- [6-cyano-5-(trifluoromemyl)pyridin-3-yl]-8- 2-fluoro-N-methylbenzamide) and related compounds which appears to be a blocker of the AR LBD and is currently in development as a treatment for prostate cancer; the chemical entity known as abiraterone acetate (or CB-7630; (3S,8R,9S,10R,13S,14S)-10,13-dimethyl-17- (pyridin-3-yl) 2,3,4,7,8,9,10,1 l,12,13,14,15-dodecahydro-lH-cyclopenta[a]phenanthren-3-ol), and related molecules, which appears to block the production of androgen and is currently in development for the treatment of prostate cancer; the chemical entity known as bicalutamide (N-[4-cyano-3-(trifluoromemyl)phenyl]-3-[(4-fluorophenyl)sul fonyl]-2-h^

methylpropanamide) and related compounds, which appears to be a blocker of the AR LBD and which is currently used to treat prostate cancer, the chemical entity known as nilutamide (5,5- dimethyl-3-[4-nitro-3-(trifluoromethyl)phenyI] imidazolidine-2,4-dione) and related compounds, which appears to be a blocker of the AR LBD and which is currently used to treat prostate cancer, the chemical entity known as flutamide (2-methyl-N-[4-nitro-3- (trifluoromethyl)phenyl]-propanamide) and related compounds, which appears to be a blocker of the AR LBD and which is currently used to treat prostate cancer, the chemical entities know as cyproterone acetate (6-chloro- 1 p,2p-dihydro-l 7-hydroxy-3'H-cyclopropa[ 1 ,2]pregna-4,6- diene-3,20-dione) and related compounds, which appears to be a blocker of the AR LBD and which is currently used to treat prostate cancer, the chemical entity known as docetaxel (Taxotere; l,7 ,10 -trihydroxy-9-oxo-5p,20-epoxytax-l l-ene-2a,4,13a-triyl 4-acetate 2- benzoate 13-{(2R,3S)-3-[(tert-butoxycarbonyl)arnino]-2-hydroxy-3-phen ylpropanoate}) and related compounds, which appears to be a cytotoxic antimicrotubule agent and is currently used in combination with prednisone to treat prostate cancer, the chemical entity known as Bevacizumab (Avastin), a monoclonal antibody that recognizes and blocks vascular endothelial growth factor A (VEGF-A) and may be used to treat prostate cancer, the chemical entity known as OSU-HDAC42 ((S)-(+)-N-hy&oxy-4-(3-me l-2-pheny and related compounds, which appears to act as a histone deacetylase inhibitor, and is currently being developed as a treatment for prostate cancer, the chemical entity known as VITAXIN which appears to be a monoclonal antibody against the vascular integrin ανβ3 to prevent angiogenesis, and which may be used to treat prostate cancer, the chemical entity known as sunitumib (N-(2-diemylammoemyl)-5-[(Z)-(5-fluoro-2-oxo-lH-mdol-3-ylide ne)memyl]-2,^ dimethyl- lH-pyrroIe-3-carboxamide) and related compounds, which appears to inhibit multiple receptor tyrosine kinases (RTKs) and may be used for treatment of prostate cancer, the chemical entity known as ZD-4054 (N-CS-Memoxy-S-methylpyrazin^-yl^-^-il^^oxadiazol^- yl)phenyl]pyridin-3-sulfonamid) and related compounds, which appears to block the edta receptor and which may be used for treatment of prostate cancer, the chemical entity known as Cabazitaxel (XRP-6258), and related compounds, which appears to be a cytotoxic microtubule inhibitor, and which is currently used to treat prostate cancer; the chemical entity known as MDX-010 (Ipilimumab), a fully human monoclonal antibody that binds to and blocks the activity of CTLA-4 which is currently in development as an immunotherapeutic agent for treatment of prostate cancer; the chemical entity known as OGX 427 (Apatorsen) which appears to target HSP27 as an antisense agent, and which is currently in development for treatment of prostate cancer; the chemical entity known as OGX Oil (Custirsen sodium) which appears to target clusterin as an antisense agent, and which is currently in development as a treatment for prostate cancer; the chemical entity known as finasteride (Proscar, Propecia; N-(l,l- dimemylemyl)-3-oxo-(5a,17P)-4-azaandrost-l-ene-17-carboxarni de), and related compounds, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone, and may be used to treat prostate cancer; the chemical entity known as dutasteride (Avodart; 5a, 17β)-Ν-{2,5 bis(trifluoromethyl) phenyl}-3-oxo-4-azaandrost-l-ene-17-carboxamide) and related molecules, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone, and may be used in the treatment of prostate cancer; the chemical entity known as turosteride ((4aR,4bS,6aS,7S,9aS,9bS,l laR)-l,4a J 6a-trimethyl-2-oxo-N-(propan-2- yl)-N-(propan-2 ylcarbamoyl)hexadecahydro-lH-mdeno[5,4-f]quinoline-7-carboxa mide), and related molecules, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone and may be used in the treatment of prostate cancer; the chemical entity known as bexlosteride (LY-191,704; (4aS,10bR)-8-chloro-4-methyl-l,2,4a,5,6,10b- hexahydrobenzo[f]quinolin-3-one), and related compounds, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone and may be used in the treatment of prostate cancer; the chemical entity known as izonsteride (LY-320,236; (4aR,10bR)-8-[(4-ethyl- 1 ,3-benzotoazol-2-yl)sulfanyl]-4,l Ob-dm^

3(2H)-one) and related compounds, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone and may be used for the treatment of prostate cancer; the chemical entity known as FCE 28260 ((4aR,4bS,6aS,7SJ9aS,9bS,l lafl)-4a,6a-dimethyI-2-oxo-N- ((2RS)-l ,1,1 -trifluoro-2-phenyl-2-propanyl)-2,4a,4b,5,6,6a,7,8,9,9a,9b, 10,11.1 la- tetradecahydro-lH-indeno[5,4-fjquinoline-7-carboxamide) and related compounds, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone and may be used for the treatment of prostate cancer; the chemical entity known as SKF105,1H ((17β)-17- (bis-l-memylaminocarbonyl)androstane-3,5-diene-3-carboxylic acid} and related compounds, which appears to be a 5-alpha reductase inhibitor that reduces levels of dihydrotestosterone and may be used for treatment of prostate cancer.

In general, compounds of the invention should be used without causing substantial toxicity. Toxicity of the compounds of the invention can be detennined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the LD100 (the dose lethal to 100% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions. Some compounds of this invention may be toxic at some concentrations. Titration studies may be used to determine toxic and non-toxic concentrations. Toxicity may be evaluated by examining a particular compound's or composition's specificity across cell lines using PC3 cells as a negative control that do not express functional AR. Animal studies may be used to provide an indication if the compound has any effects on other tissues. Systemic therapy that targets the AR will not likely cause major problems to other tissues since antiandrogens and androgen insensitivity syndrome are not fatal.

Compounds as described herein may be administered to a subject. As used herein, a "subject" may be a human, non-human primate, mammal, rat, mouse, cow, horse, pig, sheep, goat, dog, cat and the like. The subject may be suspected of having or at risk for having a cancer, such as prostate cancer, breast cancer, ovarian cancer, salivary gland carcinoma, or endometrial cancer, or suspected of having or at risk for having acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration. Diagnostic methods for various cancers, such as prostate cancer, breast cancer, ovarian cancer, salivary gland carcinoma, or endometrial cancer, and diagnostic methods for acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration and the clinical delineation of cancer, such as prostate cancer, breast cancer, ovarian cancer, salivary gland carcinoma, or endometrial cancer, diagnoses and the clinical delineation of acne, hirsutism, alopecia, benign prostatic hyperplasia, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, or age-related macular degeneration are known to those of ordinary skill in the art.

Compounds described herein may be used for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. Compounds described herein may be used for treatment of prostate cancer. Compounds described herein may be used for treatment of castration-resistant prostate cancer. Compounds described herein may be used for treatment of androgen-dependent prostate cancer. Compounds described herein may be used for preparation of a medicament for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. Compounds described herein may be used for the preparation of a medicament for treatment of prostate cancer. Compounds described herein may be used for the preparation of a medicament for treatment of castration-resistant prostate cancer. Compounds described herein may be used for the preparation of a medicament for treatment of androgen-dependent prostate cancer. Compounds described herein may be used in a method for treatment of at least one indication selected from the group consisting of: prostate cancer, breast cancer, ovarian cancer, endometrial cancer, salivary gland carcinoma, hair loss, acne, hirsutism, ovarian cysts, polycystic ovary disease, precocious puberty, spinal and bulbar muscular atrophy, and age-related macular degeneration. The method may comprise administering to a subject in need thereof an effective amount of a compound described herein. Compounds described herein may be used in a method of treatment of prostate cancer, the method comprismg administering to a subject in need thereof an effective amount of a compound described herein. Compounds described herein may be used in a method of treatment of castration resistant prostate cancer, the method comprising administering to a subject in need thereof an effective amount of a compound described herein. Compounds described herein may be used in a method of treatment of androgen-dependent prostate cancer, the method comprismg administering to a subject in need thereof an effective amount of a compound described herein. Compounds described herein may also be used in assays and for research purposes. Definitions used include ligand-dependent activation of the androgen receptor (AR) by androgens such as dihydrotestosterone (DHT) or the synthetic androgen (R1881) used for research purposes. Ligand-independent activation of the AR refers to transactivation of the AR in the absence of androgen (ligand) by, for example, stimulation of the cAMP-dependent protein kinase (PKA) pathway with forskolin (FSK). Some compounds and compositions of this invention may inhibit both FSK and androgen (e.g., R1881) induction of ARE-luciferase (ARE- luc). Constituative activity of the AR refers to splice variants lacking the AR ligand-binding domain. Such compounds may block a mechanism that is common to both ligand-dependent and ligand-independent activation of the AR, as well as constitutively active splice variants of the AR that lack ligand-binding domain. This could involve any step in activation of the AR including dissociation of heatshock proteins, essential posttranslational modifications (e.g., acetylation, phosphorylation), nuclear translocation, protein-protein interactions, formation of the transcriptional complex, release of co-repressors, and/or increased degradation. Some compounds and compositions of this invention may inhibit ligand-only activity and may interfere with a mechanism specific to ligand-dependent activation (e.g., accessibility of the ligand binding domain (LBD) to androgen). Numerous disorders in addition to prostate cancer involve the androgen axis (e.g., acne, hirsutism, alopecia, benign prostatic hyperplasia) and compounds interfering with this mechanism may be used to treat such conditions. Some compounds and compositions of this invention may only inhibit FSK induction and may be specific inhibitors to ligand-independent activation of the AR. These compounds and compositions may interfere with the cascade of events that normally occur with FSK and/or PKA activity or any downstream effects that may play a role on the AR (e.g., FSK increases MAPK activity which has a potent effect on AR activity). Examples may include an inhibitor of cAMP and or PKA or other kinases. Some compounds and compositions of this invention may induce basal levels of activity of the AR (no androgen or stimulation of the PKA pathway). Some compounds and compositions of this invention may increase induction by R1881 or FSK. Such compounds and compositions may stimulate transcription or transactivation of the AR. Some compounds and compositions of this invention may inhibit activity of the androgen receptor. Interleukin-6 (TL-6) also causes ligand-independent activation of the AR in LNCaP cells and can be used in addition to FSK.

Compounds for use in the present invention may be obtained from medical sources or modified using known methodologies from naturally occurring compounds. In addition, methods of preparing or synthesizing compounds of the present invention will be understood by a person of skill in the art having reference to known chemical synthesis principles. For example, Auzou et al 1974 European Journal of Medicinal Chemistry 9(5), 548-554 describes suitable synthetic procedures that may be considered and suitably adapted for preparing compounds of any one of the Formula I set out above. Other references that may be helpful include: Debasish Das, Jyh-Fu Lee and Soofrn Cheng "Sulfonic acid functionalized mesoporous MCM-41 silica as a convenient catalyst for Bisphenol-A synthesis" Chemical Communications, (2001) 2178-2179; US Patent 2571217 Davis, Orris L.; Knight, Horace S.; Skinner, John R. (Shell Development Co.) "Halohydrin ethers of phenols." (1951); and Rokicki, G.; Pawlicki, J.; Kuran, . "Reactions of 4-chloromethyl-l,3-dioxolan-2-one with phenols as a new route to polyols and cyclic carbonates." Journal fuer Praktische Chemie (Leipzig) (1985) 327, 718-722. Methods for preparation of bisphenol compounds are also described in published PCT application Nos. CA2009/000902; CA2011/000019 and CA2011/000021, and in pending PCT application Nos. US2012/032584; US2012/051481 and US2012/051923. Each of the above references and patent applications are hereby incorporated by reference in their entireties for all purposes.

For example, compounds of the present invention may be prepared with reference to the following General Reaction Scheme I:

General Reaction Scheme I

Referring to Synthetic Scheme 1, compounds of structure A can be purchased from commercial sources or prepared according to methods known in the art. Reaction of A with an appropriately epoxide under basic conditions, yields compounds of structure B. Optically pure or racemic epoxides may be employed to yield the desired stereochemistry. Reaction of B with an appropriately substituted chloropropane, for example a bromo-chloropropane, results in compounds of structure C.

Fluorination of C results in D. Methods for such fluorination are well known. For example, in one embodiment a fluorine atom is introduced by treatment with memylaminosiufurtrifluoride (DAST) or Xtalfluor-E or M (see J. Org. Chem. 2010, 75, 3401- 3411, which is hereby incorporated by reference in its entirety). In other embodiments, the hydroxyl moiety in C may be converted to an appropriate leaving group, for example by reaction with tosyl chloride or mesyl anhydride, followed by reaction with [K + /2,2,2-cryptand]F " or tetrabutylammonium fluoride. Other methods for fluorination of C are known to those of skill in the art. For descriptions of fluorination procedures see J. Org. Chem. 2010, 75, 3401-3411, Bioorg Med. Chem. 2009, 17, 7441-7448, and J. Med. Chem, 1990, 33, 2430-2437, each of which is hereby incorporated by reference in its entirety.

Alternatively, the compounds can be prepared according to the following General Reaction Scheme II:

General Reaction Scheme II

F

Referring to General Reaction Scheme Π, compound E, wherein P is an appropriate alcohol protecting group, such as tetrahydropyran, can be prepared according to methods known in the art and described herein. Similarly, fluorinated reagent F can be prepared according to the general procedures outlined in the Examples herein or by other means known in the art. Reaction of E and Γ under basic conditions, followed by deprotection yields compounds of structure G. Compounds of structure D are then prepared using an appropriate chloropropane as described above.

One skilled in the art will recognize that variations to the order of the steps and reagents discussed in reference to Synthetic Scheme I are possible. Further, fluorine atoms may be introduced via any number of reagents, and fluorination is not limited to those methods depicted or described above. Methods for such fluorination are well known in the art. Finally, prodrugs of Formula I can be prepared by functionalizing a free hydroxyl in structure C or D. Methods for such functionalization are well-known in the art, for example reaction with an acid chloride analogue of a moiety from Table 1 or any other suitable reagent. Methodologies for preparation of compounds of Formula I are described in more detail in the following non-limiting exemplary schemes. Various alternative embodiments and examples of the invention are described herein. These embodiments and examples are illustrative and should not be construed as limiting the scope of the invention.

EXAMPLES

All non-aqueous reactions were performed in flame-dried round bottomed flasks. The flasks were fitted with rubber septa and reactions were conducted under a positive pressure of argon unless otherwise specified. Stainless steel syringes were used to transfer air- and moisture-sensitive liquids. Flash column chromatography was performed as described by Still et al. (Still, W. C; Kahn, M.; Mitra, A. J. Org. Chem. 1978, 43, 2923) using 230-400 mesh silica gel. Thin-layer chromatography was performed using aluminum plates pre-coated with 0.25 mm 230-400 mesh silica gel impregnated with a fluorescent indicator (254 nm). Thin-layer chromatography plates were visualized by exposure to ultraviolet light and a "Seebach" staining solution (700 mL water, 10.5 g Cerium (IV) sulphate tetrahydrate, 15.0 g molybdato phosphoric acid, 17.5 g sulphuric acid) followed by heating (~1 min) with a heating gun (~250 °C), Organic solutions were concentrated on Biichi R-l 14 rotatory evaporators at reduced pressure (15-30 torr, house vacuum) at 25-40 °C.

Commercial regents and solvents were used as received. All solvents used for extraction and chromatography were HPLC grade. Normal-phase Si gel Sep paks™ were purchased from waters, Inc. Thin-layer chromatography plates were Kieselgel 60F 25 4. All synthetic reagents were purchased from Sigma Aldrich and Fisher Scientific Canada.

Proton nuclear magnetic resonance (¾ NMR) spectra were recorded at 25 °C using a Bruker 400 with inverse probe and Bruker 400 spectrometers, are reported in parts per million on the δ scale, and are referenced from the residual protium in the NMR solvent (DMSO-ifa δ 2.50 (DMSO-i¾), CDC1 3 : δ 7.24 (CHC1 3 )). Carbon-13 nuclear magnetic resonance ( 13 C NMR) spectra were recorded with a Bruker 400 spectrometer, are reported in parts per million on the δ scale, and are referenced from the carbon resonances of the solvent (DMSO-i¾: δ 39.51, CDC1 3 : δ 77.00). Spectral features are tabulated in the following order: chemical shift (δ, ppm); multiplicity (s = singlet, d = doublet, t = triplet, m = multiplet, br = broad); coupling constant (J, Hz, number of protons).

LNCaP cells are employed initially for all experiments because they are well-differentiated human prostate cancer cells in which ligand-independent activation of the AR by FSK has been characterized (Nazareth et al 1996 J. Biol. Chem. 271, 19900-19907; and Sadar 1999 J. Biol. Chem. 274, 7777-7783). LNCaP cells express endogenous AR and secrete prostate-specific antigen (PSA) (Horoszewicz et al 1983 Cancer Res. 43, 1809-1818). LNCaP cells can be grown either as monolayers in cell culture or as tumors in the well-characterized xenograft model that progresses to castration resistance in castrated hosts (Sato et al 1996 J. Steroid Biochem. Mol. Biol. 58, 139-146; Gleave et al 1991 Cancer Res. 51, 3753-3761; Sato et al 1997 Cancer Res. 57, 1584-1589; and Sadar et al 2002 Mol. Cancer Ther. 1(8), 629-637). R1881 is employed since it is stable and avoids problems associated with the labile physiological ligand dihydrotestosterone (DHT). Reporter specificity may be determined using several alternative reporter gene constructs. Some well characterized ARE-driven reporter gene constructs that have been used extensively are the PSA (6.1 kb) enhance/promoter which contains several AREs and is highly inducible by androgens as well as by FSK (Ueda et al 2002 A J. Biol. Chem. 277, 7076-7085) and the ARR3-thymidine kinase (tk)-luciferase, which is an artificial reporter construct that contains three tandem repeats of the rat probasin AREl and ARE2 regions upstream of a luciferase reporter (Snoek et al 1996 J. Steroid Biochem. Mol. Biol. 59, 243-250).

EXAMPLE 1

SYNTHESIS OF (S)-3-(4-(2-(4-(3-CHLO OP OPOXY)PHENYL)PROPAN-2-YL)PHENOXY)PROPANE- 1 ,2-

DIOL

Potassium carbonate anhydrous (14 g, 103.80 mmol, 2 equiv) was added to a stirred solution of Bisphenol A (11.83 g, 51.88 mmol, 1 equiv) in anhydrous dimethyl formamide (35 mL), at room temperature, and the contents were stirred under an atmosphere of argon for 20 min. (S)-(-)-glycidol (4.48 mL, 67.45 mmol, 1.3 equiv) was added slowly via syringe, and the mixture was allowed to react at 80 °C for 5 h. Then, the reaction was quenched by the addition of a saturated solution of ammonium chloride (10 mL), and the mixture was extracted with ethyl acetate (3 x 35 mL). The organic layer was washed with deionized water (20 mL), was dried over anhydrous magnesium sulfate, was filtered, and was concentrated under reduced pressure. Flash column chromatography on silica gel (eluent: 40% to 80% ethyl acetate in hexane) provided Bisphenol A (2,3-dihydroxypropyI) ether (8.06 g, 50%) as colorless foam.

Potassium carbonate anhydrous (7.3 g, 52.91 mmol, 2 equiv) was added to a stirred solution of Bisphenol A (2,3-dihydroxypropyl) ether (8.06 g, 26.45 mmol, 1 equiv) in anhydrous dimethyl formamide (25 mL), at room temperature, and the contents were stirred under an atmosphere of argon for 20 min. l-Bromo-3-chloropropane (7.8 mL, 79.35 mmol, 3 equiv) was added slowly via syringe and the mixture was allowed to react at room temperature for 4 days. Then, the reaction was quenched by the addition of a saturated solution of ammonium chloride (10 mL), and the mixture was extracted with ethyl acetate (3 x 25 mL). The organic layer was washed with deionized water (15 mL), was dried over anhydrous magnesium sulfate, was filtered, and was concentrated under reduced pressure. Flash column chromatography on silica gel (eluent: 40% to 70% ethyl acetate in hexane) provided titled compound (7.0 g, 70%) as a colorless foam.

EXAMPLE 2

(R)-l-(4-(2-(4-(3-CHLOROPROPOXY)PHE YL)PROPAN-2-YL)PHENOXY)-3-FLUOROPROPAN-2-OL

(IB)

To a solution of (S)-3-(4-(2-(4-(3-chloropropoxy)phenyl)propan-2-yl)phenoxy)p ropane- 1,2-diol (1 equiv) in dichloromethane were successively added triethylamine trihydrofluorideXl equiv) and XTalFluor-M (1 equiv). After 24 h, the reaction mixture was quenched at room temperature with a 5% aqueous sodium bicarbonate solution and stirred for 15 min, and the resulting mixture was extracted twice with dichloromethane. The organic phased were combined, dried over anhydrous magnesium sulfate, and filtered. Solvents were evaporated, and the resulting crude material was purified by silica gel flash chromatography to provide compound lb.

EXAMPLE 3

P EPARATION OF TIPS PROTECTED FLUOROHYDRIN

To a solution of racemic epifluorohydrin (187 ί, 2.62 mmol, 1 equiv) in acetonitrile (10 mL) was added p-toluenesulfonic acid monohydrate (747 mg, 3.93 mmol, 1.5 equiv) and solid Bismuth(III) trifiuoromethanesulfonate (172 mg, 0.262 mmol, 1/10 equiv) in one portion and the mixture was stirred at room temperature for 4 h. Sodium bicarbonate was added (1 mL), the organic solvent was evaporated under reduced pressure and the residue was extracted with dichloromethane (3 x 5 mL). The combined organic extracts were dried over Na 2 S0 4 and concentrated under reduced pressure to give fluorohydrin (546 mg, 84%) as clear oil that was directly used in the next step.

A round-bottomed flask was charged sequentially with fluorohydrin (1003 mg, 4.17 mmol, 1 equiv) and imidazole (567 mg, 8.34 mmol, 2 equiv), and the contents were placed under an atmosphere of argon. Anhydrous dimethyl formamide (2 mL) was introduced via syringe and the resulting mixture was stirred at room temperature for 5 min. Subsequently, a solution of triisopropylsilylchloride (1.15 mL, 5.43 mmol, 1.3 equiv) in anhydrous DMF (2 mL) was added dropwise at room temperature. The reaction mixture was stirred for 2 h at rt and quenched with deionized water (3 mL). The mixture was extracted with ethyl acetate (3 4 mL). The organic layer was washed with deionized water (2 mL), was dried over anhydrous magnesium sulfate, was filtered, and was concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel (eluent: 100% hexane to 20% ethyl acetate in hexane) to provide the titled compound (965 mg, 58%) as a clear liquid.

EXAMPLE 4

P -(2-(4-(TETRAHYD O-2H-PYRAN-2-YLOXY)P -2-YL)PHENOL

To a solution of racemic derivative Bisphenol A (2 g, 8.76 mmol, 1 equiv) in anhydrous tetrahydrofuran (3 mL) and methylene chloride (3 mL) was added 3,4-Dihydro-2H-pyran (640 yL, 7.01 mmol, 0.8 equiv) followed by catalytic amounts of Pyridinium p-toluene sulfonate and the mixture was stirred at room temperature for 17 h. The resulting solution was concentrated under reduced pressure and the residue was purified by flash column chromatography on silica gel (eluent: 20% to 30% ethyl acetate in hexane) to provide the title compound (1.5 g, 55%) as colorless foam. EXAMPLE 5

(L-FLUORO-3-(4-(2-(4-(TETRAHYD O-2H-PYRAN-2-YLOXY)PHENYL)PROPAN-2- -2-YLOXY)TRIISOPROPYLSILANE

Potassium carbonate anhydrous (44 mg, 0.32 mmol, 2 equiv) was added to a stirred solution of Bisphenol A mono-THP (50 mg, 0.16 mmol, 1 equiv) in anhydrous dimethyl formamide (2 mL), at room temperature, and the contents were stirred under an atmosphere of argon for 20 min. A solution of TIPS protected fluorohydrin (129 mg, 0.32 mmol, 2 equiv) in anhydrous dimethyl formamide (1 mL) was added slowly via syringe and the mixture was allowed to react at 80 D C overnight. Then, the reaction was quenched by the addition of a saturated solution of ammonium chloride (1 mL), and the mixture was extracted with ethyl acetate (3 x 5 mL). The organic layer was washed with deionized water (5 mL), was dried over anhydrous magnesium sulfate, was filtered, and was concentrated under reduced pressure. Flash column chromatography on silica gel (eluent: 20% to 30% ethyl acetate in hexane) provided titled compound (82, 94%) as colorless foam.

EXAMPLE 6

4- -(4-(3-FLUORO-2-HYDROXYPROPOXY)PHENYL)PROPAN-2-YL)PHENOL

To a solution of derivative bisphenol A (82 mg, 0.15 mmol, 1 equiv) in methanol (2 mL) was added catalytic amounts of p-toluenesulfonic acid monohydrate and the contents were stirred at room temperature overnight. Sodium bicarbonate was added (1 mL), the organic solvent was evaporated under reduced pressure and the residue was extracted with dichloromethane (3 x 5 mL). The combined organic extracts were dried over Na2S04 and concentrated under reduced pressure. Flash column chromatography on silica gel (eluent: 20% to 40% ethyl acetate in hexane) provided titled compound (44, 96%) as colorless foam.

EXAMPLE 7

1

Potassium carbonate anhydrous (30 mg, 0.22 mmol, 1.5 equiv) was added in one portion to a stirred solution of bisphenol A derivative (44 mg, 0.14 mmol, 1 equiv) in anhydrous dimethyl formarnide (2 mL), at room temperature, and the contents were stirred under an atmosphere of argon for 20 min. l-Bromo-3-chloropropane (28 uL, 0.28 mmol, 2 equiv) was added slowly via syringe and the mixture was allowed to react at room temperature for 3 days. Then, the reaction was quenched by the addition of a saturated solution of ammonium chloride (3 mL), and the mixture was extracted with ethyl acetate (3 x 10 mL). The organic layer was washed with deionized water (15 mL), was dried over anhydrous magnesium sulfate, was filtered, and was concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel (eluent: 20% to 40% ethyl acetate in hexane) to provide titled compound (33 mg, 88%) as pale foam.

EXAMPLE 8

IN VITRO ACTIVITY OF COMPOUNDS

LNCaP cells are transiently cotransfected with PSA (6.1 kb)-luciferase (0.25 μg/weH) in 24-well plates for 24 h prior to pre-treatment with compounds of the invention for 1 hour before the addition of synthetic androgen, R1881 (1 nM) to induce PSA production or vehicle. The total amount of plasmid DNA transfected is normalized to 0.75 pg well by the addition of the empty vector. After 48 h of incubation with R1881, the cells are harvested, and relative luciferase activity is determined. Test compounds are added to the cells at various concentrations and activity for each treatment is normalized to the predicted maximal activity induction (in the absence of test compounds, vehicle only). Plotting of sigmoidal curves (Boltzmann Function) and IC50 calculations are done using OriginPro 8.1 Sofware (Northampton, MA, USA).

Furthermore, toxicity is assessed by both microscopic examination and reduction of protein levels. Solubility is assessed both macroscopically (cloudy media) and microscopically (formation of granules or crystals).

EXAMPLE 9

IN VIVO DOSE RESPONSE OF COMPOUNDS

In vivo dose response of compounds of the invention is determined according to the following procedure: Male athymic SCID-NOD mice, 6- to 8-weeks old, are inoculated subcutaneously with LNCaP cells (1 x 10 6 ) suspended in 75 μΐ of RPMI 1640 (5% FBS) and 75 μΐ of Matrigel (Becton Dickinson Labware) in the flank region via a 27-gauge needle under isofluorane anesthesia. Mice bearing LNCaP subcutaneous tumors are castrated when tumor volumes are approximately 100 mm 3 . Seven days after castration, mice are injected intravenously by tail vein every other day for a total of 7 doses with compounds of the invention in 15% DMSO and 25.5% PEG. The experiment is complete 2 days after the last injection. Tumours are measured with calipers and their volumes calculated by the formula L x W x H x 0.5236. Tumor volume as a function of compound dose is plotted.

Dose responses of comparative compounds are also determined according to the above procedure.

Although various embodiments of the invention are disclosed herein, many adaptations and modifications may be made within the scope of the invention in accordance with the common general knowledge of those skilled in this art. Such modifications include the substitution of known equivalents for any aspect of the invention in order to achieve the same result in substantially the same way. Numeric ranges are inclusive of the numbers defining the range. The word "comprising" is used herein as an open-ended term, substantially equivalent to the phrase "including, but not limited to", and the word "comprises" has a corresponding meaning. As used herein, the singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a thing" includes more than one such thing. Citation of references herein is not an admission that such references are prior art to the present invention. Any priority document(s) and all publications, including but not limited to patents and patent applications, cited in this specification are incorporated herein by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein and as though fully set forth herein. The invention includes all embodiments and variations substantially as hereinbefore described and with reference to the examples and drawings.