Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SMALL MOLECULE INHIBITORS OF THE NUCLEAR TRANSLOCATION OF ANDROGEN RECEPTOR FOR THE TREATMENT OF CASTRATION-RESISTANT PROSTATE CANCER
Document Type and Number:
WIPO Patent Application WO/2019/222105
Kind Code:
A1
Abstract:
A compound, or a pharmaceutically acceptable salt or ester thereof, according to formula I: R20 –(Z)b–(Y)c–(R21)a–(X)d–R22–R23 wherein R20 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, a thio-containing group, or a seleno-containing group; Z is alkanediyl, substituted alkanediyl, cycloalkanediyl, or substituted cycloalkanediyl; Y is S, O, S(=O), –S(=O)(=O)-, or NR10, wherein R10 is H or alkyl; R21 is alkanediyl, substituted alkanediyl, cycloalkanediyl, substituted cycloalkanediyl, alkadienyl, substituted alkadienyl, cycloalkenediyl, substituted cycloalkenediyl, alkatrienyl, substituted alkatrienyl; X is ‑C(=O)-, ‑S(=O)(=O)-, or–N(H)C(=O)-; R22 includes at least one divalent amino radical; R23 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, a thio-containing group, or a seleno-containing group; a, b, c, and d independently are 0 or 1.

Inventors:
WIPF PETER (US)
JOHNSON JAMES K (US)
SKODA ERIN M (US)
NELSON JOEL B (US)
WANG ZHOU (US)
TAI SERENE (US)
TAKUBO KEITA (US)
MILLIGAN JOHN (US)
Application Number:
PCT/US2019/032033
Publication Date:
November 21, 2019
Filing Date:
May 13, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV PITTSBURGH COMMONWEALTH SYS HIGHER EDUCATION (US)
WIPF PETER (US)
JOHNSON JAMES K (US)
SKODA ERIN M (US)
NELSON JOEL B (US)
WANG ZHOU (US)
TAI SERENE (US)
TAKUBO KEITA (US)
MILLIGAN JOHN (US)
International Classes:
A61K31/12; C07D295/185; A61K31/165; A61K31/40; A61K31/403; A61K31/407; A61K31/495; A61K31/496; A61K31/498; A61P35/00; C07C49/563; C07D207/06; C07D207/08; C07D209/52; C07D233/61; C07D409/02; C07D487/04
Domestic Patent References:
WO2017165822A12017-09-28
Foreign References:
US20190022093A12019-01-24
Other References:
DATABASE Registry 1 March 2018 (2018-03-01), "CA Index Name: 1-Piperazineacetic acid, alpha-(2- chlorophenyl)-4-[[2-[2-(trifluoromethyl)phenyl]cyclopropy1]carbonyl]- , methyl ester", retrieved from STN Database accession no. RN 2180239-98-7
DATABASE Registry 11 August 2011 (2011-08-11), "CA Index Name: 1-Piperazineacetic acid, alpha-(2- chlorophenyl)-4-[[2-(2-chlorophenyl)cyclopropyl]carbonyl]- , methyl ester", retrieved from STN Database accession no. RN- 1316002-05-7
DATABASE Registry 12 May 2011 (2011-05-12), "CA Index Name: Methanone, [4-[1-(2-chlorophenyl) ethyl]-1-piperazinyl][2-(2-fluorophenyl)cyclopropyl", retrieved from STN Database accession no. RN 1293729-94-8
DATABASE Registry 19 September 2014 (2014-09-19), "CA Index Name: 1-Piperazineacetic acid, alpha-(4- chlorophenyl)-4-[[2-(3,4-difluorophenyl)cyclopropyl]carbonyl]-, methyl ester", retrieved from STN Database accession no. RN 1623249-86-4
See also references of EP 3793982A4
Attorney, Agent or Firm:
GARDNER, Gillian (US)
Download PDF:
Claims:
What is claimed is:

1. A compound, or a stereoisomer, pharmaceutically acceptable salt, or ester thereof, according to:

(i) any one of formulas IV-XVI

formula VI, formula VII,

formula X, formula XI,

formula XII, formula XIII,

formula XVI,

wherein R20 is phenyl substituted with C1-C3 perfluoroalkyl, halo, or pentafluorosulfanyl; R24-R27 independently are hydrogen, deuterium, or halo;

R28 is O, N(CH3), or C¾;

R29 is N, O, or S;

R30 is CH or N;

each R31 independently is C1-C3 alkyl, C1-C3 perfluoroalkyl, halo, pentafluorosulfanyl, - C(0)0alkyl, or C(0)N(H)alkyl; and

q is 1, 2, or 3, or

2 The compound of claim 1, wherein the compound has a structure according to any one of formulas IV -XVI and at least one of the following conditions is satisfied:

q is not 1, or

R20 is not

3. The compound of claim 1 or claim 2, wherein R20 is phenyl substituted with -CF3, -SF5, or -F.

4. The compound of any one of claims 1-3, wherein R20 is substituted at the C3 or C4 position.

5. The compound of any one of claims 1-4, wherein each R31 independently is C1-C3 alkyl, Ci- C3 perfluoroalkyl, or halo.

6. The compound of any one of claims 1-5, wherein each R31 independently is methyl, trifluoromethyl, or chloro. 7. The compound of any one of claims 1-6, wherein q is 2, and the R31 substituents are para to one another.

8. The compound of claim 1, wherein the compound is:

9. A pharmaceutical composition comprising at least one pharmaceutically acceptable additive, and a compound of any one of claims 1-8.

10. A method for treating prostate cancer in a subject, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-8.

11. The method of claim 10, wherein the prostate cancer is castration-resistant prostate cancer.

12. The method of claim 10 or claim 11, wherein the compound is orally administered.

13. The method of any one of claims 10-12, wherein the method is used in combination with androgen deprivation therapy.

14. The method of any one of claims 10-13, wherein the compound is co-administered with abiratrone.

15. The method of any one of claims 10-13, wherein the compound is co-administered with enzalutamide. 16. The method of any one of claims 10-15, wherein the method further comprises identifying a subject that is in need of treatment with the compound.

Description:
SMALL MOLECULE INHIBITORS OF THE NUCLEAR TRANSLOCATION OF

ANDROGEN RECEPTOR FOR THE TREATMENT OF CASTRATION-RESISTANT

PROSTATE CANCER

CROSS-REFERENCE TO RELATED APPLICATION

This application claims the benefit of the earlier filing date of U.S. provisional application No. 62,671,254, filed May 14, 2018, which is incorporated by reference in its entirety herein.

ACKNOWLEDGMENT OF GOVERNMENT SUPPORT

This invention was made with government support under grant #GM067082 awarded by the National Institutes of Health. The government has certain rights in the invention.

SUMMARY

Disclosed herein is a compound, or a pharmaceutically acceptable salt or ester thereof, having a formula I of:

R 20 - (Z)b- (Y)c- (R 21 )a- (X)d - R 22 - R 23

wherein R 20 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, amino, a thio-containing group, or a seleno- containing group; Z is alkanediyl, substituted alkanediyl, cycloalkanediyl, or substituted cycloalkanediyl; Y is S, O, S(=0), -S(=0)(=0)-, or NR 10 , wherein R 10 is H or alkyl; R 21 is alkanediyl, substituted alkanediyl, cycloalkanediyl, substituted cycloalkanediyl, alkadienyl, substituted alkadienyl, cycloalkenediyl, substituted cycloalkenediyl, alkatrienyl, substituted alkatrienyl; X is -C(=0)-, -S(=0)(=0)-, or-N(H)C(=0)-; R 22 is a moiety that includes at least one divalent amino radical; R 23 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, amino, a thio-containing group, a seleno-containing group; a is 0 or 1; b is 0 or 1; c is 0 or 1; and d is 0 or 1. In some embodiments, if X is -C(=0)- then Y is not S. In certain embodiments, R 21 is cycloalkanediyl. When R 21 is cycloalkanediyl, R 20 may be a phenyl optionally substituted with at least one halogen and/or R 23 may be a phenyl substituted with at least one halogen and/or at least one alkyl.

Also disclosed herein is a method for treating prostate cancer in a subject, comprising administering a therapeutically effective amount of an agent to the subject, wherein the agent is a compound, or a pharmaceutically acceptable salt or ester thereof, of formula I or formula II.

The foregoing will become more apparent from the following detailed description, which proceeds with reference to the accompanying figures.

BRIEF DESCRIPTION OF THE DRAWINGS

Figs. 1A through II show compound structures. Fig. 2 is a reaction scheme showing the synthesis of 2-((isoxazol-4-ylmethyl)thio)-l-(4- phenylpiperazin-l-yl)ethanone 1.

Fig. 3 is a chemical structure of 2-((isoxazol-4-ylmethyl)thio)-l-(4-phenylpiperazin-l-yl)etha none showing zones of modification.

Figs. 4-25 are reaction schemes showing synthesis of certain embodiments of the disclosed compounds.

DETAILED DESCRIPTION

The following explanations of terms and methods are provided to better describe the present compounds, compositions and methods, and to guide those of ordinary skill in the art in the practice of the present disclosure. It is also to be understood that the terminology used in the disclosure is for the purpose of describing particular embodiments and examples only and is not intended to be limiting.

“Administration of’ and“administering a” compound should be understood to mean providing a compound, a prodrug of a compound, or a pharmaceutical composition as described herein. The compound or composition can be administered by another person to the subject (e.g., intravenously) or it can be self- administered by the subject (e.g., tablets).

“Alkanediyl” or“cycloalkanediyl” refers to a divalent radical of the general formula -C n th n - or -C„H2 n -2-, respectively, derived from aliphatic or cycloaliphatic hydrocarbons. “Cycloalkenediyl” refers to a divalent radical of the general formula -C„H2 n -4- derived from a cycloalkene.

The term "aliphatic" is defined as including alkyl, alkenyl, alkynyl, halogenated alkyl and cycloalkyl groups as described above. A "lower aliphatic" group is a branched or unbranched aliphatic group having from 1 to 10 carbon atoms.

The term "alkyl" refers to a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, «-propyl, isopropyl, «-butyl, isobutyl, f-butyl, pentyl, hexyl, heptyl, octyl, decyl, tetradecyl, hexadecyl, eicosyl, tetracosyl and the like. A "lower alkyl" group is a saturated branched or unbranched hydrocarbon having from 1 to 6 carbon atoms. Preferred alkyl groups have 1 to 4 carbon atoms. Alkyl groups may be“substituted alkyls” wherein one or more hydrogen atoms are substituted with a substituent such as halogen, cycloalkyl, alkoxy, amino, hydroxyl, aryl, alkenyl, or carboxyl. For example, a lower alkyl or (Ci-Ce)alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3- pentyl, or hexyl; (C3-C6)cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; (C3- C 6 )cycloalkyl(Ci-Ce)alkyl can be cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl,

cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethyl, or 2-cyclohexylethyl; (Ci- Ce)alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec-butoxy, pentoxy, 3- pentoxy, or hexyloxy; (C2-Ce)alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3- butenyl, 1,-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1- hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5- hexenyl; (C2-Ce)alkynyl can be ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1- pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1- hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, or 5-hexynyl; (Ci-Ce)alkanoyl can be acetyl, propanoyl or butanoyl; halo(Ci-C 6 )alkyl can be iodomethyl, bromomethyl, chloromethyl, fluoromethyl, trifluoromethyl, 2-chloroethyl, 2-fluoroethyl, 2,2,2-trifluoroethyl, or pentafluoroethyl; hydroxy(Ci-Ce)alkyl can be hydroxymethyl, 1 -hydro xyethyl, 2 -hydroxy ethyl, 1- hydroxypropyl, 2-hydroxypropyl, 3-hydro xypropyl, 1-hydroxybutyl, 4-hydroxybutyl, 1-hydroxypentyl, 5- hydroxypentyl, 1 -hydro xyhexyl, or 6-hydro xyhexyl; (Ci-Ce)alkoxycarbonyl can be methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, or

hexyloxycarbonyl; (Ci-Ce)alkylthio can be methyl thio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, pentylthio, or hexylthio; (C2-Ce)alkanoyloxy can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy.

The term "alkylaryl" refers to a group in which an alkyl group is substituted for a hydrogen atom of an aryl group. An example is -Ar-R, wherein Ar is an arylene group and R is an alkyl group.

The term“alkoxy” refers to a straight, branched or cyclic hydrocarbon configuration and combinations thereof, including from 1 to 20 carbon atoms, preferably from 1 to 8 carbon atoms (referred to as a“lower alkoxy”), more preferably from 1 to 4 carbon atoms, that include an oxygen atom at the point of attachment. An example of an "alkoxy group" is represented by the formula -OR, where R can be an alkyl group, optionally substituted with an alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, halogenated alkyl, alkoxy or heterocycloalkyl group. Suitable alkoxy groups include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, sec-butoxy, tert-butoxy cyclopropoxy, cyclohexyloxy, and the like.

"Alkoxycarbonyl" refers to an alkoxy substituted carbonyl radical, -C(0)OR, wherein R represents an optionally substituted alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl or similar moiety.

“Alkynyl” refers to a cyclic, branched or straight chain group containing only carbon and hydrogen, and unless otherwise mentioned typically contains one to twelve carbon atoms, and contains one or more triple bonds. Alkynyl groups may be unsubstituted or substituted. “Lower alkynyl” groups are those that contain one to six carbon atoms.

The term "amide" or“amido” is represented by the formula -C(0)NRR', where R and R' independently can be a hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, halogenated alkyl, or heterocycloalkyl group described above. A suitable amido group is acetamido.

The term "amine" or "amino" refers to a group of the formula -NRR', where R and R' can be, independently, hydrogen or an alkyl, alkenyl, alkynyl, aryl, arylalkyl, carbonyl (e.g, -C(0)R”, where R” can be hydrogen, an alkyl, alkenyl, alkynyl, aryl, or an arylalkyl), cycloalkyl, halogenated alkyl, or heterocycloalkyl group. For example, an“alkylamino” or“alkylated amino” refers to -NRR', wherein at least one of R or R' is an alkyl.

"Aminocarbonyl" alone or in combination, means an amino substituted carbonyl (carbamoyl) radical, wherein the amino radical may optionally be mono- or di-substituted, such as with alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkanoyl, alkoxycarbonyl, aralkoxycarbonyl and the like. An aminocarbonyl group may be -C(0)-N(R) (wherein R is a substituted group or H). An“aminocarbonyl” is inclusive of an amido group. A suitable aminocarbonyl group is acetamido. An“analog” is a molecule that differs in chemical structure from a parent compound, for example a homolog (differing by an increment in the chemical structure or mass, such as a difference in the length of an alkyl chain or the inclusion of one of more isotopes), a molecular fragment, a structure that differs by one or more functional groups, or a change in ionization. An analog is not necessarily synthesized from the parent compound. Structural analogs are often found using quantitative structure activity relationships (QSAR), with techniques such as those disclosed in Remington ( The Science and Practice of Pharmacology, 19th Edition (1995), chapter 28). A derivative is a molecule derived from the base structure.

An“animal” refers to living multi-cellular vertebrate organisms, a category that includes, for example, mammals and birds. The term mammal includes both human and non-human mammals.

Similarly, the term“subject” includes both human and non-human subjects, including birds and non-human mammals, such as non-human primates, companion animals (such as dogs and cats), livestock (such as pigs, sheep, cows), as well as non-domesticated animals, such as the big cats. The term subject applies regardless of the stage in the organism’s life-cycle. Thus, the term subject applies to an organism in utero or in ovo, depending on the organism (that is, whether the organism is a mammal or a bird, such as a domesticated or wild fowl).

The term "aryl" refers to any carbon-based aromatic group including, but not limited to, phenyl, naphthyl, etc. The term "aryl" also includes "heteroaryl group," which is defined as an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorous. The aryl group can be substituted with one or more groups including, but not limited to, alkyl, alkynyl, alkenyl, aryl, halide, nitro, amino, ester, ketone, aldehyde, hydroxy, carboxylic acid, or alkoxy, or the aryl group can be unsubstituted.

The term "arylalkyl" refers to an alkyl group where at least one hydrogen atom is substituted by an aryl group. An example of an arylalkyl group is a benzyl group.

"Carbonyl" refers to a group of the formula -C(O)-. Carbonyl-containing groups include any substituent containing a carbon-oxygen double bond (C=0), including acyl groups, amides, carboxy groups, esters, ureas, carbamates, carbonates and ketones and aldehydes, such as substituents based on -COR or - RCHO where R is an aliphatic, heteroaliphatic, alkyl, heteroalkyl, hydroxyl, or a secondary, tertiary, or quaternary amine.

"Carboxyl" refers to a -COO group. Substituted carboxyl refers to -COOR where R is aliphatic, heteroaliphatic, alkyl, heteroalkyl, or a carboxylic acid or ester.

The term "cycloalkyl" refers to a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. The term "heterocycloalkyl group" is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorous.

The term“co- administration” or“co-administering” refers to administration of a first agent with a second agent within the same general time period, and does not require administration at the same exact moment in time (although co- administration is inclusive of administering at the same exact moment in time). Thus, co- administration may be on the same day or on different days, or in the same week or in different weeks. The first agent and the second agent may be included in the same composition or they may each individually be included in separate compositions. In certain embodiments, the two agents may be administered during a time frame wherein their respective periods of biological activity overlap. Thus, the term includes sequential as well as coextensive administration of two or more agents.

"Derivative" refers to a compound or portion of a compound that is derived from or is theoretically derivable from a parent compound.

The terms "halogenated alkyl" or "haloalkyl group" refer to an alkyl group as defined above with one or more hydrogen atoms present on these groups substituted with a halogen (F, Cl, Br, I).

The term "hydroxyl" is represented by the formula -OH.

The term "hydroxyalkyl" refers to an alkyl group that has at least one hydrogen atom substituted with a hydroxyl group. The term "alkoxyalkyl group" is defined as an alkyl group that has at least one hydrogen atom substituted with an alkoxy group described above.

“Inhibiting” refers to inhibiting the full development of a disease or condition. “Inhibiting” also refers to any quantitative or qualitative reduction in biological activity or binding, relative to a control.

“N-heterocyclic” refers to mono or bicyclic rings or ring systems that include at least one nitrogen heteroatom. The rings or ring systems generally include 1 to 9 carbon atoms in addition to the heteroatom(s) and may be saturated, unsaturated or aromatic (including pseudoaromatic). The term "pseudoaromatic" refers to a ring system which is not strictly aromatic, but which is stabilized by means of delocalization of electrons and behaves in a similar manner to aromatic rings. Aromatic includes pseudoaromatic ring systems, such as pyrrolyl rings.

Examples of 5-membered monocyclic N-heterocycles include pyrrolyl, H-pyrrolyl, pyrrolinyl, pyrrolidinyl, oxazolyl, oxadiazolyl, (including 1,2,3 and 1,2,4 oxadiazolyls) isoxazolyl, furazanyl, thiazolyl, isothiazolyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, triazolyl (including 1,2,3 and 1,3,4 triazolyls), tetrazolyl, thiadiazolyl (including 1,2,3 and 1,3,4 thiadiazolyls), and dithiazolyl. Examples of 6-membered monocyclic N-heterocycles include pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and triazinyl. The heterocycles may be optionally substituted with a broad range of substituents, and preferably with Ci- 6 alkyl, Ci- 6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, halo, hydroxy, mercapto, trifluoromethyl, amino, cyano or mono or di(Ci- 6 alkyl)amino. The N-heterocyclic group may be fused to a carbocyclic ring such as phenyl, naphthyl, indenyl, azulenyl, fluorenyl, and anthracenyl.

Examples of 8, 9 and 10-membered bicyclic heterocycles include 1H thieno[2,3-c]pyrazolyl, indolyl, isoindolyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, indazolyl, isoquinolinyl, quinolinyl, quinoxalinyl, purinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, benzotriazinyl, and the like. These heterocycles may be optionally substituted, for example with Ci - 6 alkyl, Ci- 6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, halo, hydroxy, mercapto, trifluoromethyl, amino, cyano or mono or di(Ci- 6 alkyl) amino. Unless otherwise defined optionally substituted N-heterocyclics includes pyridinium salts and the N-oxide form of suitable ring nitrogens.

Examples of N-heterocycles also include bridged groups such as, for example, azabicyclo (for example, azabicyclooctane).

“Stereoisomers” are isomers that have the same molecular formula and sequence of bonded atoms, but which differ only in the three-dimensional orientation of the atoms in space. By convention, bold wedge bonds are used to indicate bonds coming out of the page toward the reader, and hashed wedge bonds are used to indicate bonds going behind the page away from the reader. Pairs of bold and hashed bonds that are not wedged are used to indicate bonds of the same orientation, i.e., a pair of bonds that are both coming out of the page or going behind the page.

“Pharmaceutical compositions” are compositions that include an amount (for example, a unit dosage) of one or more of the disclosed compounds together with one or more non-toxic pharmaceutically acceptable additives, including carriers, diluents, and/or adjuvants, and optionally other biologically active ingredients. Such pharmaceutical compositions can be prepared by standard pharmaceutical formulation techniques such as those disclosed in Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, PA (19th Edition).

The terms "pharmaceutically acceptable salt or ester" refers to salts or esters prepared by conventional means that include salts, e.g., of inorganic and organic acids, including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, fumaric acid, succinic acid, maleic acid, salicylic acid, benzoic acid, phenylacetic acid, mandelic acid and the like. "Pharmaceutically acceptable salts" of the presently disclosed compounds also include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, diethylamine, piperazine,

tris(hydroxymethyl)aminomethane, and tetramethylammonium hydroxide. These salts may be prepared by standard procedures, for example by reacting the free acid with a suitable organic or inorganic base. Any chemical compound recited in this specification may alternatively be administered as a pharmaceutically acceptable salt thereof. “Pharmaceutically acceptable salts” are also inclusive of the free acid, base, and zwitterionic forms. Descriptions of suitable pharmaceutically acceptable salts can be found in Handbook of Pharmaceutical Salts, Properties, Selection and Use, Wiley VCH (2002). When compounds disclosed herein include an acidic function such as a carboxy group, then suitable pharmaceutically acceptable cation pairs for the carboxy group are well known to those skilled in the art and include alkaline, alkaline earth, ammonium, quaternary ammonium cations and the like. Such salts are known to those of skill in the art.

For additional examples of "pharmacologically acceptable salts," see Berge et ah, J. Pharm. Set 66: 1 (1977). “Pharmaceutically acceptable esters” includes those derived from compounds described herein that are modified to include a carboxyl group. An in vivo hydrolysable ester is an ester which is hydrolysed in the human or animal body to produce the parent acid or alcohol. Representative esters thus include carboxylic acid esters in which the non-carbonyl moiety of the carboxylic acid portion of the ester grouping is selected from straight or branched chain alkyl (for example, methyl, n-propyl, t-butyl, or n-butyl), cycloalkyl, alkoxyalkyl (for example, methoxymethyl), arylalkyl (for example benzyl), aryloxyalkyl (for example, phenoxymethyl), aryl (for example, phenyl, optionally substituted by, for example, halogen, C M alkyl, or C M alkoxy) or amino); sulphonate esters, such as alkyl- or arylalkylsulphonyl (for example, methanesulphonyl); or amino acid esters (for example, L-valyl or L-isoleucyl). A“pharmaceutically acceptable ester” also includes inorganic esters such as mono-, di-, or tri-phosphate esters. In such esters, unless otherwise specified, any alkyl moiety present advantageously contains from 1 to 18 carbon atoms, particularly from 1 to 6 carbon atoms, more particularly from 1 to 4 carbon atoms. Any cycloalkyl moiety present in such esters advantageously contains from 3 to 6 carbon atoms. Any aryl moiety present in such esters advantageously comprises a phenyl group, optionally substituted as shown in the definition of carbocycylyl above. Pharmaceutically acceptable esters thus include C1-C22 fatty acid esters, such as acetyl, t-butyl or long chain straight or branched unsaturated or omega-6 monounsaturated fatty acids such as palmoyl, stearoyl and the like. Alternative aryl or heteroaryl esters include benzoyl, pyridylmethyloyl and the like any of which may be substituted, as defined in carbocyclyl above. Additional pharmaceutically acceptable esters include aliphatic L-amino acid esters such as leucyl, isoleucyl and especially valyl.

For therapeutic use, salts of the compounds are those wherein the counter-ion is pharmaceutically acceptable. However, salts of acids and bases which are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.

The pharmaceutically acceptable acid and base addition salts as mentioned hereinabove are meant to comprise the therapeutically active non-toxic acid and base addition salt forms which the compounds are able to form. The pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid. Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e.

ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic (i.e. hydroxybutanedioic acid), tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p- aminosalicylic, pamoic and the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.

The compounds containing an acidic proton may also be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases. Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. the benzathine, N- methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.

The term“addition salt” as used hereinabove also comprises the solvates which the compounds described herein are able to form. Such solvates are for example hydrates, alcoholates and the like.

The term "quaternary amine" as used hereinbefore defines the quaternary ammonium salts which the compounds are able to form by reaction between a basic nitrogen of a compound and an appropriate quaternizing agent, such as, for example, an optionally substituted alkylhalide, arylhalide or arylalkylhalide, e.g. methyliodide or benzyliodide. Other reactants with good leaving groups may also be used, such as alkyl trifluoromethanesulfonates, alkyl methanesulfonates, and alkyl p-toluenesulfonates. A quaternary amine has a positively charged nitrogen. Pharmaceutically acceptable counterions include chloro, bromo, iodo, trifluoro acetate and acetate. The counterion of choice can be introduced using ion exchange resins.

It will be appreciated that the compounds described herein may have metal binding, chelating, complex forming properties and therefore may exist as metal complexes or metal chelates.

Some of the compounds described herein may also exist in their tautomeric form.

The term "subject" includes both human and veterinary subjects.

A "therapeutically effective amount" or "diagnostically effective amount" refers to a quantity of a specified agent sufficient to achieve a desired effect in a subject being treated with that agent. Ideally, a therapeutically effective amount or diagnostically effective amount of an agent is an amount sufficient to inhibit or treat the disease without causing a substantial cytotoxic effect in the subject. The therapeutically effective amount or diagnostically effective amount of an agent will be dependent on the subject being treated, the severity of the affliction, and the manner of administration of the therapeutic composition.

“Treatment” refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition after it has begun to develop. As used herein, the term“ameliorating,” with reference to a disease or pathological condition, refers to any observable beneficial effect of the treatment. The beneficial effect can be evidenced, for example, by a delayed onset of clinical symptoms of the disease in a susceptible subject, a reduction in severity of some or all clinical symptoms of the disease, a slower progression of the disease, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease. The phrase“treating a disease” is inclusive of inhibiting the full development of a disease or condition, for example, in a subject who is at risk for a disease, or who has a disease, such as cancer or a disease associated with a compromised immune system. “Preventing” a disease or condition refers to prophylactic administering a composition to a subject who does not exhibit signs of a disease or exhibits only early signs for the purpose of decreasing the risk of developing a pathology or condition, or diminishing the severity of a pathology or condition.

Prodmgs of the disclosed compounds also are contemplated herein. A prodrug is an active or inactive compound that is modified chemically through in vivo physiological action, such as hydrolysis, metabolism and the like, into an active compound following administration of the prodrug to a subject. The term "prodrug" as used throughout this text means the pharmacologically acceptable derivatives such as esters, amides and phosphates, such that the resulting in vivo biotransformation product of the derivative is the active drug as defined in the compounds described herein. Prodrugs preferably have excellent aqueous solubility, increased bioavailability and are readily metabolized into the active inhibitors in vivo. Prodmgs of a compounds described herein may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either by routine manipulation or in vivo, to the parent compound. The suitability and techniques involved in making and using prodrugs are well known by those skilled in the art. For a general discussion of prodrugs involving esters see Svensson and Tunek, Drug Metabolism Reviews 165 (1988) and Bundgaard, Design of Prodrugs, Elsevier (1985).

The term "prodrug" also is intended to include any covalently bonded carriers that release an active parent drug of the present invention in vivo when the prodrug is administered to a subject. Since prodmgs often have enhanced properties relative to the active agent pharmaceutical, such as, solubility and bioavailability, the compounds disclosed herein can be delivered in prodrug form. Thus, also contemplated are prodmgs of the presently disclosed compounds, methods of delivering prodmgs and compositions containing such prodmgs. Prodmgs of the disclosed compounds typically are prepared by modifying one or more functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to yield the parent compound. Prodmgs include compounds having a phosphonate and/or amino group functionalized with any group that is cleaved in vivo to yield the corresponding amino and/or phosphonate group, respectively. Examples of prodmgs include, without limitation, compounds having an acylated amino group and/or a phosphonate ester or phosphonate amide group. In particular examples, a prodrug is a lower alkyl phosphonate ester, such as an isopropyl phosphonate ester.

Protected derivatives of the disclosed compounds also are contemplated. A variety of suitable protecting groups for use with the disclosed compounds are disclosed in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed.; John Wiley & Sons, New York, 1999.

In general, protecting groups are removed under conditions which will not affect the remaining portion of the molecule. These methods are well known in the art and include acid hydrolysis, hydrogenolysis and the like. One preferred method involves the removal of an ester, such as cleavage of a phosphonate ester using Lewis acidic conditions, such as in TMS-Br mediated ester cleavage to yield the free phosphonate. A second preferred method involves removal of a protecting group, such as removal of a benzyl group by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof. A t-butoxy-based group, including t-butoxy carbonyl protecting groups can be removed utilizing an inorganic or organic acid, such as HC1 or trifluoroacetic acid, in a suitable solvent system, such as water, dioxane and/or methylene chloride. Another exemplary protecting group, suitable for protecting amino and hydroxy functions amino is trityl. Other conventional protecting groups are known and suitable protecting groups can be selected by those of skill in the art in consultation with Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed.; John Wiley & Sons, New York, 1999. When an amine is deprotected, the resulting salt can readily be neutralized to yield the free amine. Similarly, when an acid moiety, such as a phosphonic acid moiety is unveiled, the compound may be isolated as the acid compound or as a salt thereof.

Particular examples of the presently disclosed compounds include one or more asymmetric centers; thus these compounds can exist in different stereoisomeric forms. Accordingly, compounds and compositions may be provided as individual pure enantiomers or as stereoisomeric mixtures, including racemic mixtures. In certain embodiments the compounds disclosed herein are synthesized in or are purified to be in substantially enantiopure form, such as in a 90% enantiomeric excess, a 95% enantiomeric excess, a 97% enantiomeric excess or even in greater than a 99% enantiomeric excess, such as in enantiopure form.

Groups which are substituted (e.g. substituted alkyl), may in some embodiments be substituted with a group which is substituted (e.g. substituted aryl). In some embodiments, the number of substituted groups linked together is limited to two (e.g. substituted alkyl is substituted with substituted aryl, wherein the substituent present on the aryl is not further substituted). In some embodiments, a substituted group is not substituted with another substituted group (e.g. substituted alkyl is substituted with unsubstituted aryl).

Overview

CRPC is responsible for all prostate cancer deaths, and eventually all prostate cancer will develop into CRPC. The current best treatment for CRPC is MDV3100 (enzalutamide), which binds to androgen receptor. It is effective against a number of androgen-dependent prostate cancer cell lines. However, it is ineffective against the androgen-dependent prostate cancer cell line 22Rvl. Compounds disclosed herein are effective against all androgen-dependent cell lines tested including 22Rvl, a promising and unique property.

Several of the compounds show sub-micromolar inhibition of PS A-luciferase expression in C4-2 cells. Further, cell proliferation in androgen-dependent cell lines is significantly decreased while proliferation in androgen-independent cell lines is unaffected.

Agents

Disclosed herein are agents that can be used for treating prostate cancer, particularly castration- resistant prostate cancer. The agents may inhibit AR nuclear localization and/or reduce AR levels in castration-resistant prostate cancer.

In one embodiment, the agent is a compound, or a pharmaceutically acceptable salt or ester thereof, having a formula I of:

R 20 - (Z) b - (Y) c - (R 21 ) a - (X) d - R 22 - R 23

wherein R 20 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, a thio-containing group, a seleno-containing group, halide, or a nitro-containing group;

Z is alkanediyl, substituted alkanediyl, cycloalkanediyl, or substituted cycloalkanediyl;

Y is S, O, S(=0), -S(=0)(=0)-, or NR 10 , wherein R 10 is H or alkyl (preferably methyl); R 21 is alkanediyl, substituted alkanediyl, cycloalkanediyl, substituted cycloalkanediyl, alkadienyl, substituted alkadienyl, cycloalkenediyl, substituted cycloalkenediyl, alkatrienyl, or substituted alkatrienyl;

X is -C(=0)-, -S(=0)(=0)-, or -N(H)C(=0)-;

R 22 is a moiety that includes at least one divalent amino radical;

R 23 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, amino, a thio-containing group, or a seleno- containing group;

a is 0 or 1;

b is 0 or 1;

c is 0 or 1 ; and

d is 0 or 1.

In certain embodiments, the compound, or a stereoisomer, pharmaceutically acceptable salt, or ester thereof, has a formula according to any one of formulas IV-XVII:

formula VI, formula VII,

formula VIII, formula IX,

formula XII, formula XIII,

formula XVI, or formula XVII, wherein R 20 is phenyl substituted with C 1 -C 3 perfluoroalkyl, halo, or pentafluorosulfanyl; R 24 -R 27 independently are hydrogen, deuterium, or halo; R 28 is O, N(CI¾), or CFh; R 29 is N, O, or S; R 30 is CH or N; each R 31 independently is C 1 -C 3 alkyl, C 1 -C 3 perfluoroalkyl, halo, pentafluorosulfanyl, -C(0)0alkyl, or

C(0)N(H)alkyl; and q is 1, 2, or 3. In some embodiments, q is not 1 and/or R 20 is not

In any or all of the above embodiments, R 20 may be phenyl substituted with -CF3, -SF5, or -F. In some embodiments, R 20 is substituted at the C3 or C4 position.

In any or all of the above embodiments, each R 31 independently may be C1-C3 alkyl, C1-C3 perfluoroalkyl, or halo. In some embodiments, each R 31 independently is methyl, trifluoromethyl, or chloro. In any of the foregoing embodiments, q may be 2, and the R 31 substituents are para to one another. In certain embodiments, the compound is:

Other illustrative compounds are shown in Figs. 1A-1I. With respect to Figs. 1A-1I, each R independently is C1-C3 perfluoroalkyl, halo, pentafluorosulfanyl, -C(0)0alkyl, or C(0)N(H) alkyl.

Fig. 2 shows a synthesis of a parent structure that is amenable to the modifications lined out in a zone model. Isoxazole 2a can be obtained from the chloromethylation of 3,5-dimethylisoxazole, or via the corresponding alcohol, and can be converted to thiol 2b. In situ alkylation of 2b with chloride 2d under the basic conditions of thiolate formation leads to 1. There are many methods known for pyridazine synthesis, and the preparation of 2c can follow one of these methods, for example starting with the aniline. Acylation of 2c with chloroacetyl chloride provides 2d. Fig. 3 shows zones of modification for compound 1. The building blocks for zones 1 and 4 have been selected to cover a large range of chemical diversity; in addition, they are commercially available and are therefore readily funneled into the segment-based synthesis plan. Zone 2 contains a few diamines that preserve the distance between zone 1 and zone 3, i.e. where the nitrogens are appropriately spaced, but this zone can also be contracted to a simple nitrogen linker in order to probe the need to maintain the overall distance and orientation between zone 1 and zone 4. Zone 3 contains another spacer functionality, but the amide carbonyl group might also be involved in specific interactions with the binding site on the protein. Therefore, the distance between the carboxyl function and the halide electrophile can be varied, and the carbonyl group can also be replaced by a sulfonyl function. Pharmaceutical Compositions and Method of Use

The agents disclosed herein may be administered to a subject for treating prostate cancer, particularly castration-resistant prostate cancer. In certain embodiments a subject is identified as having castration-resistant prostate cancer that may be responsive to the agents disclosed herein. For example, patients that are offered any form of androgen deprivation therapy or anti-androgen therapy, including treatment with abiraterone or MDV3100, for the management of prostate cancer would be candidates for treatment with the agents disclosed herein.

Administration of the agent may reduce the nuclear level of androgen receptor in castration-resistant prostate cancer (CRPC) cells relative to the untreated control CRPC cells. Reducing nuclear androgen receptor levels is expected to inhibit its activation. Reduction of androgen receptor activation can be determined via measuring androgen-responsive genes, such as prostate-specific antigen (PSA).

In certain embodiments, the agent may be co- administered with another therapeutic agent such as, for example, an immunostimulant, an anti-cancer agent, an antibiotic, or a combination thereof. In particular, the agents targeting AR nuclear localization could be used in combination with standard androgen deprivation therapy (ADT) or with abiratrone in the treatment of CRPC. In one embodiment, the agent is co- administered with MDV3100 (enzalutamide), which may produce synergistic results since MDV3100 targets the ligand binding domain whereas the agent targets other domain(s) of the androgen receptor.

The agents disclosed herein can be included in a pharmaceutical composition for administration to a subject. The pharmaceutical compositions for administration to a subject can include at least one further pharmaceutically acceptable additive such as carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice. Pharmaceutical compositions can also include one or more additional active ingredients such as antimicrobial agents, anti-inflammatory agents, anesthetics, and the like. The pharmaceutically acceptable carriers useful for these formulations are conventional. Remington’s Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition (1995), describes compositions and formulations suitable for pharmaceutical delivery of the compounds herein disclosed.

The pharmaceutical compositions may be in a dosage unit form such as an injectable fluid, an oral delivery fluid (e.g., a solution or suspension), a nasal delivery fluid (e.g., for delivery as an aerosol or vapor), a semisolid form (e.g., a topical cream), or a solid form such as powder, pill, tablet, or capsule forms.

In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations usually contain injectable fluids that include

pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. For solid compositions (for example, powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.

The agents disclosed herein can be administered to subjects by a variety of mucosal administration modes, including by oral, rectal, intranasal, intrapulmonary, or transdermal delivery, or by topical delivery to other surfaces. Optionally, the agents can be administered by non-mucosal routes, including by intramuscular, subcutaneous, intravenous, intra-arterial, intra-articular, intraperitoneal, intrathecal, intracerebroventricular, or parenteral routes. In other alternative embodiments, the agents can be administered ex vivo by direct exposure to cells, tissues or organs originating from a subject.

To formulate the pharmaceutical compositions, the agents can be combined with various pharmaceutically acceptable additives, as well as a base or vehicle for dispersion of the compound. Desired additives include, but are not limited to, pH control agents, such as arginine, sodium hydroxide, glycine, hydrochloric acid, citric acid, and the like. In addition, local anesthetics (for example, benzyl alcohol), isotonizing agents (for example, sodium chloride, mannitol, sorbitol), adsorption inhibitors (for example, Tween 80 or Miglyol 812), solubility enhancing agents (for example, cyclodextrins and derivatives thereof), stabilizers (for example, serum albumin), and reducing agents (for example, glutathione) can be included. Adjuvants, such as aluminum hydroxide (for example, Amphogel, Wyeth Laboratories, Madison, NJ), Freund’s adjuvant, MPL™ (3-O-deacylated monophosphoryl lipid A; Corixa, Hamilton, IN) and IL-12 (Genetics Institute, Cambridge, MA), among many other suitable adjuvants well known in the art, can be included in the compositions. When the composition is a liquid, the tonicity of the formulation, as measured with reference to the tonicity of 0.9% (w/v) physiological saline solution taken as unity, is typically adjusted to a value at which no substantial, irreversible tissue damage will be induced at the site of administration. Generally, the tonicity of the solution is adjusted to a value of about 0.3 to about 3.0, such as about 0.5 to about 2.0, or about 0.8 to about 1.7.

The agents can be dispersed in a base or vehicle, which can include a hydrophilic compound having a capacity to disperse the compound, and any desired additives. The base can be selected from a wide range of suitable compounds, including but not limited to, copolymers of polycarboxylic acids or salts thereof, carboxylic anhydrides (for example, maleic anhydride) with other monomers (for example, methyl

(meth) acrylate, acrylic acid and the like), hydrophilic vinyl polymers, such as polyvinyl acetate, polyvinyl alcohol, polyvinylpyrrolidone, cellulose derivatives, such as hydroxymethylcellulose,

hydroxypropylcellulose and the like, and natural polymers, such as chitosan, collagen, sodium alginate, gelatin, hyaluronic acid, and nontoxic metal salts thereof. Often, a biodegradable polymer is selected as a base or vehicle, for example, polylactic acid, poly(lactic acid-glycolic acid) copolymer, polyhydroxybutyric acid, poly(hydroxybutyric acid-glycolic acid) copolymer and mixtures thereof. Alternatively or additionally, synthetic fatty acid esters such as polyglycerin fatty acid esters, sucrose fatty acid esters and the like can be employed as vehicles. Hydrophilic polymers and other vehicles can be used alone or in combination, and enhanced structural integrity can be imparted to the vehicle by partial crystallization, ionic bonding, cross- linking and the like. The vehicle can be provided in a variety of forms, including fluid or viscous solutions, gels, pastes, powders, microspheres and films for direct application to a mucosal surface.

The agents can be combined with the base or vehicle according to a variety of methods, and release of the agents can be by diffusion, disintegration of the vehicle, or associated formation of water channels. In some circumstances, the agent is dispersed in microcapsules (microspheres) or nanocapsules (nanospheres) prepared from a suitable polymer, for example, isobutyl 2-cyanoacrylate (see, for example, Michael et ah, J. Pharmacy Pharmacol. 43: 1-5, 1991), and dispersed in a biocompatible dispersing medium, which yields sustained delivery and biological activity over a protracted time.

The compositions of the disclosure can alternatively contain as pharmaceutically acceptable vehicles substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, and triethanolamine oleate. For solid compositions, conventional nontoxic pharmaceutically acceptable vehicles can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.

Pharmaceutical compositions for administering the agents can also be formulated as a solution, microemulsion, or other ordered structure suitable for high concentration of active ingredients. The vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), and suitable mixtures thereof. Proper fluidity for solutions can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of a desired particle size in the case of dispersible formulations, and by the use of surfactants.

In many cases, it will be desirable to include isotonic agents, for example, sugars, polyalcohols, such as mannitol and sorbitol, or sodium chloride in the composition. Prolonged absorption of the compound can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin.

In certain embodiments, the agents can be administered in a time release formulation, for example in a composition which includes a slow release polymer. These compositions can be prepared with vehicles that will protect against rapid release, for example a controlled release vehicle such as a polymer, microencapsulated delivery system or bioadhesive gel. Prolonged delivery in various compositions of the disclosure can be brought about by including in the composition agents that delay absorption, for example, aluminum monostearate hydrogels and gelatin. When controlled release formulations are desired, controlled release binders suitable for use in accordance with the disclosure include any biocompatible controlled release material which is inert to the active agent and which is capable of incorporating the compound and/or other biologically active agent. Numerous such materials are known in the art. Useful controlled-release binders are materials that are metabolized slowly under physiological conditions following their delivery (for example, at a mucosal surface, or in the presence of bodily fluids). Appropriate binders include, but are not limited to, biocompatible polymers and copolymers well known in the art for use in sustained release formulations. Such biocompatible compounds are non-toxic and inert to surrounding tissues, and do not trigger significant adverse side effects, such as nasal irritation, immune response, inflammation, or the like. They are metabolized into metabolic products that are also biocompatible and easily eliminated from the body.

Exemplary polymeric materials for use in the present disclosure include, but are not limited to, polymeric matrices derived from copolymeric and homopolymeric polyesters having hydrolyzable ester linkages. A number of these are known in the art to be biodegradable and to lead to degradation products having no or low toxicity. Exemplary polymers include polyglycolic acids and polylactic acids, poly(DL- lactic acid-co-glycolic acid), poly(D-lactic acid-co-glycolic acid), and poly(L-lactic acid-co-glycolic acid). Other useful biodegradable or bioerodable polymers include, but are not limited to, such polymers as poly(epsilon-caprolactone), poly(epsilon-caprolactone-CO-lactic acid), poly(epsilon.-caprolactone-CO- glycolic acid), poly(beta-hydroxy butyric acid), poly(alkyl-2-cyanoacrilate), hydrogels, such as

poly(hydroxyethyl methacrylate), polyamides, poly(amino acids) (for example, L-leucine, glutamic acid, L- aspartic acid and the like), poly(ester urea), poly(2-hydroxyethyl DL-aspartamide), polyacetal polymers, polyorthoesters, polycarbonate, polymaleamides, polysaccharides, and copolymers thereof. Many methods for preparing such formulations are well known to those skilled in the art (see, for example, Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978). Other useful formulations include controlled-release microcapsules (U.S. Patent Nos. 4,652,441 and 4,917,893), lactic acid-glycolic acid copolymers useful in making microcapsules and other formulations (U.S. Patent Nos. 4,677,191 and 4,728,721) and sustained-release compositions for water-soluble peptides (U.S. Patent No. 4,675,189).

The pharmaceutical compositions of the disclosure typically are sterile and stable under conditions of manufacture, storage and use. Sterile solutions can be prepared by incorporating the compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the compound and/or other biologically active agent into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated herein. In the case of sterile powders, methods of preparation include vacuum drying and freeze-drying which yields a powder of the compound plus any additional desired ingredient from a previously sterile-filtered solution thereof. The prevention of the action of microorganisms can be accomplished by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.

In accordance with the various treatment methods of the disclosure, the agent can be delivered to a subject in a manner consistent with conventional methodologies associated with management of the disorder for which treatment or prevention is sought. In accordance with the disclosure herein, a prophylactically or therapeutically effective amount of the agent is administered to a subject in need of such treatment for a time and under conditions sufficient to prevent, inhibit, and/or ameliorate a selected disease or condition or one or more symptom(s) thereof. The administration of the agent can be for either prophylactic or therapeutic purpose. When provided prophylactically, the agent is provided in advance of any symptom. The prophylactic

administration of the agents serves to prevent or ameliorate any subsequent disease process. When provided therapeutically, the compound is provided at (or shortly after) the onset of a symptom of disease or infection.

For prophylactic and therapeutic purposes, the agent can be administered to the subject by the oral route or in a single bolus delivery, via continuous delivery (for example, continuous transdermal, mucosal or intravenous delivery) over an extended time period, or in a repeated administration protocol (for example, by an hourly, daily or weekly, repeated administration protocol). The therapeutically effective dosage of the agent can be provided as repeated doses within a prolonged prophylaxis or treatment regimen that will yield clinically significant results to alleviate one or more symptoms or detectable conditions associated with a targeted disease or condition as set forth herein. Determination of effective dosages in this context is typically based on animal model studies followed up by human clinical trials and is guided by administration protocols that significantly reduce the occurrence or severity of targeted disease symptoms or conditions in the subject. Suitable models in this regard include, for example, murine, rat, avian, porcine, feline, non human primate, and other accepted animal model subjects known in the art. Alternatively, effective dosages can be determined using in vitro models. Using such models, only ordinary calculations and adjustments are required to determine an appropriate concentration and dose to administer a therapeutically effective amount of the compound (for example, amounts that are effective to elicit a desired immune response or alleviate one or more symptoms of a targeted disease). In alternative embodiments, an effective amount or effective dose of the agents may simply inhibit or enhance one or more selected biological activities correlated with a disease or condition, as set forth herein, for either therapeutic or diagnostic purposes.

The actual dosage of the agents will vary according to factors such as the disease indication and particular status of the subject (for example, the subject’s age, size, fitness, extent of symptoms, susceptibility factors, and the like), time and route of administration, other drugs or treatments being administered concurrently, as well as the specific pharmacology of the agent for eliciting the desired activity or biological response in the subject. Dosage regimens can be adjusted to provide an optimum prophylactic or therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental side effects of the agent is outweighed in clinical terms by therapeutically beneficial effects. A non-limiting range for a therapeutically effective amount of an agent within the methods and formulations of the disclosure is about 0.01 mg/kg body weight to about 20 mg/kg body weight, such as about 0.05 mg/kg to about 5 mg/kg body weight, or about 0.2 mg/kg to about 2 mg/kg body weight. Dosage can be varied by the attending clinician to maintain a desired concentration at a target site (for example, the lungs or systemic circulation). Higher or lower concentrations can be selected based on the mode of delivery, for example, trans-epidermal, rectal, oral, pulmonary, or intranasal delivery versus intravenous or subcutaneous delivery. Dosage can also be adjusted based on the release rate of the administered formulation, for example, of an intrapulmonary spray versus powder, sustained release oral versus injected particulate or transdermal delivery formulations, and so forth. Examples

1. Biological Materials and Methods

Materials

Phosphate buffered saline (PBS) solution was purchased from Fisher Scientific (MA, USA).

Trypsin-EDTA solution, dimethyl sulfoxide (DMSO), Roswell Park Memorial Institute (RPMI) 1640 medium, ethanol (200 proof), puromycin powder, and G418 powder were purchased from Sigma-Aldrich (MO, USA). Fetal bovine Serum (FBS), penicillin-streptomycin solution were purchased from Invitrogen (NY, USA). Dual-Luciferase® Reporter Assay System was purchased from Promega (WI, USA). PSA6.1— luc plasmid was a gift from Dr. Marianne Sadar at the University of British Columbia (BC, CA) and pRL- TK Renilla luciferase reporter plasmid was purchased from Promega (WI, USA). The C4-2 castration- resistant prostate cancer cell line was kindly provided by Dr. Leland W.K. Chung (Cedars-Sinai Medical Center).

2. Chemistry

General

Moisture and air-sensitive reactions were performed under N2 or Ar atmosphere and glassware used for these reactions was flamed dried and cooled under N2 or Ar prior to use. THF and Et 2 0 were distilled from sodium/benzophenone ketyl. DMF and CH2CI2 were distilled from CaFE. 1,4-Dioxane was purchased from Acros (Sure/Seal bottle) and used as received. EhN was distilled from Ca¾ and stored over KOH. Toluene was purified by passage through an activated alumina filtration system. Melting points were determined using a Mel-Temp II instrument and are not corrected. Infrared spectra were determined using a Smiths Detection Identify IR FT-IR spectrometer. High-resolution mass spectra were obtained on a

Micromass UK Limited, Q-TOF Ultima API, Thermo Scientific Exactive Orbitrap LC-MS. Automated column chromatography was done using an Isco Combiflash Rf. H and 13 C NMR spectra were obtained on Bruker Advance 300 MHz, 400 MHz, or 500 MHz instruments. Chemical shifts (d) were reported in parts per million with the residual solvent peak used as an internal standard, d H/ I3 C (Solvent): 7.26/77.00 (CDCE); 2.05/29.84 (acetone-d6); 2.50/39.52 (DMSO-d6), 3.31/49.00 (CD 3 OD); and are tabulated as follows: chemical shift, multiplicity (s = singlet, brs = broad singlet, d = doublet, brd = broad doublet, t = triplet, app t = apparent triplet, q = quartet, m = multiplet), number of protons, and coupling constant(s). 13 C NMR spectra were obtained at 75 MHz, 100 MHz, or 125 MHz using a proton-decoupled pulse sequence and are tabulated by observed peak. CDCE was filtered through dried basic alumina prior to use. Thin-layer chromatography was performed using pre-coated silica gel 60 F254 plates (EMD, 250 pm thickness) and visualization was accomplished with a 254 nm UV light and by staining with a PM A solution (5 g of phosphomolybdic acid in 100 mL of 95% EtOH), Vaughn’s reagent (4.8 g of (NH 1 ) f ,Mo 7 0 2 i ·4¾0 and 0.2 g of Ce(S04)2 in 100 mL of a 3.5 N H2SO4 solution) or a KMnCE solution (1.5 g of KMnCE and 1.5 g of K2CO3 in 100 mL of a 0.1% NaOH solution). Chromatography on S1O2 (Silicycle, Silia-P Flash Silica Gel or SiliaFlash® P60, 40-63 pm) was used to purify crude reaction mixtures. Final products were >95% purity as analyzed by RP (reverse phase) HPLC (Alltech Prevail C-18, 100 x 4.6 mm, 1 mL/min, CPbCN, H 2 0 and 0.1% TFA) with UV (210, 220 and 254 nm), ELS (nebulizer 45 °C, evaporator 45 °C, N 2 flow 1.25 SLM), and MS detection using a Thermo Scientific Exactive Orbitrap LC-MS (ESI positive). All other materials were obtained from commercial sources and used as received.

Example 5

Synthesis and Characterization of Analogs

General:

All glassware was flame-dried or dried in an oven at 120 °C for more than two hours prior to use.

All air- and moisture-sensitive reactions were performed under N 2 or Ar atmosphere. Reactions carried out at 0 °C or -78 °C employed an ice bath or an acetone/dry ice bath. Tetrahydrofuran and diethyl ether were either distilled over sodium/benzophenone ketyl, CH 2 C1 2 and toluene were distilled from CaH 2 . All other materials were obtained from commercial sources and used as received. Infrared spectra were determined neat on a Smiths Detection Identify IR FT-IR spectrometer. H and 13 C NMR spectra were obtained on a Bruker Advance 300 MHz, 400 MHz or 500 MHz NMR in CDCE unless otherwise specified. Chemical shifts (d) were reported in parts per million, with the residual solvent peak used as an internal standard d 'H/ 13 C (Solvent); 7.26/77.00 (CDCE), 2.50/39.50 (DMSO-d6); they are tabulated as follows: chemical shift, multiplicity (s = singlet, brs = broad singlet, d = doublet, brd = broad doublet, t = triplet, brt = broad triplet, q = quartet, m = multiplet), number of protons, and coupling constant(s). 13 C NMR spectra were obtained at 75 MHz, 100 MHz or 125 MHz unless otherwise specified using a proton-decoupled pulse sequence and are tabulated by observed peak. 19 F spectra were obtained at 471MHz or 376 MHz unless otherwise specified using a proton-decoupled pulse sequence and are tabulated by observed peak. Reactions were monitored by thin-layer chromatography analysis using pre-coated silica gel 60 F254 plates (EMD, 250 pm thickness), and visualization was accomplished with a 254 nm UV light. Flash chromatography was performed using Si0 2 (Silicycle, Silia-P Flash Silica Gel, 40-63 pm).

Figs. 4-25 provide exemplary reaction schemes for several of the analogs described in detail below. Fig. 4 is a general reaction scheme for propiolic acid precursor compounds. Fig. 5 is a reaction scheme for (4-(5- chloro-2-methylphenyl)piperazin- 1 -yl)(( I A’.S , ,2.S , A > )-2-(4- fluorophenyl pcyclopropyl- 1 2-d2 )-mcthanonc. Fig.

6 shows three general reaction schemes for several precursor compounds including aryl and piperazinyl moieties. Fig. 7 is a general reaction scheme for several analogs comprising cyclopropyl and piperazinyl moieties. Fig. 8 is a reaction scheme for (4-(5-chloro-2-fluorobenzoyl)piperazin-l-yl)((15,2R)-2-(4- (trifluoromethyl)phenyl)cyclopropyl)-methanone and (4-((5-chloro-2-fluorophenyl)-sulfonyl)piperazin-l- yl)((lS,2R)-2-(4-(trifluoromethyl)phenyl)-cyclopropyl)methan one. Fig. 9 is a reaction scheme for (4-(5- chloro-2-fluorophenyl)piperazin-l-yl)(2-(4-fluorophenyl)cycl opropyl)-methanone. Fig. 10 is a reaction scheme for (4-(5-chloro-2-methylphenyl)piperazin-l-yl)(2-(4-(trifluorom ethyl)phenyl)- cyclopropyl)methanone. Fig. 11 is a reaction scheme for (4-(2-methyl-5-(trifluoromethyl)phenyl)piperazin- l-yl)(2-(4-(trifluoromethyl)phenyl)cyclopropyl)-methanone. Fig. 12 is a reaction scheme for 3-fluoro-2-(4- (2-(4-(trifluoromethyl)phenyl)cyclopropane-l-carbonyl)pipera zin-l-yl)benzonitrile. Fig. 13 is a reaction scheme for (4-(2-chloro-5-(trifluoromethyl)phenyl)piperazin- 1 -yl)(2-(4-(trifluoromethyl)phenyl)- cyclopropyl)methanone. Fig. 14 is a reaction scheme for (4-cyclohexyl-piperazin-l-yl)(2-(4- (trifluoromethyl)phenyl)cyclopropyl)methanone and (4-(Tetrahydro-2H-pyran-4-yl)piperazin- 1 -yl)(2-(4- (trifluoromethyl)phenyl)cyclopropyl)methanone. Fig. 15 is a reaction scheme for ethyl 2-fluoro-2-(4- fluorophenyl)cyclopropane-l-carboxylate. Fig. 16 is a reaction scheme for (4-(2-methyl-5- (trifluoromethyl)phenyl)piperazin-l-yl)(4-(4-(trifluoromethy l)phenyl)oxetan-2-yl)methanone. Fig. 17 is a reaction scheme for r/Y//rv-(4-(5-chloro-2-mcthylphcnyl jpipera/.in- 1 -yl )(( I SR,2RS)-2-fluoro-2-(4- fluorophenyl)-cyclopropyl)methanone. Fig. 18 is a reaction scheme for r/v-(4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lSR,2RS)-2-fluoro-2-(4-fluorop henyl)cyclopropyl)-methanone. Fig. 19 is a reaction scheme for cis- and r/Y//rv-(4-(5-chloro-2-mcthylphcnyl )piperazin- 1 -yl )(2-(4-fluorophcnyl )- 1 - (trifluoromethyl)-cyclopropyl)methanone. Fig. 20 is a reaction scheme for r/Y//iv-(4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lRS,2RS)-2-(4-fluorophenyl)-2- (trifluoromethyl)cyclopropyl)-methanone. Fig. 21 is a reaction scheme for i/Y//iv-(4-(2,5-his(trifluoromcthyl )phenyl )-piperazin- 1 -yl )( I - (trifluoromethyl)-2-(4-(trifluoromethyl)-phenyl)cyclopropyl) methanone. Fig. 22 is a reaction scheme for cA-((lSR,3RS)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cycl opropyl)(4-(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)methanone. Fig. 23 is a reaction scheme for (4-(5-chloro-2-(trifluoromethyl)- phenyl)piperazin-l-yl)(3-(4-fluorophenyl)bicyclo[1.1.0]butan -l-yl)methanone. Fig. 24 is a reaction scheme for (4-(5-chloro-2-(trifluoromethyl)phenyl)piperazin-l-yl)(3-(4- (trifluoromethyl)phenyl)bicyclo[1.1.0]butan- l-yl)methanone. Fig. 25 is a reaction scheme for (4-(2-methyl-5-(trifluoromethyl)-phenyl)piperazin-l-yl)(3- (4-(trifluoromethyl)phenyl)cyclobutyl)-methanone.

3-(3-(Trifluoromethyl)phenyl)propiolic acid (Yonemoto-Kobayashi et al, Org. & Biomolec. Chem. 2013, 11 :3773-3775; Solomon et al, JACS 1963, 85:3492-3496; Austin et al, J. Org. Chem. 1981, 46:2280-2286. A solution of Pd(PPli3)2Cl2 (0.0134 g, 0.0436 mmol), Cul (0.00829 g, 0.0436 mmol), and 3-bromobenzo- triflouride (0.62 mL, 4.36 mol) in EtiN (8.7 mL) was sparged with Ar for 15 min and treated with

(trimethylsilyl) acetylene (0.93 mL, 6.53 mmol) and sparged for an additional 2 min. The resulting mixture was heated to 80 °C overnight, cooled to rt, filtered through Celite, washed (Et 2 0) until the washes appeared colorless and the filtrate was concentrated under reduced pressure. The crude residue was purified by chromatography on S1O2 (hexanes) to give the trimethyl((3-(trifluoromethyl)phenyl)ethynyl)silane (1.03 g,

4.24 mmol, 97 %) as a pale yellow oil.

A solution of CsF (0.775 g, 5.10 mmol) in dry DMSO (6.5 mL) under an atmosphere of CO2 (balloon) at rt was treated with a solution of trimethyl((3-(trifluoromethyl)phenyl)ethynyl)silane (1.03 g,

4.25 mmol) dropwise and the reaction was stirred under CO2 at rt overnight. The reaction mixture was diluted with H2O (80 mL) and extracted with CH2CI2 (2 x 25 mL). The aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and extracted with Et 2 0 (3 x 25 mL). The combined organic layers were dried (MgSOi ), concentrated under reduced pressure, and dried under high vacuum to give 3-(3- (trifluoromethyl)phenyl)propiolic acid (0.774 g, 3.62 mmol, 85%) as an pale tan orange waxy solid: H NMR (400 MHz, Acetone-d 6 ) d 11.85 (bs, 1 H), 7.95-7.87 (m, 3 H), 7.36 (t, / = 7.4 Hz, 1 H); 13 C NMR (100 MHz, Acetone-de) d 154.3, 137.1, 131.7 (q, JC F = 33.0 Hz), 130.0 (q, JC F = 3.8 Hz), 128.1 (q, JC F = 3.4 Hz), 124.5 (q, J CF = 272.0 Hz), 121.7, 83.6, 82.9.

Trimethyl((4-(pentafluoro^6-sulfaneyl)phenyl)ethynyl)silane. A solution of Pd(PPh 3 ) 2 Cl 2 (0.0365 g, 0.0519 mmol), Cul (0.0100 g, 0.0519 mmol), and (4-bromophenyl )pcntafluoro-/.6-sulfanc (1.50 g, 5.19 mmol) in EtiN (11 mL) was sparged with Ar for 10 min, treated with (trimethylsilyl)acetylene (1.10 mL,

7.79 mmol), sparged with Ar for 5 min, heated to 80 °C for 22 h, cooled to rt, filtered through Celite, washed (Et20) until the washes appeared colorless, and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by chromatography on S1O 2 (hexanes) to give trimethyl((4- (pentafluoro- 6-sulfaneyl)phenyl)ethynyl)silane (1.32 g, 4.40 mmol, 85 %) as a pale yellow oil: IR (CH 2 CI 2 ) 2963, 2165, 1599, 1493, 1401, 1251, 1095, 826, 802, 759 cm 1 ; Ή NMR (400 MHz, CDCE) d 7.68 (dt, / = 9.0, 2.0 Hz, 2 H), 7.52 (d, / = 9.0 Hz, 2 H), 0.28 (s, 9 H); 13 C NMR (100 MHz, CDC1 3 ) d 153.2 (quint, J CF = 18.0 Hz), 126.9, 125.9 (quint, JC F = 5.0 Hz), 102.7, 98.0, -0.3; 19 F NMR (376 MHz, CDCE) d 84.0 (quint, J = 150.2 Hz, 1 F), 62.6 (d, J = 150.2 Hz, 4 F); HRMS (ESI) m/z calcd for CnHi 3 F 5 SiS ([M] + ) 300.0427, found 300.0400.

3-(4-(Pentafluoro^6-sulfaneyl)phenyl)propiolic add. A solution of CsF (0.801 g, 5.27 mmol) in dry DMSO (3 mL) under CO2 (balloon) at rt was treated with a solution of tri methyl ((4-( pen tail uoro-/.6- sulfaneyl)phenyl)ethynyl)silane (1.32 g, 4.40 mmol) in DMSO (5.8 mL) dropwise and the reaction was stirred under CO2 at rt overnight, diluted with ¾0 (90 mL) and extracted with CH2CI2 (2 x 50 mL). The aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and extracted with Et 2 0 (3 x 50 mL). The combined organic layers were washed with H2O (50 mL), dried (MgSO i ), concentrated under reduced pressure, and dried under high vacuum to give 3-(4-(pentafluoro- 6-sulfaneyl)phenyl)propiolic acid (0.338 g, 1.24 mmol, 28%) as brown solid. Mp 156.5-159.6 °C; IR (CHCE) 2979, 2876, 2577, 2235, 1677, 1416, 1298, 1217, 886, 829, 751 cm 1 ; Ή NMR (400 MHz, CDCE) d 10.00 (bs, 1 H), 7.82 (dt, / = 9.0, 2.0 Hz, 2 H), 7.73 (d, J = 9.0 Hz, 2 H); 13 C NMR (100 MHz, CDCE) d 157.7, 155.2 (quint, JC F = 19.0 Hz), 133.3, 126.5 (quint, J CF = 4.0 Hz), 122.7, 85.9, 81.7; 19 F NMR (376 MHz, CDCE) d 82.6 (quint, J = 150.5 Hz, 1 F), 62.3 (d, / = 150.3 Hz, 4 F); HRMS (ESI) m/z calcd for C9H4O2F5S ([M-H] ) 270.9858, found 270.9858.

Tnmcthyl((3-(pcntafluoro->.6-sulfancyl)phcnyl)cthynyl)sil anc. A solution of Pd(PPh 3 )2Cl2 (0.0365 g, 0.0519 mmol), Cul (0.0100 g, 0.0519 mmol), and ( 3 - b ro m o phenyl) pc nta 11 uo ro - l6 - s u I fan c (1.50 g, 5.19 mmol) in Et 3 N (11 mL) was sparged with Ar for 10 min, treated with (trimethylsilyl)acetylene (1.10 mL, 7.79 mmol), sparged with Ar for 5 min heated to 80 °C for 22 h, cooled to rt, filtered through Celite, washed (Et 2 0) until the washes appeared colorless. The combined filtrates were concentrated under reduced pressure. The crude residue was purified by chromatography on SiCL (hexanes) to give trimethyl((3- (pcntal1uoro-/.6-sulfancyl )phcnyl )ethynyl )silane (1.29 g, 4.29 mmol, 83%) as a yellow oil: IR (CH2CI2) 2963, 2902, 2168, 1601, 1479, 1421, 1251, 1109, 831, 803, 789, 759, 683 cm 1 ; Ή NMR (400 MHz, CDC1 3 ) d 7.87 (t, J = 2.0 Hz, 1 H), 7.68 (ddd, J = 8.0, 2.0, 0.8 Hz, 1 H), 7.58 (d, / = 8.0 Hz, 1 H), 7.38 (t, J = 8.0 Hz, 1 H), 0.29 (s, 9 H); 13 C NMR (100 MHz, CDC1 3 ) d 153.7 (quint, J CF = 17.8 Hz), 134.8, 129.5 (quint, J CF = 4.6 Hz), 128.6, 125.8 (quint, J CF = 4.8 Hz), 124.4, 102.8, 96.7, -0.3; 19 F NMR (376 MHz, CDC1 3 ) d 83.6 (quint, / = 150.4 Hz, 1 F), 62.5 (d, / = 150.3 Hz, 4 F); HRMS (ESI) m/z calcd for CnHi 3 F 5 SiS ([M] + ) 300.0427, found 300.0405.

3-(3-(Pentafluoro-h6-sulfaneyl)phenyl)propiolic add. A solution of CsF (0.783 g, 5.15 mmol) in dry DMSO (3 mL) under CO2 (balloon) at rt was treated a solution of trimcthyK(3-(pcntafluoro-/.6- sulfaneyl)phenyl)ethynyl)silane (1.29 g, 4.30 mmol) in DMSO (5.6 mL) dropwise and the reaction was stirred under CO2 at rt overnight. The reaction mixture was diluted with ¾0 (90 mL) and extracted with CH2CI2 (2 x 50 mL). The aqueous layer was acidified (> pH 1) with 6M aqueous HC1 at 0 °C and then extracted with Et 2 0 (3 x 50 mL). The combined organic layers were washed with ¾0 (50 mL), dried (MgSOr), concentrated under reduced pressure, and further dried under high vacuum to give 3-(3- (pcntal1uoro-/.6-sulfancyl ) phenyl )propiolic acid (0.935 g, 3.43 mmol, 80%) as tan solid: Mp 121.1- 124.4 °C; IR (CHC1 3 ) 2831, 2218, 1688, 1479, 1426, 1214, 831, 791, 768, 680 cm 1 ; Ή NMR (400 MHz, CDCLs) d 10.92 (s, 1 H), 8.01 (t, J = 2.0 Hz, 1 H), 7.87 (ddd, J = 8.0, 2.0, 0.9 Hz, 1H), 7.75 (d, J = 8.0 Hz, 1 H), 7.54 (t, / = 8.0 Hz, 1 H); 13 C NMR (100 MHz, CDCLs) d 158.1, 153.8 (quint, J CF = 19.0 Hz), 135.9,

130.7 (quint, J CF = 5.0 Hz), 129.3, 128.4 (quint, J CF = 4.0 Hz), 120.2, 86.2, 81.0; 19 F NMR (376 MHz, CDCL) d 82.5 (quint, J = 150.4 Hz, 1 F), 62.5 (d, / = 150.4 Hz, 4 F); HRMS (ESI) m/z calcd for C 9 H 4 O 2 F5S ([M-H]-) 270.9858, found 270.9856. 3-(5-Methylthiophen-2-yl)propiolic add (He et al. , Chem. Sci. 2013, 4:3478-3483; Paegle et al, Euro. J. Org. Chem. 2015; 2015:4389-4399; Kub et al., Macromolecules 2010, 34:2124-2129). A solution of Pd(PPli3)2Cl2 (0.0269 g, 0.0877 mmol), Cul (0.0167 g, 0.0877 mmol), and 2-bromo-5-methyl thiophene (1.00 mL, 8.77 mmol) in EtiN (17.5 mL) sparged with Ar for 15 min and treated with

(trimethylsilyl) acetylene (1.9 mL, 13.2 mmol) and the mixture was further sparged for 2 min, heated to 80 °C overnight, cooled to rt, filtered through Celite, washed (Et 2 0) until the washes appeared colorless and the filtrate was concentrated under reduced pressure. The crude residue was purified by chromatography on S1O2 (hexanes) to give the trimethyl((5-methylthiophen-2-yl)ethynyl)silane 1.06 g, 5.47 mmol, 62%) as a pale yellow oil.

To a solution of CsF (0.994 g, 6.54 mmol) in dry DMSO (8 mL) under an atmosphere of CO2 (balloon) at rt was added a solution of trimethyl((5-methylthiophen-2-yl)ethynyl)silane (1.06 g, 5.45 mmol) dropwise and the reaction was stirred under CO2 at rt overnight. The reaction mixture was diluted with H2O (100 mL) and extracted with CH2CI2 (2 x 25 mL). The aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and extracted with Et 2 0 (3 x 25 mL). The combined organic layers were washed with brine (50 mL), dried (MgSOi ), filtered, and concentrated under reduced pressure to give the product (0.571 g, 3.44 mmol, 63%) as a brown solid: Ή NMR (300 MHz, CDCL) d 10.14 (bs, 1 H), 7.36 (d, / = 3.3 Hz, 1 H), 6.73 (dd, / = 3.3 Hz, 1 H), 2.52 (s, 3 H).

3-(3,5-Bis(trifluoromethyl)phenyl)propiolic add. A solution of Pd(PPli3)2Cl2 (0.239 g, 0.341 mmol), Cul (0.0650 g, 0.341 mmol), and l-bromo-3,5-bis(trifluoromethyl)benzene (2.00 g, 6.83 mmol) in EtiN (13 mL) was sparged with Ar for 10 min and treated with (trimethylsilyl) acetylene (1.42 mL, 10.2 mmol) and the solution was sparged with Ar for 2 min. The resulting mixture was heated to 80 °C for 22 h, cooled to rt, and filtered through Celite, which was washed with Et 2 0 until the washes appeared colorless. The filtrate was concentrated under reduced pressure and the crude residue was purified by chromatography on S1O2 (hexanes) to give the desired product (1.79 g, 5.78 mmol) as a light yellow solid.

A solution of CsF (1.05 g, 6.92 mmol) in DMSO (4.6 mL) under CO2 at rt was treated with a solution of ((3,5-bis(trifluoromethyl)phenyl)ethynyl)trimethylsilane (1.79 g, 5.77 mmol) in DMSO (7 mL) dropwise and the reaction was stirred under CO2 at rt overnight. The reaction mixture was diluted with ¾0 (90 mL) and extracted with CH2CI2 (2 x 50 mL). The aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and then extracted with Et 2 0 (3 x 100 mL). The combined organic layers were washed with H2O (50 mL), dried (MgSOi ). The solvent was concentrated under reduced pressure and further dried under high vacuum to give 3-(3,5-bis(trifluoromethyl)phenyl)propiolic acid (0.859 g, 3.04 mmol, 45% (2 steps)) as brown solid: Mp 124.5-128.2 °C; IR (CH2CI2) 2915, 2226, 1688, 1377, 1278, 1131, 972, 903, 683 cm 1 ; Ή NMR (400 MHz, CDC1 3 ) d 11.25 (s, 1 H), 8.06 (s, 2 H), 7.98 (s, 1 H); 13 C NMR (100 MHz, CDC1 3 ) d 157.7, 132.9 (q, J CF = 3.4 Hz), 132.6 (q, J CF = 34.4 Hz), 124.5 (q, J CF = 3.5 Hz), 122.5 (q, J CF = 273.2 Hz), 121.5, 84.5, 82.0; 19 F NMR (376 MHz, CDCF) d -63.3 (s, 6 F); HRMS (ESI) m/z calcd for CnH 3 F 6 0 2 ([M- H] ) 281.0043, found 281.0039

3-(4-Chloro-2-fluorophenyl)propiolic acid. A solution of Pd(PPli 3 ) 2 Cl 2 (0.197 g, 0.281 mmol), Cul (0.0535 g, 0.281 mmol), and 4-chloro-2-fluoro-l-iodobenzene (1.80 g, 7.02 mmol) in Et 3 N (14 mL) was sparged with Ar for 10 min followed by addition of (trimethylsilyl)acetylene (1.46 mL, 10.5 mmol) and the solution was further sparged with Ar for 2 min. The resulting mixture was heated to 80 °C for 22 h. After cooling the reaction to rt, the solution was filtered through Celite, which was washed with Et 2 0 until the washes appeared colorless. The filtrate was concentrated under reduced pressure. The crude residue was purified by chromatography on S1O2 (hexanes) to give ((4-chloro-2-fluorophenyl)ethynyl)trimethylsilane (1.42 g, 6.26 mmol) as a yellow oil.

A solution of CsF (1.14 g, 7.51 mmol) in DMSO (5 mL) under CO2 at rt was treated with a solution of ((4-chloro-2-fluorophenyl)ethynyl)trimethylsilane (1.42 g, 6.26 mmol) in DMSO (7 mL) dropwise and the reaction was stirred under CO2 at rt overnight. The reaction mixture was diluted with ¾0 (90 mL) and extracted with CH2CI2 (2 x 50 mL). The aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and then extracted with Et 2 0 (3 x 100 mL). The combined organic layers were washed with ¾0 (50 mL), dried (MgSO i ), concentrated under reduced pressure, and dried under high vacuum to give 3-(4-chloro-2- fluorophenyl)propiolic acid (0.828 g, 4.17 mmol, 60% (2 steps)) as tan solid: Mp 174.1-176.4 °C; IR (CH2CI2) 2972, 2233, 1720, 1603, 1486, 1387, 1298, 1189, 1072, 883, 827 cm 1 ; Ή NMR (400 MHz, CDCL) d 9.09 (s, 1 H), 7.53 (t, / = 7.4 Hz, 1 H), 7.20 (d, J = 8.2 Hz, 2 H); 13 C NMR (100 MHz, CDC1 3 ) d 163.6 (d, JCF = 259.9 Hz), 156.9, 138.7 (d, JCF = 10.0 Hz), 135.2 (d, JCF = 0.9 Hz), 125.1 (d, JCF = 3.7 Hz), 117.0 (d, JCF— 23.6 Hz), 106.8 (d, J CF = 15.5 Hz), 85.1, 81.1; 19 F NMR (376 MHz, CDC1 3 ) d -104.3 (s, 1 F); HRMS (ESI) m/z calcd for C9H3CIFO2 ([M-H] ) 196.9811, found 196.9831.

Methyl 3-(6-(trifluoromethyl)pyridin-3-yl)propiolate. To two flasks each containing a solution of Pd(PPh3)2Cl2 (0.0466 g, 0.0664 mmol), Cul (0.0126 g, 0.0664 mmol), and 5-bromo-2-trifluoromethyl pyridine (1.50 g, 6.64 mmol) in Et 3 N (13 mL) sparged with Ar for 10 min followed by addition of

(trimethylsilyl) acetylene (1.4 mL, 9.96 mmol) and sparged with Ar for 2 min. The resulting mixtures were heated to 80 °C overnight where by TLC (hexanes/EtOAc, 4: 1) the SM had been consumed. After cooling the reaction to rt, the reactions were combined, the solution was filtered through Celite, which was washed with Et 2 0 (100 mL) until the washes appeared colorless. The filtrate was concentrated under reduced pressure. The crude residue was purified by chromatography on S1O2 (hexanes/EtOAc, 9: 1) to give 2- (trifluoromethyl)-5-((trimethylsilyl)ethynyl)pyridine (3.37 g, 13.9 mmol) as orange/brown waxy solid that was taken on to the carboxylation. A solution of CsF (2.52 g, 16.6 mmol) in DMSO (20 mL) under CO2 at rt was treated with a solution of 2-(trifluoromethyl)-5-((trimethylsilyl)ethynyl)pyridine (3.37 g, 13.9 mmol) in DMSO (7 mL) dropwise and the reaction was stirred under CO2 (balloon) at rt for 5 h, treated with Mel (0.95 mL,

15.2 mmol) was added and the solution was stirred for 1 h at rt. The reaction mixture was diluted with ¾0 (200 mL), brine (100 mL) and extracted with Et20 (3 x 150 mL). The combined organic layers were washed with H 2 O (100 mL), dried (MgSO i ), and concentrated under reduced pressure. The crude product was purified by chromatography on S1O 2 (hexanes/EtOAc, 4: 1) to give methyl 3-(6-(trifluoromethyl)pyridin-3- yl)propiolate (1.87 g, 8.17 mmol, 59% (2 steps)) as tan solid: Mp 98.2-99.7 °C; IR (neat) 2962, 2233, 1712, 1433, 1337, 1242, 1127, 1085, 864, 745 cm 1 ; Ή NMR (400 MHz, CDCL) d 8.87 (s, 1 H), 8.04 (ddd, / = 8.0, 1.2, 0.6 Hz, 1 H), 7.71 (d, / = 8.0 Hz, 1 H), 3.86 (s, 3 H); 13 C NMR (100 MHz, CDCL) d 153.4, 153.1, 148.6 (q, J CF 35.5 Hz), 141.2, 121.0 (q, J CF = 274.4 Hz), 120.1 (q, J CF = 2.8 Hz), 120.0, 84.7, 80.7, 53.2; 19 F NMR (376 MHz, CDCL) d -68.3 (s, 3 F); HRMS (ESI) m/z calcd for C 10 H 7 F 3 NO 2 ([M+H] + ) 230.0423, found 230.0422.

l-(4-(5-Chloro-2-methylphenyl)piperazin-l-yl)-3-(4-(penta fluoro^6-sulfaneyl)phenyl)prop-2-yn-l- one. A solution of 3-(4-(pcntal1uoro-/.6-sulfancyl ) phenyl )propiolic acid (0.100 g, 0.367 mmol) and l-(l-(5- chloro-2-methylphenyl)piperazine hydrochloride (0.0999 g, 0.404 mmol) in CH2CI2 (3.7 mL) cooled to 0 °C was treated with EtiN (0.20 mL, 1.47 mmol). The cooled solution was treated with T3P (50 wt. % solution in EtOAc, 0.39 mL, 0.551 mmol) dropwise and the reaction was stirred at 0 °C for 30 min, warmed to rt overnight, diluted with EtOAc (30 mL), washed with ¾0 (20 mL), satd. aqueous NaHCOs (20 mL), dried (MgSO i ), filtered, and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 30% EtO Ac/hexanes), to give 1- (4-(5 -chloro-2-methylphenyl)piperazin- 1 -yl )-3-(4-(pcntafluoro-/.6-sulfancyl )phenyl )prop-2-yn- 1 -one (0.127 g, 0.272 mmol, 74%) as a pale yellow foam: IR (CHCL) 2981, 2222, 1627, 1490, 1432, 1275, 832, 792, 727 cm 1 ; Ή NMR (400 MHz, CDCL) d 7.76 (d, / = 8.8 Hz, 2 H), 7.64 (d, / = 8.8 Hz, 2 H), 7.11 (d, / = 8.1 Hz,

1 H), 6.99 (dd, / = 8.1, 2.1 Hz, 1 H), 6.95 (d, / = 2.1 Hz, 1 H), 3.96 (app t, / = 5.0 Hz, 2 H), 3.84 (app t, / = 5.0 Hz, 2 H), 2.97 (app t, / = 5.0 Hz, 2 H), 2.89 (app t, / = 5.0 Hz, 2 H), 2.29 (s, 3 H); 13 C NMR (100 MHz, CDCL) d 154.4 (quint, J CF = 18.3 Hz), 152.4, 151.6, 132.4, 132.1, 131.9, 131.0, 126.2 (quint, J CF = 4.6 Hz), 124.1, 123.8, 119.8, 88.2, 83.2, 51.9, 51.3, 47.4, 42.0, 17.3; 19 F NMR (376 MHz, CDCL) d 83.0 (quint, / = 150.3 Hz, 1 F), 62.4 (d, J = 150.3 Hz, 4 F); HRMS (ESI) m/z calcd for C20H19CIF5N2OS ([M+H] + ) 465.0821, found 465.0819.

(4-(5-Chloro-2-methylphenyl)piperazin- l-yl)(( l/?S,2S/?)-2-(4-(pcntafluoro-}.6-sulfancyl)phcnyl)- cyclopropyl)methanone. A solution of l-(4-(5-chloro-2-methylphenyl)piperazin-l-yl)-3-(4-(pentaflu oro- /.6-sul faneyl )phcnyl )prop-2-yn- 1 -one (0.0850 g, 0.183 mmol) in EtOAc (1.8 mL) was treated with Lindlar's catalyst (5% Pd on CaCCE, lead poisoned, 0.0195 g, equivalent to 5 mol% Pd). The reaction was placed under a balloon of ¾ (3 vacuum/backfill cycles) and stirred at rt for 2 d, filtered through Celite, washed (EtOAc), and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 40%

EtO Ac/hexanes, product eluted at 35% EtO Ac/hexanes) to give (Z)-l-(4-(5-chloro-2-methylphenyl)- piperazin- 1 -yl )-3-(4-(pcntafluoro-/.6-sul fancyl )phcnyl )prop-2-en- 1 -one (0.0811 g, 0.174 mmol) as a colorless solid.

A solution of CrCE (0.126 g, 1.03 mmol) and (Z)-l-(4-(5-chloro-2-methylphenyl)piperazin-l-yl)-3- (4-(pentafluoro- 6-sulfaneyl)phenyl)prop-2-en-l-one (0.0800 g, 0.171 mmol) in dry degassed THF (1.7 mL) was sparged with Ar for 5 min and added CH2ICI (0.10 mL, 0.857 mmol) at rt, heated for 2 d at 80 °C, cooled to rt, diluted with EtOAc (50 mL) and washed with 1 M aqueous HC1 (3 x 20 mL). The organic layer was dried (MgSO i ), filtered and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 30% EtO Ac/hexanes, product eluted at 30% EtO Ac/hexanes) to give a clear oil that was filtered through basic AI2O3

(CH2Cl2/EtOAc, 1 : 1), concentrated under reduced pressure, and dried under high vacuum to give (4-(5- chloro-2-methylphenyl)piperazin-l-yl)((li¾2SR)-2-(4-(pentaf luoro- 6-sulfaneyl)phenyl)cyclopropyl)- methanone (0.0451 g, 0.0938 mmol, 52% (2 steps) (100% purity by ELSD)) as a colorless solid: Mp 169.8- 172.0 °C; IR (CH2CI2) 2919, 1639, 1490, 1466, 1438, 1225, 1035, 835, 750 cm 1 ; Ή NMR (400 MHz, CDCE) d 7.66 (d, / = 8.6 Hz, 2 H), 7.25 (d, / = 8.6 Hz, 2 H), 7.06 (dd, / = 8.1, 0.4 Hz, 1 H), 6.95 (dd, J = 8.1, 2.1 Hz, 1 H), 6.69 (d, / = 2.1 Hz, 1 H), 3.82 (bd, / = 14.0 Hz, 1 H), 3.70-3.58 (m, 2 H), 3.33 (ddd, / = 12.4, 8.9, 3.0 Hz, 1 H), 2.75 (tdd, / = 14.0, 7.7, 3.4 Hz, 2 H), 2.50 (td, / = 8.9, 7.0 Hz, 1 H), 2.32-2.23 (m, 2 H), 2.20 (s, 3 H), 2.10 (ddd, / = 11.1, 8.2, 3.0 Hz, 1 H), 1.91 (q, / = 6.3 Hz, 1 H), 1.44 (td, / = 8.4, 5.6 Hz, 1 H); 13 C NMR (100 MHZ, CDC1 3 ) d 166.5, 152.1 (quint, J CF = 17.3 Hz), 151.6, 141.9, 131.9 (2 C), 130.9, 127.7, 125.7 (quint, J CF = 4.6 Hz), 123.7, 119.5, 51.9, 51.5, 45.5, 42.2, 24.6, 23.7, 17.3, 11.3; 19 F NMR (376 MHz, CDCE) d 84.8 (quint, / = 150.4 Hz, 1 F), 63.2 (d, / = 150.4 Hz, 4 F); HRMS (ESI) m/z ealed for C21H23CIF5N2OS ([M+H] + ) 481.1134, found 481.1134.

Racemic (4-(5-chloro-2-mcthylphcnyl )pi pcrazi n- 1 -yl )(( I A > .S , ,2.S , A > )-2-(4-(pcntafl uoro-/.6-sul fancyl )phcnyl )- cyclopropyl)methanone was separated on a SEC Chiralpak-IC semiprep (250 x 10 mm) column (30% Methanol:C0 2 , 7 mL/min, p = 100 bar, 220 nm) injection volume 90 pL, 20 mg/mL) to give (4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lS,2R)-2-(4-(pentafluoro- 6-sulfaneyl)phenyl)cyclopropyl)methanone (retention time 5.14 min) as a colorless viscous oil (100% purity by ELSD): [CC] 17 D -134.2 (c 0.60, MeOH); Ή NMR (300 MHz, CDCI3) d 7.66 (d, / = 8.7 Hz, 2 H), 7.26 (overlap, 2 H), 7.06 (d, / = 8.1 Hz, 1 H), 6.96 (dd, / = 8.1, 1.8 Hz, 1 H), 6.70 (d, J = 1.8 Hz, 1 H), 3.87-3.79 (m, 1 H), 3.73-3.58 (m, 3 H), 3.38-3.31 (m, 1 H), 2.81-2.70 (m, 2 H), 2.50 (q, / = 8.7 Hz, 1 H), 2.33-2.24 (m, 2 H), 2.20 (s, 3 H), 2.16-2.08 (m, 1 H), 1.91 (q, J = 5.7 Hz, 1 H), 1.44 (td, J = 8.1, 5.7 Hz, 1 H). The enantiomeric excess was >99.9% ee (SEC

Chiralpak-IC (250 x 10 mm); 30% MethanohCCE, 7 mL/min, p = 100 bar, 220 nm; retention time: 5.1 min). (4-(5-Chloro-2-methylphenyl)piperazin-l-yl)((15,2R)-2-(4-(pe ntafluoro- 6-sulfaneyl)phenyl)cyclopropyl)- methanone (retention time 6.57 min) was obtained as a colorless viscous oil (100% purity by ELSD): [CC] 17 D + 136.3 (c 0.59, MeOH); Ή NMR (300 MHz, CDCE) d 7.66 (d, / = 8.7 Hz, 2 H), 7.26-7.24 (overlap, 2 H), 7.06 (d, / = 8.4 Hz, 1 H), 6.96 (dd, / = 8.4, 1.8 Hz, 1 H), 6.70 (d, / = 1.8 Hz, 1 H), 3.85-3.78 (m, 1 H), 3.67- 3.61 (m, 2 H), 3.39-3.30 (m, 1 H), 2.81-2.70 (m, 2 H), 2.50 (q, / = 8.7 Hz, 1 H), 2.33-2.24 (m, 2 H), 2.20 (s, 3 H), 2.16-2.10 (m, 1 H), 1.91 (q, / = 5.7 Hz, 1 H), 1.44 (td, / = 8.4, 5.7 Hz, 2 H). The enantiomeric excess was >99.9% ee (SFC Chiralpak-IC (250 x 10 mm); 30% Methanol:C0 2 , 7 mL/min, p = 100 bar, 220 nm; retention time: 6.5 min).

l-(4-(5-Chloro-2-methylphenyl)piperazine-l-yl)-3-(3-(pent afluoro-k6-sulfaneyl)phenyl)prop-2-yn-l- one. A solution of 3-(3-(pentafluoro-/.6-sulfaneyl ) phenyl jpropiol ic acid (0.100 g, 0.367 mmol) and l-(l-(5- chloro-2-methylphenyl)piperazine hydrochloride (0.0999 g, 0.404 mmol) in CH2CI2 (3.7 mL) cooled to 0 °C was treated with EtsN (0.20 mL, 1.47 mmol). The cooled solution was treated with T3P (50 wt. % solution in EtOAc, 0.39 mL, 0.551 mmol) dropwise and the reaction was stirred at 0 °C for 30 min and allowed to warm to rt overnight. The reaction was diluted with EtOAc (30 mL) and washed with H2O (20 mL), satd. aqueous NaHCO s (20 mL), dried (MgSO i ), filtered, and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 30% EtO Ac/hexanes), to give I -(4-(5-chloro-2-mcthylphcnyl )pipcrazinc- 1 -yl )-3-(3-(pcntafluoro-/.6- sulfaneyl)phenyl)prop-2-yn-l-one (0.113 g, 0.244 mmol, 66%) as a pale yellow solid: Mp 127.9-133.0 °C; IR (CHCls) 2921, 2219, 1627, 1432, 1288, 1223, 1041, 838, 797, 728 cm 1 ; Ή NMR (400 MHz, CDCI3) d 7.94 (t, / = 2.0 Hz, 1 H), 7.80 (ddd, / = 8.4, 2.0, 0.8 Hz, 1 H), 7.69 (d, / = 7.7 Hz, 1 H), 7.49 (t, / = 8.1 Hz, 1 H), 7.11 (d, / = 8.1 Hz, 1 H), 6.99 (dd, / = 8.1, 2.1 Hz, 1 H), 6.96 (d, / = 2.1 Hz, 1 H), 3.97 (app t, / = 5.0 Hz, 2 H), 3.84 (app t, J = 5.0 Hz, 2 H), 2.98 (app t, / = 5.0 Hz, 2 H), 2.89 (app t, / = 5.0 Hz, 2 H), 2.29 (s, 3 H); 13 C NMR (100 MHZ, CDC1 3 ) d 153.8 (quint, J CF = 18.5 Hz), 152.4, 151.6, 135.1, 132.1, 131.9, 131.0, 129.7 (quint, J CF = 7.1 Hz), 129.0, 127.3 (quint, J CF = 4.5 Hz), 123.8, 121.6, 119.8, 88.4, 82.3, 51.9, 51.3, 47.4, 42.0, 17.3; 19 F NMR (376 MHz, CDC1 3 ) d 82.9 (quint, J = 150.6 Hz, 1 F), 62.6 (d, J = 150.5 Hz, 4 F); HRMS (ESI) m/z calcd for C20H19CIF5N2OS ([M+H] + ) 465.0821, found 465.0821.

(4-(5-Chloro-2-methylphenyl)piperazine- l-yl)(( l/?S,2,S7?)-2-(3-(pcntafluoro-}.6- sulfaneyl)phenyl)cyclopropyl)methanone. A solution of l-(4-(5-chloro-2-methylphenyl)piperazine-l-yl)- 3-(4-(pentafluorosulfanyl)phenyl)prop-2-yn-l-one (0.0930 g, 0.200 mmol) in EtOAc (2 mL) was treated with Lindlar’s catalyst (5% Pd on CaCCE, lead poisoned, 0.0213 g, equivalent to 5 mol% Pd). The reaction was placed under a balloon of ¾ (3 vacuum/backfill cycles) and stirred at rt for 3 d, filtered through Celite, washed (EtOAc) and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by automated chromatography on SiCE (4g column, liquid load CH2CI2, 100% hexanes to 30% EtO Ac/hexanes, product eluted at 20% EtO Ac/hexanes) to give (Z)-l-(4-(5-chloro-2-methylphenyl)- pipcrazinc- 1 -yl )-3-(3-(pcntafluoro-/.6-sulfancyl )phcnyl )prop-2-en- 1 -one (0.0470 g, 0.101 mmol) as a colorless solid.

To a flame dried 5 mL microwave vial containing CrC'l· (0.0742 g, 0.604 mmol) was added a solution of (Z)-l-(4-(5-chloro-2-methylphenyl)piperazine-l-yl)-3-(3-(pen tafluoro- 6-sulfaneyl)phenyl)prop- 2-en-l-one (0.0470 g, 0.101 mmol) in dry degassed THF (1 mL) and the mixture was sparged with Ar for 15 min and added CH2ICI (0.058 mL, 0.503 mmol) at rt, heated for 2 d at 80 °C. The reaction was cooled to rt, diluted with EtOAc (50 mL) and washed with 1 M aqueous HC1 (3 x 20 mL). The organic layer was dried (MgSO i ), filtered and concentrated under reduced pressure. The crude residue was purified by automated chromatography on SiCE (4g column, liquid load CH2CI2, 100% hexanes to 30% EtO Ac/hexanes, product eluted at 20% EtO Ac/hexanes) to give the product as a clear oil. The product was filtered through basic AI2O3 (CH2Cl2/EtOAc, 1 : 1) concentrated and dried under high vacuum to give (4-(5-chloro-2- methylphenyl)piperazine-l-yl)((lR5,25R)-2-(3-(pentafluoro- 6-sulfaneyl)phenyl)cyclopropyl)methanone (0.0182 g, 0.0378 mmol, 19% (2 steps) (100% purity by ELSD)) as a colorless solid: Mp 112.9-116.1 °C; IR (CH2CI2) 2854, 1640, 1490, 1439, 1225, 1036, 841 cm 1 ; Ή NMR (400 MHz, CDC1 3 ) d 7.62-7.59 (m, 2 H), 7.38 (t, / = 8.1 Hz, 1 H), 7.29 (d, / = 7.8 Hz, 1 H), 7.06 (d, / = 8.5 Hz, 1 H), 6.95 (dd, / = 8.1, 2.1 Hz, 1 H), 6.70 (d, / = 2.1 Hz, 1 H), 3.84 (bd, / = 12.8 Hz, 1 H), 3.76-3.71 (m, 1 H), 3.61 (ddd, / = 12.8, 8.9, 3.2 Hz, 1 H), 3.29 (ddd, J = 12.8, 9.0, 3.1 Hz, 1 H), 2.80-2.71 (m, 2 H), 2.53 (td, / = 8.9, 6.9 Hz, 1 H), 2.31-2.23 (m, 2 H), 2.20 (s, 3 H), 2.15-2.09 (m, 1 H), 1.91 (q, / = 6.2 Hz, 1 H), 1.43 (td, / = 8.4, 5.6 Hz, 1 H); 13 C NMR (100 MHz, CDCI3) d 166.6, 153.9 (quint, J CF = 16.8 Hz), 151.8, 139.0, 132.0, 131.8, 131.0, 130.2, 128.6, 125.9 (quint, J CF = 4.5 Hz), 124.0 (quint, J CF = 4.6 Hz), 123.7, 119.7, 51.8, 51.6, 45.6, 42.2, 24.1, 24.0, 17.3, 11.0; 19 F NMR (376 MHz, CDCI3) d 84.7 (quint, / = 150.1 Hz, 1 F), 62.9 (d, / = 149.9 Hz, 4 F); HRMS (ESI) m/z calcd for C21H23CIF5N2OS ([M+H] + ) 481.1134, found 481.1133.

l-(4-(2-Methyl-5-(trifluoromethyl)phenyl)piperazine-l-yl) -3-(3-(pentafluoro^6-sulfaneyl)phenyl)- prop-2-yn-l-one. A solution of 3-(3-(pentafluoro-/.6-sulfaneyl ) phenyl )propiolic acid (0.100 g, 0.367 mmol) and l-(2-methyl-5-(trifluoromethyl)phenyl)piperazine hydrochloride (0.113 g, 0.404 mmol) in CH2CI2 (3.7 mL) cooled to 0 °C was treated with EtsN (0.20 mL, E47 mmol). The cooled solution was treated with T3P (50 wt. % solution in EtOAc, 0.39 mL, 0.551 mmol) dropwise and the reaction was stirred at 0 °C for 30 min, warmed to rt overnight, diluted with EtOAc (30 mL), washed with ¾0 (20 mL), satd. aqueous NaHCOs (20 mL), dried (MgSO i ), filtered, and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 40%

EtO Ac/hexanes), to give I -(4-(2-mcthyl-5-(trifluoromcthyl )phcnyl ) piperazine- 1 -yl )-3-(3-( pen tail uoro-/.6- sulfaneyl)phenyl)prop-2-yn-l-one (0.150 g, 0.300 mmol, 82%) as a tan foam: IR (CH2CI2) 2919, 2825,

2219, 1629, 1418, 1309, 1152, 1119, 840, 797, 730 cm 1 ; Ή NMR (400 MHz, CDC1 3 ) d 7.94 (t, / = 1.8 Hz,

1 H), 7.81 (dt, / = 8.2, 1.1 Hz, 1 H), 7.70 (d, / = 7.8 Hz, 1 H), 7.50 (t, / = 8.2 Hz, 1 H), 7.29 (q, / = 7.8 Hz, 2 H), 7.22 (s, 1 H), 4.00 (app t, / = 5.0 Hz, 2 H), 3.87 (app t, / = 5.0 Hz, 2 H), 3.04 (app t, J = 5.0 Hz, 2 H), 2.94 (app t, J = 5.0 Hz, 2 H), 2.39 (s, 3 H); 13 C NMR (100 MHz, CDCL) d 153.8 (quint, J CF = 18.2 Hz), 152.5, 150.9, 136.8, 135.1, 131.6, 129.7 (quint, J CF = 4.7 Hz), 129.1 (q, J CF = 323 HZ), 129.1, 127.4 (quint, J CF — 4.7 Hz), 124.1 (q, J CF = 271.8 Hz), 121.5, 120.6 (q, J CF = 3.9 Hz), 116.1 (q, J CF = 3.6 Hz), 88.4, 82.3, 51.9, 51.3, 47.4, 42.0, 17.9; 19 L NMR (376 MHz, CDCL) d 82.9 (quint, / = 150.6 Hz, 1 L), 62.6 (d, / = 150.4 Hz, 4 L), -62.3 (s, 3 L); HRMS (ESI) m/z calcd for C2iHi 9 F 8 N 2 OS ([M+H] + ) 499.1085, found 499.1086.

(4-(2-Methyl-5-(trifluoromethyl)phenyl)piperazine-l-yl)((l/? S,2S/?)-2-(3-(pentafluoro^6- sulfaneyl)phenyl)cyclopropyl)methanone. A solution of l-(4-(2-methyl-5-(trifluoromethyl)phenyl)- piperazine- 1 -yl )-3-(3-(pcntafluoro-/.6-sulfancyl )phcnyl )prop-2-yn- 1 -one (0.100 g, 0.201 mmol) in EtOAc (2 mL) was treated with Lindlar’s catalyst (5% Pd on CaCCE, lead poisoned, 0.0214 g, equivalent to 5 mol% Pd). The reaction was placed under a balloon of H 2 (3 vacuum/backfill cycles) and stirred at rt for 2 d, filtered through Celite, washed (EtOAc), and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, hexanes to 40% EtO Ac/hexanes, product eluted at 30% EtO Ac/hexanes) to give (Z)-l-(4-(2-methyl- 5-(trifluoromethyl)phenyl)piperazine- 1 -yl )-3-(3-(pcntafluoro-/.6-sulfancyl )phenyl )prop-2-en- 1 -one (0.0938 g, 0.187 mmol) as a colorless foam.

A solution of CrCE (0.138 g, 1.12 mmol) and (Z)-l-(4-(2-methyl-5-(trifluoromethyl)phenyl)- pipcrazinc- 1 -yl )-3-(3-(pcntafluoro-/.6-sulfancyl )phenyl )prop-2-en- 1 -one (0.0938 g, 0.187 mmol) in dry degassed THF (1.9 mL) (degassed by sparging with Ar for 15 min) was treated with CH2ICI (0.11 mL,

0.937 mmol) at rt, heated for 2 d at 80 °C, cooled to rt, diluted with EtOAc (50 mL), and washed with 1 M aqueous HC1 (3 x 20 mL). The organic layer was dried (MgSOi ), filtered and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 40% EtO Ac/hexanes, product eluted at 30% EtO Ac/hexanes), filtered through basic AI2O3 (CFbCh/EtOAc, 1 : 1), concentrated under reduced pressure, and dried under high vacuum to give (4-(2-methyl-5-(trifluoromethyl)phenyl)piperazine-l-yl)((lR5 ,25R)-2-(3-(pentafluoro- 6-sulfaneyl)- phenyl)cyclopropyl)methanone (0.0641 g, 0.125 mmol, 62% (2 steps) (100% purity by ELSD)) as a pale yellow oil: IR (CH2CI2) 2917, 1638, 1438, 1417, 1337, 1307, 1120, 835, 758 cm 1 ; Ή NMR (400 MHz, CDCE) d 7.62-7.58 (m, 2 H), 7.38 (t, 7 = 7.8 Hz, 1 H), 7.30 (d, 7 = 7.8 Hz, 1 H), 7.24-7.21 (m, 2 H), 6.96 (s, 1 H), 3.93 (d, 7 = 13.0 Hz, 1 H), 3.79 (dt, 7 = 13.0, 3.8 Hz, 1 H), 3.60 (ddd, 7 = 12.6, 9.2, 3.1 Hz, 1 H), 3.25 (ddd, 7 = 12.6, 9.2, 3.0 Hz, 1 H), 2.79 (ddt, 7 = 20.7, 11.9, 3.8 Hz, 2 H), 2.53 (td, 7 = 8.9, 6.9 Hz, 1 H), 2.31- 2.20 (m, 5 H), 2.16-2.10 (m, 1 H), 1.92 (q, 7 = 6.2 Hz, 1 H), 1.43 (td, 7 = 8.4, 5.6 Hz, 1 H); 13 C NMR (100 MHz, CDCE) d 166.5, 153.9 (quint, J CF = 16.9 Hz), 151.0, 139.0, 136.8, 131.3, 130.3, 129.0 (q, JCF = 33.1 Hz), 128.5, 125.7 (quint, JCF = 4.6 Hz), 124.2 (q, Jn = 272.0 Hz), 123.9 (quint, JCF = 4.6 Hz), 120.3 (q, JCF = 3.9 Hz), 115.9 (q, JCF = 3.6 Hz), 51.7, 51.6, 45.5, 42.2, 24.0 (2 C), 24.0, 17.8, 10.9; 19 F NMR (376 MHz, CDCE) d 84.5 (quint, 7 = 150.0 Hz, 1 F), 62.7 (d, 7 = 149.8 Hz, 4 F), -62.4 (s, 3 F); HRMS (ESI) m/z ealed for C22H 2 3F 8 N 2 OS ([M+H] + ) 515.1398, found 515.1380.

Racemic (4-(2-methyl-5-(trifluoromethyl)phenyl)- 31 -iperazine- 1 -yl)(( I A’.S', 2.S' A’ ) - 2- ( 3 - ( pc n t all u o ro -l6 - sulfaneyl)phenyl)cyclopropyl)methanone was separated on a SFC Chiralpak-IC semiprep (250 x 10 mm) column (30% Methanol:C0 2 , 7 mL/min, p = 100 bar, 220 nm) injection volume 90 pL, 20 mg/mL) to give (4-(2-methyl-5-(trifluoromethyl )phcnyl )piperazin- 1 -yl )(( I .S , ,2A > )-2-(3-(pcntafluoro-/.6-sulfancyl )phcnyl )- cyclopropyl)methanone (retention time 3.25 min) as a colorless viscous oil (100% purity by ELSD): [CC] 18 D - 104.7 (c 0.84, MeOH); Ή NMR (300 MHz, CDCE) d 7.62-7.58 (m, 2 H), 7.38 (t, 7 = 7.7 Hz, 1 H), 7.31- 7.24 (m, 3 H), 6.96 (s, 1 H), 3.94-3.89 (m, 1 H), 3.81-3.75 (m, 1 H), 3.65-3.56 (m, 1 H), 3.31-3.22 (m, 1 H), 2.84-2.74 (m, 2 H), 2.53 (td, 7 = 9.3, 6.9 Hz, 1 H), 2.32-2.21 (m, 5 H), 2.19-2.11 (m, 1 H), 1.93 (q, 7 = 5.7 Hz, 1 H), 1.43 (td, 7 = 8.4, 5.7 Hz, 1 H). The enantiomeric excess was >99.9% ee (SFC Chiralpak-IC (250 x 10 mm); 30% MethanohCCE, 7 mL/min, p = 100 bar, 220 nm; retention time: 3.3 min).

(4-(2-Mcthyl-5-(trifluoromcthyl )phenyl )piperazin- 1 -yl )(( I A > ,2.S , )-2-(3-(pcntafluoro-/.6- sulfaneyl)phenyl)-cyclopropyl)methanone (retention time 3.60 min) was obtained as a colorless viscous oil (100% purity by ELSD): [a] 18 D +103.4 (c 0.87, MeOH); Ή NMR (300 MHz, CDCE) d 8.54-7.81 (m, 2 H), 7.62-7.59 (m, 2 H), 7.38 (t, 7 = 7.8 Hz, 1 H), 7.31-7.24 (m, 3 H), 6.96 (s, 1 H), 3.94-3.88 (m, 1 H), 3.81-3.75 (m, 1 H), 3.65-3.56 (m, 1 H), 3.31-3.22 (m, 1 H), 2.84-2.74 (m, 2 H), 2.53 (td, 7 = 9.0, 7.2 Hz, 1 H), 2.32- 2.22 (m, 5 H), 2.18-2.11 (m, 1 H), 1.93 (q, 7 = 5.7 Hz, 1 H), 1.43 (td, 7 = 8.4, 5.7 Hz, 1 H). The enantiomeric excess was >99.9% ee (SFC Chiralpak-IC (250 x 10 mm); 30% Methanol:C0 2 , 7 mL/min, p = 100 bar, 220 nm; retention time: 3.6 min).

3-(4-Chloro-2-fluorophenyl)-l-(4-(5-chloro-2-methylphenyl)pi perazin-l-yl)prop-2-yn-l-one. A solution of 3-(4-chloro-2-fluorophenyl)propiolic acid (0.190 g, 0.957 mmol) and l-(5-chloro-2-methylphenyl)- piperazine hydrochloride (0.284 g, 1.15 mmol) in CH2CI2 (9.6 mL) cooled to 0 °C was treated with EhN (0.53 mL, 3.83 mmol). The cooled solution was treated with T3P (50% solution in EtOAc) (1.0 mL, 1.44 mmol) dropwise and the reaction was stirred at 0 °C for 30 min, warmed to rt overnight, diluted with EtOAc (40 mL) and washed with ¾0 (20 mL), satd. aqueous NaHCOs (20 mL), dried (MgSO i ), filtered, and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, gradient 100% hexanes to 100% EtOAc) to give 3-(4-chloro-2- fluorophenyl)-l-(4-(5-chloro-2-methylphenyl)piperazin-l-yl)p rop-2-yn-l-one (0.302 g, 0.772 mmol, 81%) as a pale yellow solid: Mp 135.1-137.3 °C; IR (CH2CI2) 2917, 2820, 2219, 1632, 1489, 1431, 1291, 1224, 1039, 898, 819 cm 1 ; Ή NMR (400 MHz, CDCE) d 7.50 (dd, 7 = 8.8, 8.0 Hz, 1 H), 7.17 (t, 7 = 2.0 Hz, 1 H), 7.15 (q, 7 = 2.0 Hz, 1 H), 7.11 (d, 7 = 8.4 Hz, 1 H), 6.98 (dd, 7 = 8.0, 2.0 Hz, 1 H), 6.94 (d, 7 = 2.4 Hz, 1 H), 3.97 (t, 7 = 5.0 Hz, 2 H), 3.83 (t, 7 = 5.0 Hz, 2 H), 2.96 (t, 7 = 5.0 Hz, 2 H), 2.88 (t, 7 = 5.0 Hz, 2 H), 2.28 (s, 3 H); 13 C NMR (100 MHz, CDCE) d 163.0 (d, JCF = 257.4 Hz), 152.5, 151.6, 137.4 (d, JCF = 10.1 Hz), 134.7 (d, JCF = 1.4 Hz), 131.9 (d, J CF = 25.0 Hz), 130.9, 125.0 (d, J CF = 3.7 Hz), 123.7, 119.8, 116.8, 116.6, 107.9 (d, JCF = 15.9 Hz), 86.6 (d, JCF = 3.6 Hz), 83.0, 51.8, 51.2, 47.3, 41.9, 17.4; 19 F NMR (376 MHz, CDCE) d - 105.8 (s, 1 F); HRMS (ESI) m/z calcd for C 2O H I8 C FN 2 0 ([M+H] + ) 391.0775, found 391.0781.

((l/?S,2S/?)-2-(4-Chloro-2-fluorophenyl)cyclopropyl)(4-(5-ch loro-2-methylphenyl)piperazin-l- yl)methanone A solution of 3-(4-chloro-2-fluorophenyl)-l-(4-(5-chloro-2-methylphenyl)pi perazin-l- yl)prop-2-yn-l-one (0.300 g, 0.767 mmol) in EtOAc (7.8 mL) was treated with Lindlar's catalyst (5% Pd on CaCCE, lead poisoned, 0.0816 g, equivalent to 5 mol% Pd). The reaction was placed under a balloon of ¾

(3 vacuum/backfill cycles) and stirred at rt for 3 d, filtered through Celite, washed (EtOAc), and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 40% EtO Ac/hexanes, pdt eluted at 30% EtO Ac/hexanes) to give (Z)-3-(4-chloro-2-fluorophenyl)-l-(4-(5-chloro-2-methylpheny l)piperazin- l-yl)prop-2-en-l-one (0.138 g, 0.351 mmol, 46%) as a pale yellow oil.

A solution of CrCl· (0.259 g, 2.11 mmol) and (Z)-3-(4-chloro-2-fluorophenyl)-l-(4-(5-chloro-2- methylphenyl)piperazin-l-yl)prop-2-en-l-one (0.138 g, 0.351 mmol) in dry degassed THF (3.5 mL) was sparged with Ar for 5 min and treated with CH2ICI (0.20 mL, 1.75 mmol) at rt, further sparged for 5 min, heated for 20 h at 80 °C, cooled to rt, diluted with EtOAc (50 mL) and washed with 1 M aqueous HC1 (3 x 20 mL). The organic layer was dried (MgSO i ), filtered, and concentrated under reduced pressure. The crude residue was purified by automated chromatography on S1O2 (4g column, liquid load CH2CI2, 100% hexanes to 40% EtO Ac/hexanes), filtered through basic AI2O3 (1 : 1 CPbCE/EtOAc) and dried under high vacuum to give (( I A > .S , ,2.S , A > )-2-(4-chloro-2-fluorophcnyl jcyclopropyl )(4-(5-chloro-2-mcthylphcnyl jpipera/.in- 1 - yl)methanone (0.0930 g, 0.228 mmol, 65% (100% purity by ELSD)) as a colorless solid: Mp 100.2- 103.0 °C; IR (CH2CI2) 2950, 2819, 1638, 1490, 1435, 1225, 1034, 899, 818, 731 cm 1 ; Ή NMR (400 MHz, CDCE) d 7.09-7.03 (m, 4 H), 6.97 (dd, / = 8.0, 2.0 Hz, 1 H), 6.81 (d, / = 2.0 Hz, 1 H), 3.81-3.66 (m, 3 H), 3.38 (ddd, J = 12.4, 8.0, 3.4 Hz, 1 H), 2.91-2.86 (m, 1 H), 2.77-2.72 (m, 1 H), 2.58 (qd, / = 8.4, 0.8 Hz, 1 H), 2.50 (ddd, / = 11.2, 8.0, 2.8 Hz, 1 H), 2.40 (ddd, / = 11.2, 8.0, 2.8 Hz, 1 H), 2.29 (ddd, / = 9.2, 8.0, 5.6 Hz, 1 H), 2.23 (s, 3 H), 1.90 (dt, / = 6.8, 5.6 Hz, 1 H), 1.34 (td, / = 8.4, 5.6 Hz, 1 H); 13 C NMR (100 MHz, CDCE) d 167.0, 161.9 (d, J CF = 247.9 Hz), 151.9, 132.8 (d, J CF = 10.5 Hz), 132.0, 131.8, 130.9, 129.8 (d, J CF = 4.5 Hz), 124.2 (d, Jc F = 3.4 Hz), 123.5, 123.1 (d , J CF = 14.3 Hz), 119.6, 115.4 (d, / CF = 25.3 Hz), 51.8, 51.6, 45.6, 42.2, 22.2, 17.4, 17.4, 9.3; 19 F NMR (376 MHz, CDC1 3 ) d -116.4 (s, 1 F); HRMS (ESI) m/z calcd for C21H22CI2FN2O ([M+H] + ) 407.1088, found 407.1089.

l-(2-Methyl-5-(trifluoromethyl)phenyl)piperazine hydrochloride. A solution of Boc-piperazine (2.14 g, 11.5 mmol), KOtBu (2.35 g, 20.9 mmol), (racemic)-BINAP (0.671 g, 1.05 mmol), Pd 2 (dba) 3 (0.194 g, 0.209 mmol) in dry toluene (105 mL) was sparged with Ar for 20 min, treated with 2-bromo-l-methyl-4- (trifluoromethyl)benzene (2.50 g, 10.5 mmol), and the mixture was heated under Ar at 100 °C overnight, cooled to rt, diluted with Et 2 0 (125 mL), filtered through Celite, washed (Et 2 0), and the combined filtrates were concentrated under reduced pressure. The crude residue was purified by chromatography on S1O2 (hexanes/EtOAc, 9: 1) to give tert-butyl 4-(2-methyl-5-(trifluoromethyl)phenyl)piperazine-l-carboxyla te (2.49 g, 7.23 mmol) as an orange oil.

A solution of tert-butyl 4-(2-methyl-5-(trifluoromethyl)phenyl)piperazine-l-carboxyla te (2.49 g,

7.23 mmol) in THF (3.5 mL) was cooled to 0 °C and treated with 4M HCI in dioxane (9.0 mL, 36.2 mmol) was added and the reaction was stirred at 0 °C for 30 min and then rt for 4 h. The solution was concentrated under reduced pressure and the tan solid was precipitated in ether, filtered off from the solution, washed with Et 2 0, dried under vacuum to give the crude product. The crude material was recrystallized from

EtOH/hexanes (1: 1) to give the product (1.71 g, 6.10 mmol, 58% (2 steps)) as colorless needles. Mp 296 °C (decomp); IR (CH2CI2) 2939, 2799, 2498, 1610, 1418, 1338, 1307, 1153, 1076, 950, 827 cm 1 ; Ή NMR (400 MHz, DMSO-de) d 9.44 (bs, 2 H), 7.42 (d, / = 7.9 Hz, 1 H), 7.35 (dd, / = 7.9, 1.0 Hz, 1 H), 7.25 (d, / = 1.0 Hz, 1 H), 3.22 (dd, / = 6.2, 3.6 Hz, 4 H), 3.12 (dd, / = 6.2, 3.6 Hz, 4 H), 2.33 (s, 3 H); 13 C NMR (100 MHz, DMSO-de) d 150.8, 137.2, 131.9, 127.5 (q, JCF = 31.6 Hz), 124.2 (q, J CF = 272.1 Hz), 120.1 (q, JCF = 3.9 Hz), 115.3 (q, J CF = 3.8 Hz), 47.9, 43.1, 17.7; 19 F NMR (376 MHz, CDCE) d -60.7 (s, 3 F); HRMS (ESI) m/z calcd for C12H16F3N2 ([M+H] + ) 245.1260, found 245.1261.

l-(5-Chloro-2-(trifluoromethyl)phenyl)piperazine hydrochloride. Two flasks each containing a solution Boc-piperazine (1.58 g, 8.48 mmol), KOtBu (1.73 g, 15.4 mmol), (racemic)-BINAP (0.480 g, 0.771 mmol), Pd2dba3 (0.142 g, 0.154 mmol) in toluene (8 mL) was sparged with argon for 15 min and treated with 2- bromo-4-chloro-l-(trifluoromethyl)benzene (2.00 g, 7.71 mmol), heated under N2 at 80 °C for 24 h, cooled to rt, combined, diluted with Et 2 0 (100 mL) and added Celite and filtered through Celite, washed (Et 2 0), and the combined organic layers were concentrated in vacuo. The crude residue was purified by chromatography on S1O2 (hexanes/EtOAc, 9: 1) to give 4-(5-chloro-2-(trifluoromethyl)phenyl)piperazine-l- carboxylate (3.12 g, 8.55 mmol) as an orange oil. A solution of tert-butyl 4-(5-chloro-2-(trifluoromethyl)phenyl)piperazine-l-carboxyla te (3.12 g, 8.55 mmol) in THF (8 mL) was treated with 4M HC1 in dioxane (10.7 mL, 42.8 mmol), stirred at rt overnight, diluted with hexanes (100 mL), and the resulting precipitate was filtered and washed with additional hexanes and Et 2 0. The crude material was recrystallized at rt (EtOH/hexanes), crystals were collected by vacuum filtration, washed (hexanes) and dried under high vacuum to give l-(5-chloro-2-(trifluoromethyl)phenyl)- piperazine hydrochloride (1.88 g, 6.24 mmol, 41% (2 steps)) as tan solid: Mp 272 °C (decomp); IR (neat) 2947, 2726, 2480, 1599, 1576, 1307, 1118, 1037, 946, 819 cm 1 ; Ή NMR (400 MHz, DMSO-de) d 9.58 (s, 2 H), 7.72 (d, 7 = 8.8 Hz, 1 H), 7.57 (d, 7 = 1.6 Hz, 1 H), 7.46 (dd, 7 = 8.8, 1.6 Hz, 1 H), 3.13 (bs, 8 H); 13 C NMR (100 MHz, DMSO-d 6 ) d 152.2, 138.3, 129.0 (q, 7 = 5.4 Hz), 125.9, 124.7, 124.2 (q, 7 = 29.7 Hz),

123.7 (q, 7 = 273.0 Hz), 49.6, 43.1; 19 F NMR (376 MHz, CDCL) d -59.0 (s, 3 F); HRMS (ESI) m/z calcd for

265.0714, found 265.0712.

4-Bromo-2-(piperazin-l-yl)benzonitrile hydrochloride. A suspension of 4-bromo-2-fluorobenzonitrile (2.00 g, 10.0 mmol), 1-boc-piperazine (1.86 g, 10.0 mmol), and Et N (1.4 mL, 10.0 mmol) in anhydrous MeCN (5.0 mL) was heated to 110 °C for 21 h, cooled to rt, and concentrated under reduced pressure. The crude residue was purified by chromatography on S1O 2 (hexanes/EtOAc, 9: 1) to give tert-butyl 4-(5-bromo- 2-cyanophenyl)piperazine-l-carboxylate (3.03 g, 8.27 mmol) as a yellow oil.

A solution of tert-butyl 4-(5-bromo-2-cyanophenyl)piperazine-l-carboxylate (3.03 g, 8.27 mmol) in THF (4 mL) was cooled to 0 °C and treated with 4M HC1 in dioxane (10.3 mL, 41.4 mmol), stirred at 0 °C for 30 min, rt for 16 h, diluted with hexanes (200 mL), and the resulting precipitate was collected by vacuum filtration, washed with hexanes, ether, and dried under high vacuum to give 4-bromo-2-(piperazin-l- yl)benzonitrile hydrochloride (3.82 g, 13.6 mmol, 79% (2 steps)) as a colorless solid: Mp 288 °C (decomp); IR (neat) 2901, 2748, 1459, 1129, 1581, 1411, 1241, 1118, 949, 874 cm 1 ; Ή NMR (400 MHz, MeOD-d4) d 7.59 (d, 7 = 8.0 Hz, 1 H), 7.44 (d, 7 = 1.6 Hz, 1 H), 7.37 (dd, 7 = 8.0, 1.6 Hz, 1 H), 3.49 (td, 7 = 4.0, 1.2 Hz,

4 H), 3.44 (td, 7 = 4.0, 1.2 Hz, 4 H); 13 C NMR (100 MHz, MeOD-d4) d 156.5, 136.4, 129.9, 127.8, 124.2, 118.2, 106.7, 49.6, 44.9; HRMS (ESI) m/z calcd for CnHi 3 N 3 Br ([M+H] + ) 266.0287, found 266.0286.

l-(5-Chloro-2-fluorophenyl)piperazine hydrochloride. Under N 2 atmosphere, CuBr (0.471 g, 3.22 mmol), l,l’-bi-2-naphthol (0.692 g, 2.41 mmol) and DMF (8.04 mL) was added to the flame-dried flask. The mixture was stirred for 10 minutes before the addition of 1-Boc-piperazine (4.49 g, 24.1 mmol), K 3 PO 4 (7.04 g, 32.2 mmol) and 2-bromo-4-chloro-l-fluorobenzene (2.00 mL, 16.1 mmol). The reaction mixture was stirred at 120 °C for 22.0 h. After cooling to room temperature, the mixture was diluted with EtOAc (50 mL) and filtered through Celite pad. The filtrate was sequentially washed with saturated aqueous NH4CI (50 mL) and brine (50 mLx3). The resulting organic phase was dried (MgSOi ), filtered and concentrated in vacuo. The crude product was filtered through a short column of S1O2 (EtO Ac/hexanes, 1 : 15) to yield a mixture of ferf-butyl 4-(5-chloro-2-fluorophenyl)piperazine-l-carboxylate (1.50 g) as a light yellow oil. This mixture was used without further purification.

To a solution of the above product (1.50 g) in 1,4-dioxane (11.7 mL), HC1 (4 M in 1,4-dioxane,

4.76 mL) was added at 0 °C. The mixture was stirred at RT for 14 h. The thick suspension was diluted with hexanes (50 mL) and the resulting solid was collected by filtration, washed with hexanes and Et20, and dried to give the desired compound as a light yellow solid (0.408 g, 10% over 2 steps). Mp: 173-174 °C; IR (neat): 3013, 2956, 2838, 2528, 2484, 2391, 1516, 1478, 1456, 1393, 1269, 1123, 1042, 1019 cm 1 ; 'H-NMR (500 MHz; DMSO-d6): d 9.29 (s, 2 H), 7.23 (ddd, 7 = 12.5, 8.7, 0.8 Hz, 1 H), 7.13 (dd, 7 = 7.7, 2.5 Hz, 1 H), 7.08-7.05 (m, 1 H), 3.30-3.25 (brd, 4 H), 3.25-3.18 (brd, 4 H); 13 C-NMR (125 MHz; DMSO-d6): d 153.5 (d, 7 = 244.6 Hz), 139.8 (d, 7 = 9.9 Hz), 128.6 (d, 7 = 2.9 Hz), 122.4 (d, 7 = 8.3 Hz), 119.4 (d, 7 = 3.1 Hz), 117.6 (d, 7 = 22.6 Hz), 46.6 (d, 7 = 3.7 Hz), 42.6; 19 L-NMR (471 MHz; DMSO-d6): d -124.65 (ddd, 7 = 11.4, 7.0, 3.8 Hz, 1 L); HRMS (ESI): m/z calculated for C10H13CIL ([M+H] + ) 215.0746, found 215.0747

l-(4-(5-Chloro-2-fluorophenyl)piperazin-l-yl)-3-(4-fluoro phenyl)prop-2-yn-l-one. To a solution of 3- (4-fluorophenyl)propiolic acid (0.200 g, 1.22 mmol) in CH2CI2 (6.09 mL) at 0°C was added l-(5-chloro-2- fluorophenyl)piperazine hydrochloride (0.367 g, 1.46 mmol), and EtiN (0.432 mL, 3.05 mmol). T3P (1.29 mL, 1.83 mmol) was added dropwise and the reaction was stirred at 0 °C for 30 min and allowed to warm to room temperature for 33 h. The reaction was diluted with CH2CI2 (30 mL) and sequentially washed with 1 M HC1 (30 mLx2) and saturated aqueous NaHCCL (30 mLx2). The resulting organic phase was dried (MgSO i ), filtered and concentrated in vacuo. The crude material was purified by chromatography on S1O2 (hexanes/EtOAc, 1 : 1) to give l-(4-(5-chloro-2-fluorophenyl)piperazin-l-yl)-3-(4-fluorophe nyl)prop-2-yn-l- one (0.296 g, 0.821 mmol, 67%) as a light yellow crystal. Mp: 139-140 °C; IR (neat): 2217, 1629, 1601, 1506, 1498, 1432, 1286, 1244, 1227, 1205, 1156, 1039 cm 1 ; 'H-NMR (400 MHz; CDC1 3 ): d 7.58-7.54 (m, 2 H), 7.11-7.05 (m, 2 H), 7.01-6.89 (m, 3 H), 3.99 (t, 7 = 5.1 Hz, 2 H), 3.86 (t, 7 = 5.1 Hz, 2 H), 3.15 (t, 7 = 5.1 Hz, 2 H), 3.09 (t, 7 = 5.1 Hz, 2 H); 13 C-NMR (100 MHz; CDC1 3 ): d 163.5 (d, 7 = 252.8 Hz), 154.0 (d, 7 = 245.8 Hz), 140.3 (d, 7 = 9.9 Hz), 134.6 (d, 7 = 8.8 Hz), 129.5 (d, 7 = 3.3 Hz), 122.8 (d, 7 = 8.1 Hz), 119.6 (d, 7 = 2.9 Hz), 117.2 (d, 7 = 22.4 Hz), 116.4 (d, 7 = 3.7 Hz), 116.1 (d, 7 = 22.4 Hz), 90.1, 80.7, 50.7 (d, 7 = 3.2 Hz), 50.0 (d, 7 = 3.4 Hz), 46.9, 41.4; 19 L-NMR (376 MHz; CDCL): d -107.24 (tt, 7 = 8.1, 5.5 Hz, 1 L), - 125.19 (ddd, 7 = 11.4, 7.3, 4.6 Hz, 1 L); HRMS (ESI): m/z calculated for C19H16CION2L2 ([M+H] + ) 361.0914, found 361.0912.

(4-(5-chloro-2-fluorophenyl)piperazin-l-yl)(2-(4-fluoropheny l)cyclopropyl)methanone. To a solution of l-(4-(5-chloro-2-fluorophenyl)piperazin-l-yl)-3-(4-fluorophe nyl)prop-2-yn-l-one (0.275 g, 0.763 mmol) in EtOAc (7.63 mL, 0.1 M) was added Lindlar's catalyst (5% Pd on CaCCE, lead poisoned, 0.0812 g,

5.0 mol%) . The reaction vessel was placed under vacuum and backfilled with ¾ (balloon, 2x) and allowed to stir for 14 h. The reaction mixture was then filtered through celite, washed with EtOAc and concentrated in vacuo. The crude residue was purified by chromatography on S1O2 (hexanes/EtOAc, 1 : 1) to afford (Z)-l- (4-(5-chloro-2-fluorophenyl)piperazin-l-yl)-3-(4-fluoropheny l)prop-2-en-l-one (0.107 g, 39%) as a light yellow oil. Ή-NMR (300 MHz; CDCE): d 7.38 (dd, 7 = 8.6, 5.4 Hz, 2 H), 7.03 (t, 7 = 8.6 Hz, 2 H), 6.95- 6.91 (m, 2 H), 6.78 (dd, 7 = 7.7, 1.9 Hz, 1 H), 6.68 (d, 7 = 12.5 Hz, 1 H), 6.05 (d, 7 = 12.5 Hz, 1 H), 3.83 (t, 7 = 5.1 Hz, 2 H), 3.51 (t, 7 = 5.1 Hz, 2 H), 3.02 (t, 7 = 5.1 Hz, 2 H), 2.71 (t, 7 = 5.1 Hz, 2 H).

To a solution of anhydrous CrCh (0.169 g, 1.37 mmol) in THF (2.29 mL) that was degassed by sparging with Ar for 30 min followed by the addition of (Z)-l-(4-(5-chloro-2-fluorophenyl)piperazin-l-yl)- 3-(4-fluorophenyl)prop-2-en-l-one (83.0 mg, 0.229 mmol) and CH2ICI (0.132 mL, 1.14 mmol) at RT and under Ar atmosphere. The reaction mixture was stirred at 80 °C. After stirring for 14 h, the mixture was cooled to RT and quenched by the addition of 1.0 M aqueous HC1 (10 mL) and extracted with EtOAc (3x20 mL). The combined organic layer was concentrated, then the residue was diluted in EtOAc (3 mL) and the solution was filtered through a plug of basic alumina, and concentrated. The crude material was purified by chromatography on S1O2 (EtO Ac/hexanes, 1 : 1) to afford (4-(5-chloro-2-fluorophenyl)piperazin- l-yl)(2-(4-fluorophenyl)cyclopropyl)methanone as a white crystal (67 mg, 78 %): Mp: 108-109 °C; IR (neat): 1638, 1606, 1512, 1498, 1436, 1230, 1209, 1032 cm 1 ; 'H-NMR (400 MHz; CDCE): d 7.12 (dd, 7 = 8.5, 5.5 Hz, 2 H), 6.97-6.88 (m, 4 H), 6.72 (dd, 7 = 7.3, 1.9 Hz, 1 H), 3.79 (d, 7 = 12.8 Hz, 1 H), 3.69 (t, 7 = 17.1 Hz, 2 H), 3.39 (t, 7 = 9.5 Hz, 1 H), 2.99 (t, 7 = 13.9 Hz, 2 H), 2.52-2.42 (m, 2 H), 2.40-2.34 (m, 1 H), 2.20-2.15 (m, 1 H), 1.84-1.80 (m, 1 H), 1.37-1.32 (m, 1 H). 13 C-NMR (125 MHz; CDC1 3 ): d 167.2 , 161.6 (d, 7 = 245.2 Hz), 154.1 (d, 7 = 246.1 Hz), 140.4 (d, 7 = 9.9 Hz), 133.0 (d, 7 = 2.8 Hz), 129.4 (d, 7 = 3.3 Hz), 129.0 (d, 7 = 7.5 Hz), 122.4 (d, 7 = 8.1 Hz), 119.3 (d, 7 = 2.8 Hz), 117.1 (d, 7 = 22.7 Hz), 115.0 (d, 7 = 21.2 Hz), 50.6, 50.1, 45.1, 41.7, 23.7, 23.5, 10.6. HRMS (ESI): m/z calculated for C20H20N2OCIF2 ([M+H] + ) 377.1227, found 377.1221 .

(4-Cyclohexylpiperazin-l-yl)(2-(4-(trifluoromethyl)phenyl)cy clopropyl)methanone. To a solution of piperazin-l-yl(2-(4-(trifluoromethyl)phenyl)cyclopropyl)meth anone hydrochloride (0.09 g, 0.269 mmol) and cyclohexanone (0.0279 mL, 0.269 mmol) in dichloromethane (1.00 mL) was added NaBH(OAc)3 (0.256 g, 1.21 mmol) and acetic acid (0.0154 mL, 0.269 mmol). The mixture was stirred for 45 h at room temperature. Then the mixture was quenched with IN aqueous NaOH (40 mL) and extracted with EtOAc (40 mLx3). The combined organic phase was dried (Na 2 S0 4 ), filtered and concentrated in vacuo. The crude product was purified by chromatography on S1O2 (MeOH/CtLCL, 1 :9) to yield (4-cyclohexylpiperazin-l-yl)(2-(4- (trifluoromethyl)phenyl)cyclopropyl)methanone (0.0883 g, 86 %) as a light yellow solid: Mp: 99-101 °C; IR (neat): 2933, 1634, 1618, 1468, 1439, 1325, 1161, 1116, 1070, 1017 cm 1 ; Ή-NMR (500 MHz; CDCL): d 7.49 (d, J = 8.2 Hz, 2 H), 7.24 (d, J = 8.2 Hz, 2 H), 3.78 (d, J = 12.8 Hz, 1 H), 3.58 (d, J = 12.8 Hz, 1 H), 3.38 (ddd, J = 12.5, 9.4, 3.0 Hz, 1 H), 3.06 (ddd, / = 12.5, 9.4, 3.0 Hz, 1 H), 2.47-2.41 (m, 3 H), 2.22 (td, / = 8.8, 6.3 Hz, 1 H), 2.09 (tt, / = 11.4, 3.1 Hz, 1 H), 1.87-1.82 (m, 2 H), 1.73 (d, / = 12.8 Hz, 2 H), 1.61-1.57 (m, 4 H), 1.39 (td, / = 8.4, 5.6 Hz, 1 H), 1.19-1.10 (m, 2 H), 1.06-0.94 (m, 3 H); 13 C-NMR (125 MHz; CDCL): d 166.3, 142.2, 128.5 (q, / = 32.4 Hz), 127.8, 124.9 (q, J = 3.9 Hz), 124.2 (q, / = 270.0 Hz), 63.5, 49.2, 48.4, 45.6, 42.3, 28.6, 28.5, 26.2, 25.8, 24.8, 23.8, 11.2; 19 F-NMR (471 MHz; CDCL): d -62.43 (s, 3 F); HRMS (ESI): m/z calculated for C23H20F9ON2 ([M+H] + ) 381.2154, found 381.2147.

(4-(Tetrahydro-2H-pyran-4-yl)piperazin-l-yl)(2-(4-(trifluoro methyl)phenyl)cyclopropyl)methanone.

To a solution of piperazin-l-yl(2-(4-(trifluoromethyl)phenyl)cyclopropyl)meth anone hydrochloride (0.09 g, 0.269 mmol) and tetrahydro-4H-pyran-4-one (0.0269 mL, 0.269 mmol) in dichloromethane (1.00 mL) was added NaBH(OAc) 3 (0.205 g, 0.269 mmol) and acetic acid (0.0154 mL, 0.269 mmol). The mixture was stirred for 13 h at room temperature. Then the mixture was quenched with IN aqueous NaOH (30 mL) and extracted with CH2CI2 (30 mFx3). The combined organic phase was dried (Na2SOr), filtered and concentrated in vacuo. The crude product was purified by chromatography on S1O2 (MeOH:CH2CL = 1:9) to give (4-(tetrahydro-2H-pyran-4-yl)piperazin-l-yl)(2-(4-(trifluoro methyl)phenyl)cyclopropyl)methanone (0.0845 g, 82 %) as a light yellow solid: Mp: 84-86 °C; IR (neat): 2949, 2845, 1637, 1619, 1468, 1439,

1325, 1161, 1115, 1069 cm 1 ; 'H-NMR (500 MHz; CDCL): d 7.49 (d, / = 8.2 Hz, 2 H), 7.24 (d, / = 8.2 Hz,

2 H), 3.95 (dd, / = 11.5, 3.4 Hz, 2 H), 3.83 (d, / = 13.1 Hz, 1 H), 3.61 (d, / = 12.9 Hz, 1H), 3.37 (ddd, J = 12.7, 9.5, 3.1 Hz, 1 H), 3.28 (tt, / = 11.8, 2.8 Hz, 2 H), 3.04 (ddd, J = 12.7, 9.5, 3.1 Hz, 1 H), 2.47-2.43 (m, 3 H), 2.28-2.19 (m, 2 H), 1.87-1.78 (m, 2 H), 1.51-1.30 (m, 6 H). 13 C-NMR (125 MHz; CDCI3): d 166.3,

142.2, 128.5 (q, J = 32.4 Hz), 127.7, 125.0 (q, / = 3.9 Hz), 124.2 (q, / = 270.4 Hz), 67.4, 67.3, 60.8, 49.0, 48.6, 45.4, 42.1, 29.1, 28.9, 24.9, 23.8, 11.3; 19 F-NMR (471 MHz; CDCI3): d -62.37 (s, 3 F); HRMS (ESI): m/z calculated for C20H26F3O2N2 ([M+H] + ) 383.1946, found 383.1938.

(3,3-Difluoro-2-(4-(trifluoromethyl)phenyl)cycloprop-l-en-l- yl)(4-(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)methanone. Under an inert atmosphere, l-(4-(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)-3-(4-(trifluoromethyl)phenyl)prop-2-y n-l-one (0.400 g, 0.908 mmol),

TMSCF2Br (0.28 mL, 1.82 mmol), «Bu iNBr (14.6 mg, 0.0454 mmol), and toluene (3.6 mL) were added into an oven-dried pressure tube at room temperature. After being heated at 110 °C for 20 h, The reaction mixture was cooled to room temperature and poured into saturated NaHCCF solution (15 mL) and extracted with EtOAc (2 x 20 mL). The combined organic layers were dried over anhydrous Na2SC>4, filtered, and concentrated under reduced pressure. The crude product was subjected to flash column chromatography on S1O2 (1:9 EtO Ac/hexanes). The fractions collected contained -10% of impurities. The product was resubjected to flash column chromatography on S1O2 (1 :2 CH2Cl2/hexanes) to afford (3,3-difluoro-2-(4- (trifluoromethyl)phenyl)cycloprop-l-en-l-yl)(4-(2-methyl-5-( trifluoromethyl)phenyl)piperazin-l- yl)methanone (0.325 g, 0.663 mmol, 73%) as a pale yellow oil that foamed up upon drying under vacuum: IR (CDCI3) 2925, 2825, 1776, 1643, 1442, 1303, 1120, 1061, 1031, 909, 850, 730 cm 1 ; Ή NMR (CDC1 3 , 500 MHz) d 8.13 (d, / = 8.0 Hz, 2 H), 7.79 (d, / = 8.1 Hz, 2 H), 7.31 (q, / = 8.7 Hz, 2 H), 7.22 (s, 1 H), 3.94 (t, J = 4.7 Hz, 2 H), 3.88 (t, J = 5.0 Hz, 2 H), 3.04 (dt, J = 25.5, 5.0 Hz, 4 H), 2.41 (s, 3 H); 13 C NMR (CDCI3, 125 MHz) d 155.1, 150.9, 137.0, 135.5 (t, / = 10 Hz), 134.4 (q, / = 33 Hz), 132.6, 131.8, 129.4 (q, J = 32 Hz), 126.3 (q, / = 3.6 Hz), 126.0, 124.3 (q, / = 272 Hz), 123.6 (q, / = 273 Hz), 120.9 (q, / = 3.9 Hz), 119.7 (t, / = 13 Hz), 116.3 (q, J = 3.6 Hz), 98.8 (t, / = 278 Hz), 52.2, 51.5, 46.9, 42.6, 18.1; 19 F NMR (CDCE, 470 MHz) d -62.3 (s, 3 F), -63.2 (s, 3 F), -102.3 (s, 2 F) ; HRMS (ESI) m/z calcd for C2 3 H I9 ON2F 8 ([M+H] + ) 491.1364, found 491.1363.

c/s-((lSR,3RS)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cyd opropyl)(4-(2-methyl-5- (trifluoromethyl)phenyl)piperazin-l-yl)methanone. A solution of (3,3-difluoro-2-(4-(trifluoromethyl)- phenyl)cycloprop-l-en-l-yl)(4-(2-methyl-5-(trifluoromethyl)p henyl)piperazin-l-yl)methanone

(260 mg, 0.530 mmol) in EtOAc (4.6 mL) was added Pd/C (10% Pd on carbon, 56.4 mg, 10.0 mol%) . The reaction vessel was placed in the parr hydrogenator (7 bar) and stirred for 24 h at room temperature. The mixture was filtered through celite and concentrated in vacuo. The crude oil was then passed through a plug of silica gel to remove baseline impurities. The crude residue (270 mg) contained a mixture of the desired r/.v-product and ring-opening side products which was inseparable by normal phase column chromatography.

The crude racemic cis-((lSR,3RS)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cyc lopropyl)(4-(2-methyl- 5-(trifluoromethyl)phenyl)piperazin-l-yl)methanone was purified and separated on a SFC Chiralpak-IC semiprep (250 x 10 mm) column (15% /PrOH, 6 mL/min, 220 nM, P=100) to afford the (-)-enantiomer (45.1 mg, 0.0916 mmol, 17%) and (-t)-enantiomer (41.3 mg, 0.0839 mmol, 16%) respectively as a white solid: Mp 123.5 - 127.8 °C; IR (CDC1 3 ) 2917, 2820, 1648, 1416, 1323, 1308, 1115, 1070, 986, 856, 731 cm 1 .

(-)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cyclopropyl )(4-(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)methanone (retention time 7.52 min) was obtained as a white solid (99.5% purity by ESLD): [a] 20 D -32.1 (c 1.03, /PrOH); Ή NMR (CDCE, 500 MHz) d 7.60 (d, J = 8.3 Hz, 2 H), 7.44 (d, J = 8.1 Hz, 2 H), 7.28-7.24 (m, 4 H), 7.02 (s, 1 H), 3.80-3.77 (m, 1 H), 3.66-3.53 (m, 3 H), 3.13 (td, / = 12.6, 2.0 Hz, 1 H), 2.90 (td, / = 12.5, 2.4 Hz, 1 H), 2.83 (dtd, / = 11.5, 5.8, 3.4 Hz, 2 H), 2.61 (ddd, / = 11.2, 8.0, 3.0 Hz, 1 H), 2.39-2.33 (m, 1 H), 2.31 (s, 3 H); 19 F NMR (CDC1 3 , 470 MHz) d -62.4 (s, 3 F), -62.8 (s, 3 F), - 118.9 (d, J F~F — 161 Hz, 1 F), -147.2 (d, J F-F = 161 Hz, 1 F); HRMS (ESI) m/z calcd for C23H2iON 2 F 8 ([M+H] + ) 493.1521, found 493.1522. The enantiomeric excess was >99% ee (SFC Chiralpak-IC (250 x 10 mm); 15% /PrOH, 220 nm, 6 mF/min; retention time: 7.54 min).

(+)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cyclopropyl)(4 -(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)methanone (retention time 9.64 min) was obtained as a white solid (99.5% purity by ESFD): [a] 20 D +33.5 (c 0.60, /PrOH); Ή NMR (CDC1 3 , 500 MHz) d 7.60 (d, / = 8.2 Hz, 2 H), 7.44 (d, / = 8.1 Hz, 2 H), 7.28-7.24 (m, 5 H), 7.02 (s, 1 H), 3.81-3.76 (m, 1 H), 3.66-3.52 (m, 3 H), 3.13 (td, / = 12.6, 2.2 Hz, 1 H), 2.90 (td, / = 12.5, 2.6 Hz, 1 H), 2.83 (dtd, / = 11.5, 5.8, 3.3 Hz, 2 H), 2.61 (ddd, / = 11.4, 7.9, 3.0 Hz, 1 H), 2.39-2.33 (m, 1 H), 2.31 (s, 3 H); 13 C NMR (CDCI3, 126 MHz) d 160.94 (s, 1 C), 150.94 (s, 1 C), 136.92 (s, 1 C), 134.96 (s, 1 C), 131.67 (s, 1 C), 130.13 (q, J = 32.8 Hz, 1 C), 129.53 (d, / = 2.7 Hz, 1 C), 129.30 (q, / = 34.0 Hz, 1 C), 125.39 (q, J = 3.6 Hz, 1 C), 124.22 (q, J = 272.0 Hz, 1 C), 124.06 (q, / = 272.1 Hz, 1 C), 120.72 (q, J = 3.7 Hz, 1 C), 116.01 (q, J = 3.6 Hz, 1 C), 110.99 (t, / = 289.7 Hz, 1 C), 51.70 (s, 1 C), 51.49 (s, 1 C), 46.17 (s, 1 C), 42.12 (s, 1 C), 31.35 (dd, J = 12.9, 9.8 Hz, 1 C), 30.28 (dd, / = 10.4, 8.9 Hz, 1 C), 18.00 (s, 1 C). 19 F NMR (CDCE, 470 MHz) d -62.4 (s, 3 F), -62.8 (s, 3 F), -118.9 (d, / F -F = 161 Hz, 1 F), -147.2 (d, / F-F = 161 Hz, 1 F). HRMS (ESI) m/z calcd for C23H21ON2F8 ([M+H] + ) 493.1521, found 493.1522. The enantiomeric excess was >99% ee (SFC Chiralpak-IC (250 x 10 mm); 15% /PrOH, 220 nm, 6 mL/min; retention time: 9.66 min).

Example 6

Androgen Receptor and Estrogen Receptor Alpha Competitor Assays

The disclosed compounds are assayed for androgen receptor activity using the PolarScreen™ Androgen Receptor Competitor Assay Kit, Green (ThermoFisher Scientific, catalog #A15880). The kit uses rat AR- ligand binding domain tagged with gluthathione-S -transferase (GST) and histidine [AR-LBD(Hist- GST)] to determine the IC50 of competitive androgen receptor compounds. AR-LBD(His-GST) is added to fluorescently-tagged androgen ligand (FluormoneAL Green) in the presence of competitor test compounds. Effective competitors prevent the formation of a AL Green/ AR-LBD(His-GST) complex, resulting in a decrease of polarization due to ligand displacement by the competitor. The shift in polarization values is used to determine the IC50 of test compounds.

The disclosed compounds are assayed for estrogen receptor (ER) alpha activity using the

PolarScreen™ ER Alpha Competitor Asssay Kit, Green (ThermoFisher Scientific, catalog #A15883). The kit uses full length, native (untagged), human estrogen receptor alpha to determine the IC50 of competitive estrogen receptor compounds. Full length ER alpha is added to a fluorescent estrogen ligand (Fluormone ES2 Green) to form an ER-Fluormone ES2 complex. An effective ER alpha competitor will displace the Fluormone ES2 ligand from the ER alpha and will result in a decrease in polarization. The shift in fluorescence polarization is used to determine the relative affinity of test compounds for ER alpha.

In view of the many possible embodiments to which the principles of the disclosed compounds, compositions and methods may be applied, it should be recognized that the illustrated embodiments are only preferred examples and should not be taken as limiting the scope of the invention.