WO2017120328A1 - Hydantoin-derived histone deacetylase (hdac) inhibitors and methods of making and using thereof - Google Patents

Hydantoin-derived histone deacetylase (hdac) inhibitors and methods of making and using thereof Download PDF

Info

Publication number
WO2017120328A1
WO2017120328A1 PCT/US2017/012323 US2017012323W WO2017120328A1 WO 2017120328 A1 WO2017120328 A1 WO 2017120328A1 US 2017012323 W US2017012323 W US 2017012323W WO 2017120328 A1 WO2017120328 A1 WO 2017120328A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
hydrogen
compounds
zbg
heteroatoms
Prior art date
Application number
PCT/US2017/012323
Other languages
French (fr)
Inventor
Adegboyega K. Oyelere
Subhasish Tapadar
David GAUL
Original Assignee
Sophia Bioscience, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sophia Bioscience, Inc. filed Critical Sophia Bioscience, Inc.
Publication of WO2017120328A1 publication Critical patent/WO2017120328A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin

Definitions

  • HDAC HYDANTOIN-DERIVED HISTONE DEACETYLASE
  • the various embodiments of the disclosure relate generally to hydantoin-derived histone deacetylase (HDAC) inhibitors that target proliferative and hyperporoliferative diseases, including prostate tumors. More particularly, the disclosure describes a series of hydantoin-derived HDAC inhibitors equipped with androgen receptor binding affinity to target androgen positive prostate. The disclosure describes the methods for making and using thereof.
  • HDAC histone deacetylase
  • PCa Prostate cancer
  • RT external beam radiation therapy
  • brachytherapy interstitial RT
  • cryotherapy cryotherapy
  • ADT androgen deprivation therapy
  • ADT drugs in clinical use, either as monotherapy or combination therapy (with castration), include bicalutamide (Casodex ® ), nilutamide (Nilandron ® ), MDV3100 (Enzalutamide) and flutamide (Eulexin ® ).
  • PCas respond well to ADT and other available therapies.
  • tumors inevitably enter an antiandrogen-resistant (i.e. castration-resistant or hormone refractory) state and subsequently exhibit chemotherapy- 1 resistance. This castration-resistant state is incurable, and the median survival following this period is just 18–24 months (Eisenberger, M. A.; Blumenstein, B.
  • the androgen receptor (AR) expression state is one of the key hallmarks of PCa sustenance and progression.
  • All PCas (early-stage and CRPC) overexpress AR with CRPC showing higher expression than the androgen-dependent early-stage tumors or benign prostate hyperplasias (Linja, M. J.; Savinainen, K. J.; Saramaki, O. R.; Tammela, T. L.; et al. Amplification and Overexpression of Androgen Receptor Gene in Hormone-Refractory Prostate Cancer. Cancer Res 2001, 61, 3550–3555).
  • most mechanistic drivers of CRPC phenotype target the AR pathway.
  • the validated drivers of the CRPC phenotype are (i) tumor expression of promiscuous mutant AR which are activated, in addition to the natural ligands testosterone (T) and dihydrotestoterone (DHT), by other steroid hormones such as progesterone, estradiol, and cortisol, (ii) tumor expression of constitutively active AR splice variants lacking the ligand binding domain, and (iii) adaptive intratumoral androgen biosynthesis powered by adrenal androgens such as DHEA-SO4, DHEA and 4- androstene-3,17-dione ( ⁇ 4 -AD) (Yuan, X.; Balk, S. P. Mechanisms Mediating Androgen Receptor Reactivation After Castration.
  • Dehydroepiandrosterone activates mutant androgen receptors expressed in the androgen-dependent human prostate cancer xenograft CWR22 and LNCaP cells. Mol Endocrinol. 1997, 11, 450–459. Knudsen, K.; Scher, H. I. Starving the addiction: New opportunities for durable suppression of AR signaling in prostate cancer. Clin. Cancer Res. 2009, 15, 4792-4798. Knudsen, K.; Penning, T. M. Partners in crime: deregulation of AR activity and androgen synthesis in prostate cancer. Trends Endocrinol. Metab. 2010, 21, 315-324. Yu, Z.; Chen, S.; Sowalsky, A. G.; Voznesensky, O.
  • Enza is not well tolerated by some patients due to dose limiting CNS toxicities (Foster, W. R.; Car, B. D.; Shi, H.; Levesque, P. C.; Obermeier, M. T.; Gan, J.; Arezzo, J. C.; et al. Drug safety is a barrier to the discovery and development of new androgen receptor antagonists. Prostate 2011, 71, 480-488.); however, ARN-509, an Enza-derived next generation super-AR antagonist, is devoid of CNS toxicity and is in a phase III clinical trial (Clegg, N.
  • PCa with adaptive androgen biosynthesis is targeted with a combination of P450c17 (17 ⁇ -hydroxylase/17,20-lyase) inhibitors, such as abiraterone acetate (Abi) and prednisone (Attard, G.; Reid, A. H. M.; Yap, T.A.; Raynaud, F.; Dowsett, M.; Settatree, S.; et al. Phase 1 clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. J. Clin. Oncol. 2008, 28, 4563-4571. Danila, D. C., Morris, M. J., de Bono, J.
  • P450c17 inhibitors such as Galeterone (TOK-001) (Vasaitis, T.; Belosay, A.; Schayowitz, A.; Khandelwal, A.; Chopra, P.; Gediya, L.K.; Guo, Z.; Fang, H. B.; Njar, V. C.; Brodie, A. M. Androgen receptor inactivation contributes to antitumor efficacy of 17 ⁇ -hydroxylase/17,20-lyase inhibitor 3 ⁇ -hydroxy-17-(1H-benzimidazole-1- yl)androsta-5,16-diene in prostate cancer. Mol. Cancer Therap. 2008, 7, 2348-2357.
  • Toren PJ Kim S, Pham S, Mangalji A, Adomat H, Guns ES, Zoubeidi A, Moore W, Gleave ME.
  • Anticancer activity of a novel selective CYP17A1 inhibitor in preclinical models of castrate-resistant prostate cancer. Mol. Cancer Ther. 2015, 14, 59–69.) which selectively inhibit the 17,20-lyase activity of P450c17 are being developed as well. Observations from the use of these agents in the clinic, both alone and in combination, have clearly indicated the need for better agents able to treat CRPC.
  • HDAC histone deacetylase
  • An embodiment of the disclosure can be a compound of Formula II,
  • An embodiment of the disclosure can be a method of treating a proliferative disorder by administering a compound of Formula II.
  • the disclosure for the compound of or method using Formula II can include W selected from O or S; X selected from C-H or N; Y selected from cyano or nitro; Z selected from trifluoromethyl or iodo; R 1 and R 2 independently selected from hydrogen, C 1 to C 8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C 3 to C 6 cycloalkyl or substituted cycloalkyl group; R 3 selected from hydrogen, fluorine, or CF 3 , preferably hydrogen or fluorine; and n as 0 to 6, and when greater than 0 represents C 1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement.
  • the disclosure for the compound of or method using Formula II can include ZBG that is a Zinc Binding Group selected from
  • m can be 0 to 7, and when m is greater than 0 represents a C 1 to C 7 group, optionally can contain one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement.
  • R 4 can be hydrogen or methyl.
  • R 5 can be hydrogen, fluoro, phenyl, or 4-pyridyl.
  • R 6 can be hydrogen or a salt counterion.
  • R 7 can be a C 1 to C 6 alkyl or acyl.
  • R 8 and R 9 can each independently be hydrogen or C 1 to C 6 alkyls.
  • the disclosure for the compound of or method using Formula II can include n as at least 1, or n equal to 1.
  • Y can be is cyano, Z can be trifluoromethyl, or Y can be cyano and Z can be trifluoromethyl.
  • X can be C-H or N, Y can be cyano, Z can be trifluoromethyl, and n can be greater than 1.
  • the disclosure for the compound of or method using Formula II can include ZBG as
  • ZBG can include .
  • ZBG can include
  • the disclosure for the compound of or method using Formula II can include X as H or N; Y as cyano; Z as trifluoromethyl; n equal to 1; and ZBG is selected from
  • Figure 1A illustrates three exemplary compounds, in accordance with exemplary embodiments of the disclosure.
  • Figure 1B illustrates intracellular activity of two compounds of Figure 1A, in accordance with exemplary embodiments of the disclosure.
  • Figure 1C illustrates an exemplary synthetic scheme for the three compounds of Fig.1A, in accordance with an exemplary embodiment of the disclosure.
  • Figure 2 illustrates exemplary structures 30-146, in accordance with exemplary embodiments of the disclosure.
  • Figures 3A-C illustrate comparative chemical compounds and activities, in accordance with an exemplary embodiment of the disclosure.
  • Figures 4A-C illustrate activity of comparative chemical compounds and intracellular target validation of comparative compounds, in accordance with an exemplary embodiment of the disclosure.
  • Figure 5 illustrates androgen receptor antagonist activity of comparative chemical compounds, in accordance with an exemplary embodiment of the disclosure.
  • Figure 6A illustrates the structures of comparative compounds and Figure 6B illustrates the stability and clearance of the compounds in Figure 6A, in accordance with an exemplary embodiment of the disclosure.
  • Figure 7 illustrates an in vivo efficacy study of comparative compounds, in accordance with an exemplary embodiment of the disclosure.
  • Figures 8A illustrates a toxicity study in healthy mice of comparative compounds
  • Figures 8B and 8C illustrate in vivo efficacy studies of comparative compounds, in accordance with an exemplary embodiment of the disclosure.
  • Ranges can be expressed herein as from“about” or“approximately” one particular value and/or to“about” or“approximately” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value.
  • Ar represents an aryl group.“Aryl”, as used herein, refers to 5-, 6- and 7- membered aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic, or bihetereocyclic ring system, optionally substituted by halogens, alkyl-, alkenyl-, and alkynyl-groups.
  • “Ar” includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • Those aryl groups having heteroatoms in the ring structure may also be referred to as“aryl heterocycles” or “heteroaromatics”.
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties,—CF 3 ,—CN, or the like.
  • substituents as described above, for example, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfony
  • the term“Ar” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are“fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocycles.
  • heterocyclic ring examples include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2- dithiazinyl, dihydrofuro [2,3b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl,
  • “Zinc binding group” or“ZBG”, as used herein, refers to moieties capable of inhibiting zinc metalloenzymes activity including HDAC. Suitable examples include, but are not limited to, hydroxamates, N-formyl hydroxylamine (or retro-hydroxamate), carboxylates, thiols, dithiols, trithiocarbonates, thioesters, benzamide, keto, mercaptoacetamides, 2-ketoamides, epoxides, epoxyketones, trifluoromethyl ketones, hydroxypyridinones, pyrones, hydroxylpyridinethiones, and thiopyrones.
  • Alkyl refers to the radical of saturated or unsaturated aliphatic groups, including straight-chain alkyl, alkenyl, or alkynyl groups, branched-chain alkyl, alkenyl, or alkynyl groups, cycloalkyl, cycloalkenyl, or cycloalkynyl (alicyclic) groups, alkyl substituted cycloalkyl, cycloalkenyl, or cycloalkynyl groups, and cycloalkyl substituted alkyl, alkenyl, or alkynyl groups.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone.
  • C1 to C4 or C6 linear alkyl preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure, and optionally contain or are substituted with heteroatoms, such as with 1, 2, 3 or more independently selected oxygen, nitrogen, or sulfur atoms.
  • Alkylaryl refers to an alkyl group substituted with an aryl group (e.g., an aromatic or hetero aromatic group).
  • “Acyl” or“acyl group” as used herein is intended to mean a—C(O)—R radical, where R is a suitable substituent (for example, an acetyl group, a propionyl group, a butyroyl group, a benzoyl group, or an alkylbenzoyl group).
  • R is a suitable substituent (for example, an acetyl group, a propionyl group, a butyroyl group, a benzoyl group, or an alkylbenzoyl group).
  • “Heterocycle” or“heterocyclic”, as used herein, refers to a cyclic radical attached via a ring carbon or nitrogen of a monocyclic or bicyclic ring containing 3-10 ring atoms, and preferably from 5-6 ring atoms, consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y) wherein Y is absent or is H, O, phenyl or benzyl, and optionally containing 1-3 double bonds and optionally substituted with one or more substituents.
  • heterocyclic ring examples include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH- carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H- 1,5,2-dithiazinyl, dihydrofuro [2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizin
  • Heteroaryl refers to a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and 1, 2, 3, or 4 heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y) where Y is absent or is H, O, (C 1 -C 8 ) alkyl, phenyl or benzyl.
  • heteroaryl groups include furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyrazolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl, (or its N-oxide), thienyl, pyrimidinyl (or its N- oxide), indolyl, isoquinolyl (or its N-oxide), quinolyl (or its N-oxide) and the like.
  • heteroaryl can include radicals of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto.
  • heteroaryl can be furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyraxolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl (or its N-oxide), thientyl, pyrimidinyl (or its N-oxide), indolyl, isoquinolyl (or its N-oxide), quinolyl (or its N- oxide), and the like.
  • Halogen refers to fluorine, chlorine, bromine, or iodine.
  • alkenyl and“alkynyl” refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively.
  • 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
  • “Pharmaceutically acceptable salt”, as used herein, refer to derivatives of the compounds defined by Formula I wherein the parent compound is modified by making acid or base salts thereof.
  • Example of pharmaceutically acceptable salts include but are not limited to mineral or organic acid salts of basic residues such as amines; and alkali or organic salts of acidic residues such as carboxylic acids.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • Such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric acids; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, naphthalenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic salts.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric acids
  • organic acids such as acetic, propionic, succinic, glycolic,
  • the pharmaceutically acceptable salts of the compounds can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 20th ed., Lippincott Williams & Wilkins, Baltimore, Md., 2000, p. 704; and“Handbook of Pharmaceutical Salts: Properties, Selection, and Use,” P. Heinrich Stahl and Camille G. Wermuth, Eds., Wiley-VCH, Weinheim, 2002.
  • “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • “Prodrug”, as used herein, refers to a pharmacological substance (drug) which is administered in an inactive (or significantly less active) form. Once administered, the prodrug is metabolized in the body (in vivo) into the active compound.
  • “Solvate”, as used herein, refers to a compound which is formed by the interaction of molecules of a solute with molecules of a solvent.
  • “Reverse ester”, as used herein, refers to interchange in the positions of the oxygen and carbon groups in a series of structurally related compounds.
  • “Reverse amide”, as used herein, refers to interchange in the positions of the nitrogen and carbon groups in a series of structurally related compounds.
  • AR pathway constitutes a viable target to develop new generation of novel, targeted agents against PCas even after the establishment of the CRPC phenotype.
  • Two possible approaches toward new agents against PCas are: (i) development of anti- androgens with stronger AR binding affinities, (ii) redesign of anti-androgens, including but not limiting to the more potent diarylhydantions, to incorporate independent cancer inhibiting chemotype(s).
  • the later approach explores the tumor AR expression state to affect a selective delivery of an independent anti-tumor chemotype.
  • Histone deacetylase (HDAC) inhibiting pharmacophores ideally functions as independent cancer inhibiting chemotype(s) for incorporation to anti-androgen pharmacophores (US 9,139,565).
  • HDAC inhibitors (HDACi) have stimulated much enthusiasm in oncology recently with over 500 cancer clinical trials initiated to date, resulting in five clinically approved drugs: SAHA and FK228, approved by the FDA for the treatment of cutaneous T cell lymphoma; Belinostat and Epidaza, approved for peripheral T cell lymphoma; and Panobinostat, approved for multiple myeloma. Despite their success in blood malignancies, current HDACi have serious limitations in solid tumors.
  • HDAC inhibition elicits a pleiotropic phenotype which is largely due to their nonselective inhibition of various cellular HDAC isoforms and possibly other non-HDAC targets as well.
  • This broad HDAC inhibition is associated with reduced in vivo potency and toxic side effects.
  • An approach that selectively target HDAC inhibitors to the diseased cells could improve their therapeutic indices.
  • Ar represents an aryl group
  • W is O or S
  • X is C-H or N
  • Y is cyano or nitro
  • Z is trifluoromethyl or iodo
  • R 1 and R 2 are independently selected from hydrogen, C 1 to C 8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C 3 to C 6 cycloalkyl or substituted cycloalkyl group;
  • n is 0 to 6, and when greater than 0 represents C 1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement;
  • ZBG is a Zinc Binding Group selected from
  • n is 0 to 7, and when m is greater than 0 represents a C 1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
  • R 4 is hydrogen or a C 1 to C 6 alkyl
  • R 5 is hydrogen, fluoro, phenyl, or 4-pyridyl
  • R 6 is hydrogen or a salt counterion
  • R 7 is a C 1 to C 6 alkyl or acyl
  • R 8 and R 9 are each independently hydrogen or C 1 to C 6 alkyls.
  • Aryl includes 5-, 6- and 7-membered aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic, or bihetereocyclic ring system, optionally substituted by halogens, alkyl-, alkenyl-, and alkynyl-groups.
  • “Ar” includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • Those aryl groups having heteroatoms in the ring structure may also be referred to as“aryl heterocycles” or“heteroaromatics”.
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, --CF 3 , --CN, or the like.
  • substituents as described above, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether,
  • Ar also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are“fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocycles.
  • heterocyclic ring examples include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H- 1,5,2-dithiazinyl, dihydrofuro[2,3 b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indoliziny
  • the hydrogen atom positions of the alkene bond bridging the Ar and the ZBG can be isotopically modified to include one or two of deuterium and/or tritium. In some embodiments, both the hydrogen atom positions can be hydrogen. In some embodiments, at least one of the hydrogen atom positions can be isotopically modified to deuterium or tritium, preferably deuterium.
  • the compounds of Formula II can be used in HDAC inhibition.
  • W is O or S
  • X is C-H or N
  • Y is cyano or nitro
  • Z is trifluoromethyl or iodo
  • R 1 and R 2 are independently selected from hydrogen, C 1 to C 8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C 3 to C 6 cycloalkyl or substituted cycloalkyl group;
  • R 3 is hydrogen, fluorine, or CF 3
  • n is 0 to 6, and when greater than 0 represents C 1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement;
  • ZBG is a Zinc Binding Group selected from
  • n is 0 to 7, and when m is greater than 0 represents a C 1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
  • R 4 is hydrogen or a C 1 to C 6 alkyl
  • R 5 is hydrogen, fluoro, phenyl, or 4-pyridyl
  • R 6 is hydrogen or a salt counterion
  • R 7 is a C 1 to C 6 alkyl or acyl
  • R 8 and R 9 are each independently hydrogen or C 1 to C 6 alkyls.
  • W can be O or S.
  • W can also be oxygen; or W can be sulfur.
  • X can be C-H or N.
  • X can be C-H to establish the substituted phenyl ring.
  • X can also be N, to establish the substituted pyridyl ring.
  • Y can be cyano or nitro, and Z can be trifluoromethyl or iodo.
  • Y can be cyano and Z can be trifluoromethyl; Y can be cyano and Z can be iodo; Y can be nitro and Z can be trifluoromethyl; or Y can be nitro and Z can be iodo.
  • R 1 and R 2 can be independently selected from hydrogen, C 1 to C 8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C 3 to C 6 cycloalkyl or substituted cycloalkyl group.
  • R 1 and R 2 can both be a short chain C 1 to C 4 alkyl.
  • R 1 and R 2 can both be methyl.
  • R 1 and R 2 can be taken together with the carbon they are attached to form a C 3 to C 6 cycloalkyl ring.
  • R 1 and R 2 can be taken together with the carbon they are attached can for a cyclopropyl ring, a cyclobutyl ring, a cyclopentyl ring or a cyclohexyl ring.
  • the cycloalkyl ring can be optionally substituted.
  • the cycloalkyl ring can be cyclopropyl, cyclobutyl, or cyclopentyl, preferably be cyclopropyl or cyclobutyl.
  • R 3 can be hydrogen, halo, or CF 3 ; or hydrogen, fluorine or CF 3 .
  • R 3 can be preferably hydrogen or fluorine.
  • R 3 can be at either of the 2 and 3 position of the aryl ring when R 3 is not hydrogen.
  • n is 0 to 6, and when greater than 0 represents C 1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement.
  • n is at least 1, i.e. n is 1 to 6. In some embodiments, n can be 1.
  • the hydrogen atom positions of the alkene bond bridging the Ar and the ZBG can be isotopically modified to include one or two of deuterium or tritium. In some embodiments, both the hydrogen atom positions be hydrogen. In some embodiments, at least one of the hydrogen atom positions can be isotopically modified to deuterium or tritium, preferably deuterium.
  • Zinc binding group refers to moieties capable of inhibiting zinc metalloenzymes activity including HDAC and matrix metalloproteinase (MMP) activity. Suitable examples include, but are not limited to, hydroxamates, N- formyl hydroxylamine (or retro-hydroxamate), carboxylates, thiols, dithiols, trithiocarbonates, thioesters, benzamide, keto, mercaptoacetamides, 2-ketoamides, epoxides, epoxyketones, trifluoromethyl ketones, hydroxypyridinones, pyrones, hydroxylpyridinethiones, and thiopyrones.
  • hydroxamates N- formyl hydroxylamine (or retro-hydroxamate)
  • carboxylates include, but are not limited to, hydroxamates, N- formyl hydroxylamine (or retro-hydroxamate), carboxylates, thiols, dithiol
  • zinc binding groups include, but are not limited to, hydroxamic acids, thiol-hydroxamic acids, n-formyl hydroxylamines, carboxylates, nitro, thiols, dithiol mercaptoacetamies trithiocarbonates,thioesters, benzamides, epoxides, epoxyketones, trifluoromethyl ketones, ketones, and ketoamides.
  • Exemplary ZBGs can also be found in U.S. Patent No. 9,139,565, incorporated herein by reference in its entirety.
  • ZBG can be a zinc binding group selected from
  • m can be 0 to 7, and when m is greater than 0, can represent a C 1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement.
  • m can be 1 to 4, and can represent a C 1 to C 4 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement.
  • R 4 can be hydrogen or a C 1 to C 6 alkyl, hydrogen or C 1 to C 4 alkyl, or hydrogen and methyl.
  • R 5 can be hydrogen, fluoro, phenyl, or 4-pyridyl.
  • R 6 can be hydrogen or a salt counterion, e.g. a sodium salt, a potassium salt, an ammonium or a typical organic salt counterion, such as an alkyl ammonium salt with one to four carbon groups on the ammonium.
  • R 7 is a C 1 to C 6 alkyl or acyl.
  • R 8 and R 9 are each independently hydrogen or C 1 to C 6 alkyls.
  • the ZBG can be any organic compound [0061] in some preferred embodiments.
  • R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and m are as noted above.
  • the ZBG can be any organic compound
  • the ZBG can be
  • the compounds of Formula III can be used.
  • X, Y, Z, R 1 , R 2 , R 3 , n and ZBG are as defined above.
  • X can be C-H or N; Y is cyano; Z is trifluoromethyl; n is greater than 1, and ZBG is selected from
  • the compound of Formula II can also include X as C-H or N; Y as cyano; Z as trifluoromethyl; and n equal to 1. Furthermore, ZBG can be selected from
  • the compound of Formula II can also include ZBG selected from
  • the compounds of Formulas I, II, and III can be used to treat and/or prevent proliferative or hyperproliferative disorders.
  • the disclosure includes a method for treating these disorders administering a compound of Formula I, II or III, as disclosed and described above, and incorporated herein.
  • the disorder can include prostate cancer, including hormone sensitive and hormone refractory prostate cancers.
  • Compounds of this disclosure and related compounds in U.S. Patent No. 9,139,565, incorporated by reference herein in its entirety, have been demonstrated to effect inhibition in histone deacetylase (HDAC.) Cinnamoyl derivatives and aryl alkenyl (e.g. styrenyl) structures as disclosed herein have shown a particular and surprising efficacy.
  • HDAC histone deacetylase
  • the compounds of Formulas I, II, and III can be administered as a pharmaceutically acceptable salt, prodrug, or solvate.
  • the compounds described herein can be formulated with a pharmaceutically acceptable carrier and, optionally one or more pharmaceutically acceptable excipients, for enteral or parenteral administration, as further detailed below.
  • compositions of the disclosure can comprise a carrier and/or excipient. While it is possible to use a compound of the present disclosure for therapy as is, it may be preferable to administer it in a pharmaceutical formulation, e.g., in admixture with a suitable pharmaceutical excipient and/or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • a suitable pharmaceutical excipient and/or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the excipient and/or carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Acceptable excipients and carriers for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington: The Science and Practice of Pharmacy. Lippincott Williams & Wilkins (A.R. Gennaro edit.2005).
  • compositions readily accommodate additional mixtures, such as, e.g., milk, yogurt, and infant formula.
  • Solid dosage forms for oral administration can also be used and can include, e.g., capsules, tablets, caplets, pills, troches, lozenges, powders, and granules.
  • Non-limiting examples of suitable excipients include, e.g., diluents, buffering agents (e.g., sodium bicarbonate, infant formula, sterilized human milk, or other agents which allow bacteria to survive and grow [e.g., survive in the acidic environment of the stomach and to grow in the intestinal environment]), preservatives, stabilizers, binders, compaction agents, lubricants, dispersion enhancers, disintegration agents, antioxidants, flavoring agents, sweeteners, and coloring agents, including hydroxypropyl- ⁇ -cyclodextrin (CD), Solutol HS 15 (a PEG15-hydroxystearate) & Cremophor RH 40 (a PEG-40 Hydrogenated Castor Oil).
  • buffering agents e.g., sodium bicarbonate, infant formula, sterilized human milk, or other agents which allow bacteria to survive and grow [e.g., survive in the acidic environment of the stomach and to grow in the intestinal environment]
  • preservatives e
  • the composition is formulated for delivery by a route such as, e.g., oral, topical, rectal, mucosal, sublingual, nasal, naso/oro-gastric gavage, parenteral, intraperitoneal, intradermal, transdermal, intrathecal, nasal, and intracheal administration.
  • the composition is in a form of a liquid, foam, cream, spray, powder, or gel.
  • the composition comprises a buffering agent (e.g., sodium bicarbonate, infant formula or sterilized human milk).
  • the composition is formulated for oral delivery.
  • Administration of the compounds and compositions in the methods of the disclosure can be accomplished by any method known in the art.
  • useful routes of delivery include oral, rectal, fecal (by enema), and via naso/oro-gastric gavage, as well as parenteral, intraperitoneal, intradermal, transdermal, intrathecal, nasal, and intracheal administration.
  • the active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.
  • the carrier material should be non-toxic to the bacteria and the subject/patient.
  • the active ingredient(s) can be administered in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions.
  • the active component(s) can be encapsulated in gelatin capsules together with inactive ingredients and powdered carriers, such as glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium saccharin, talcum, magnesium carbonate.
  • inactive ingredients examples include red iron oxide, silica gel, sodium lauryl sulfate, titanium dioxide, and edible white ink.
  • Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric-coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • the active ingredient(s) are compounded and administered in a single tablet or capsule.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • Solutions or suspensions can include any of the following components, in any combination: a sterile diluent, including by way of example without limitation, water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent; antimicrobial agents, such as benzyl alcohol and methyl parabens; antioxidants, such as ascorbic acid and sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetates, citrates and phosphates; and agents for the adjustment of tonicity, such as sodium chloride or dextrose.
  • a sterile diluent including by way of example without limitation, water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent
  • antimicrobial agents such as benzyl alcohol and methyl parabens
  • antioxidants such as ascorbic acid and sodium bisul
  • solubilizing agents may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using co-solvents, such as, e.g., dimethylsulfoxide (DMSO), ethanol, dimethylacetamide or water, using surfactants, such as TWEEN ® 80, or dissolution in aqueous sodium bicarbonate.
  • co-solvents such as, e.g., dimethylsulfoxide (DMSO), ethanol, dimethylacetamide or water
  • surfactants such as TWEEN ® 80
  • dissolution in aqueous sodium bicarbonate such as sodium bicarbonate.
  • Pharmaceutically acceptable derivatives of the agents may also be used in formulating effective pharmaceutical compositions.
  • the composition can contain along with the active agent, for example and without limitation: a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose; a lubricant, such as magnesium stearate, calcium stearate and talc; and a binder such as starch, natural gums, such as gum acacia gelatin, glucose, molasses, polyvinylpyrrolidone, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to those of skill in the art.
  • a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose
  • a lubricant such as magnesium stearate, calcium stearate and talc
  • a binder such as starch, natural gums, such as gum acacia gelatin, glucose, molasses, polyvinylpyrrolidone, celluloses and derivatives thereof, povidone
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active agent as defined above and optional pharmaceutical adjuvants in a carrier, such as, by way of example and without limitation, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a carrier such as, by way of example and without limitation, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, such as, by way of example and without limitation, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, such as, by way of example and without limitation, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, such as, by way of example and without limitation, acetate, sodium citrate, cyclodextr
  • the active agents or pharmaceutically acceptable derivatives may be prepared with carriers that protect the agent against rapid elimination from the body, such as time release formulations or coatings.
  • the compositions may include other active agents to obtain desired combinations of properties.
  • Oral pharmaceutical dosage forms include, by way of example and without limitation, solid, gel and liquid.
  • Solid dosage forms include tablets, capsules, granules, and bulk powders.
  • Oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated.
  • Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art.
  • the formulations are solid dosage forms, such as capsules or tablets.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or agents of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
  • binders include, by way of example and without limitation, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose, and starch paste.
  • Lubricants include, by way of example and without limitation, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • Diluents include, by way of example and without limitation, lactose, sucrose, starch, kaolin, salt, mannitol, and dicalcium phosphate.
  • Glidants include, by way of example and without limitation, colloidal silicon dioxide.
  • Disintegrating agents include, by way of example and without limitation, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • Coloring agents include, by way of example and without limitation, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate.
  • Sweetening agents include, by way of example and without limitation, sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors.
  • Flavoring agents include, by way of example and without limitation, natural flavors extracted from plants such as fruits and synthetic blends of agents which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
  • Wetting agents include, by way of example and without limitation, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene laural ether.
  • Emetic- coatings include, by way of example and without limitation, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
  • Film coatings include, by way of example and without limitation, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
  • the agent could be provided in a composition that protects it from the acidic environment of the stomach.
  • the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active agent in the intestine.
  • the composition may also be formulated in combination with an antacid or other such ingredient.
  • the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as fatty oil.
  • dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • the agents can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like.
  • a syrup may contain, in addition to the active agents, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics.
  • Pharmaceutically acceptable carriers included in tablets are binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, and wetting agents.
  • Enteric-coated tablets because of the enteric-coating, resist the action of stomach acid and dissolve or disintegrate in the neutral or alkaline intestines.
  • Sugar-coated tablets are compressed tablets to which different layers of pharmaceutically acceptable substances are applied.
  • Film-coated tablets are compressed tablets which have been coated with a polymer or other suitable coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned.
  • Coloring agents may also be used in the above dosage forms.
  • Flavoring and sweetening agents are used in compressed tablets, sugar-coated, multiple compressed and chewable tablets. Flavoring and sweetening agents are useful in the formation of chewable tablets and lozenges.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Aqueous solutions include, for example, elixirs and syrups.
  • Emulsions are either oil-in-water or water-in-oil.
  • Elixirs are clear, sweetened, hydroalcoholic preparations.
  • Pharmaceutically acceptable carriers used in elixirs include solvents. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative.
  • An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid.
  • Pharmaceutically acceptable carriers used in emulsions are non- aqueous liquids, emulsifying agents and preservatives.
  • Suspensions use pharmaceutically acceptable suspending agents and preservatives.
  • Pharmaceutically acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form include diluents, sweeteners and wetting agents.
  • Pharmaceutically acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form include organic acids and a source of carbon dioxide. Coloring and flavoring agents may be used in any of the above dosage forms.
  • Solvents include, by way of example and without limitation, glycerin, sorbitol, ethyl alcohol and syrup.
  • preservatives include, without limitation, glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol.
  • Non-aqueous liquids utilized in emulsions include, by way of example and without limitation, mineral oil and cottonseed oil.
  • Emulsifying agents include, by way of example and without limitation, gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • Suspending agents include, by way of example and without limitation, sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia.
  • Diluents include, by way of example and without limitation, lactose and sucrose.
  • Sweetening agents include, by way of example and without limitation, sucrose, syrups, glycerin and artificial sweetening agents such as saccharin.
  • Wetting agents include, by way of example and without limitation, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
  • Organic acids include, by way of example and without limitation, citric and tartaric acid.
  • Sources of carbon dioxide include, by way of example and without limitation, sodium bicarbonate and sodium carbonate.
  • Coloring agents include, by way of example and without limitation, any of the approved certified water soluble FD and C dyes, and mixtures thereof.
  • Flavoring agents include, by way of example and without limitation, natural flavors extracted from plants, such as fruits, and synthetic blends of agents which produce a pleasant taste sensation.
  • the solution or suspension in for example propylene carbonate, vegetable oils or triglycerides, is encapsulated in a gelatin capsule.
  • a gelatin capsule Such solutions, and the preparation and encapsulation thereof, are disclosed in US 4,328,245, US 4,409,239, and US 4,410,545.
  • the solution e.g., in a polyethylene glycol
  • a pharmaceutically acceptable liquid carrier e.g., water
  • liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active agent or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells.
  • Other useful formulations include those set forth in US RE28819 and US 4,358,603.
  • such formulations include, but are not limited to, those containing an agent provided herein, a dialkylated mono- or poly-alkylene glycol, including, but not limited to, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
  • BHT butylated
  • formulations include, but are not limited to, aqueous alcoholic solutions including a pharmaceutically acceptable acetal.
  • Alcohols used in these formulations are any pharmaceutically acceptable water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol.
  • Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes, such as acetaldehyde diethyl acetal.
  • Tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.
  • Parenteral administration generally characterized by injection, either subcutaneously, intramuscularly or intravenously, is also contemplated herein.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • Suitable excipients include, by way of example and without limitation, water, saline, dextrose, glycerol or ethanol.
  • compositions to be administered may also contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as, for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as, for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • Excipients can also include hydroxypropyl- ⁇ - cyclodextrin (CD), Solutol HS 15 (a PEG15-hydroxystearate) & Cremophor RH 40 (a PEG-40 Hydrogenated Castor Oil).
  • Implantation of a slow-release or sustained-release system is also contemplated herein.
  • the compounds can be dispersed in a solid inner matrix (e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross- linked partially hydrolyzed polyvinyl acetate) that is surrounded by an outer polymeric membrane (e.g., polyethylene, polypropylene, ethylene/propylene copoly
  • a solid inner matrix e.g., polymethylmethacrylate, polybutylme
  • Lyophilized powders can be reconstituted for administration as solutions, emulsions, and other mixtures or formulated as solids or gels.
  • the sterile, lyophilized powder is prepared by dissolving an agent provided herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent.
  • the solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder.
  • Excipients that may be used include, but are not limited to, dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agents.
  • the solvent may also contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH.
  • a buffer such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH.
  • sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation.
  • the resulting solution will be apportioned into vials for lyophilization.
  • Each vial will contain, by way of example and without limitation, a single dosage (10-1000 mg, such as 100- 500 mg) or multiple dosages of the agent.
  • the lyophilized powder can be stored under appropriate conditions, such as at about 4°C to room temperature. Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration.
  • about 1-50 mg, such as about 5-35 mg, for example, about 9-30 mg of lyophilized powder is added per
  • composition or pharmaceutically acceptable derivatives thereof may be formulated as aerosols for application e.g., by inhalation or intranasally (e.g., as described in US 4,044,126, 4,414,209, and 4,364,923).
  • These formulations can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation will, by way of example and without limitation, have diameters of less than about 50 microns, such as less than about 10 microns.
  • the agents may be also formulated for local or topical application, such as for application to the skin and mucous membranes (e.g., intranasally), in the form of nasal solutions, gels, creams, and lotions.
  • local or topical application such as for application to the skin and mucous membranes (e.g., intranasally), in the form of nasal solutions, gels, creams, and lotions.
  • Transdermal patches including iontophoretic and electrophoretic devices, are well known to those of skill in the art. For example, such patches are disclosed in US 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010,715, 5,985,317, 5,983,134, 5,948,433, and 5,860,957.
  • N 3 -(4-bromobenzyl)cyanonilutamide (22) Cyanonilutamide 10 (891 mg, 2.99 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (240 mg, 5.99 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromobenzyl bromide 19 (1.53 g, 5.99 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h.
  • N 3 -(4-bromo-2-fluorobenzyl)cyanonilutamide (23) Cyanonilutamide 10 (998 mg, 3.36 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (268 mg, 6.72 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromo-2- fluorobenzyl bromide 20 (1.85 g, 6.72 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h.
  • N 3 -(4-bromo-3-fluorobenzyl)cyanonilutamide (24) Cyanonilutamide 10 (771 mg, 2.59 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (208 mg, 5.18 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromo-3- fluorobenzyl bromide 21 (1.42 g, 5.18 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h.
  • antiandrogen equipped HDACi having shown that appending antiandrogen moiety derived from Enza skeleton as a secondary pharmacophore to the surface recognition group of a prototypical hydroxamate-based HDACi resulted in antiandrogen equipped HDACi which potently inhibit HDAC isoforms– HDACs 1 and 6 – suggested to be relevant for PCa growth, and weakly active against the less relevant HDAC 8.
  • FIG. 3a shows (a) a 1 st generation antiandrogen equipped HDACi; Antiandrogen equipped HDACi is selectively cytotoxic to (b) AR+ LNCaP PCa cell and (c) a representative member (1d) has a 10-fold in vitro therapeutic index relative to non- transformed Vero cells.
  • HDAC and AR sex hormone binding globulin
  • HDACs and AR can serve as intracellular targets of antiandrogen equipped HDACi.
  • the tubulin acetylation state is an important biomarker that is primarily associated with intracellular HDAC 6 inhibition, while anti-androgen induced perturbation of the AR intracellular localization is confirmatory of AR binding.
  • SAHA was as a positive control for HDAC inhibition and, Enza, bicalutamide and testosterone as positive controls for AR binding.
  • FIG. 4A-C show (a) Intracellular HDAC inhibition of representative compounds probed via ⁇ -tubulin acetylation; (b) Anti-androgen equipped HDACi induced nucleus translocation of AR; and (c) Correlation between HDAC1 inhibition activity and AR nuclear localization. Acetylation was more pronounced than SAHA for the most potent HDAC6 inhibitor 1d, agreeing with the cell-free HDACi assay. Similarly, 1d and 2f caused AR nuclear localization in addition to the positive controls while SAHA did not (Fig. 4b).
  • AR binding and nuclear localization are not fully predictive of the effects of small molecules on AR pathway. Binding of small molecules to the AR may result in either the undesirable agonist or desirable antagonist activity.
  • the pReceiverAR expresses AR; pARLuc is the reporter plasmid which contains firefly luciferase downstream of AR response elements while pCMX ⁇ Gal was used to express ⁇ - galactosidase as an internal control and to assess transfection efficiency.
  • pARLuc is the reporter plasmid which contains firefly luciferase downstream of AR response elements while pCMX ⁇ Gal was used to express ⁇ - galactosidase as an internal control and to assess transfection efficiency.
  • the aryl- nilutamide derivatives (1a ⁇ f and 3) were more potent antagonists than the alkyl- nilutamide compounds (2a ⁇ f and 4) (Fig. 5), correlating with the general trend seen with relative binding affinity.
  • Figure 5 shows AR antagonist activity of antiandrogen equipped HDACi (% RLU for 10 ⁇ M). All compounds competed against 200 pM T. Note data was not presented for compounds 3 and 4. Interestingly, 1b showed
  • FIG. 6A-B shows (a) structures of antiandrogen equipped HDACi studied in Phase I; and (b) stability and intrinsic clearance (CL int ) of lead compounds.
  • SAHA a clinically validated HDACi as a benchmark agent. Relative to SAHA, we observed that all of our compounds have equal or slightly enhanced stability in human plasma.
  • FIG. 8A shows a toxicity study in healthy nude male BALB/c mice and illustrates no significant weight loss in mice when dosed up to 67 mg/kg ip for 8 days.
  • Figures 8B and 8C show the in vivo efficacy study, in which antiandrogen equipped HDACi are (a) not toxic to healthy nude male BALB/c mice and (b) more efficacious than a standard HDACi (SAHA) against xenograft model of AR(+) LNCaP in nude male BALB/c mice (data shown only for 1b to simply our presentation). Inset: 1b remained in the tumor 48 h post administration while undetectable in the serum. (c) A representative antiandrogen equipped HDACi (1e) showed a preliminary indication of potency enhancement with increased AR expression.
  • SAHA standard HDACi
  • the treatment groups were administered lead compounds and SAHA at 10 mg/kg via ip route while Enza was administered (10 mg/kg) through oral gavage in 10% DMSO in hypromellose.
  • Mice in SAHA/Enza combination treatment cohort were simultaneously administered SAHA at 10 mg/kg ip and Enza (10 mg/kg) through oral gavage in 10% DMSO in hypromellose. Animals were dosed once a day, five times a week. As expected based on literature data, SAHA only resulted in modest tumor growth retardation relative to the untreated control. Compounds 1b, 1d and 3 were much more potent than SAHA (Fig. 8b).
  • compounds 1b, 1d and 3 reduced tumor growth rate by nearly 76%, and their effect on tumor growth is nearly indistinguishable from those of Enza and Enza/SAHA combination.
  • Compound 1e is less potent in this model; however it is more potent than SAHA, reducing tumor growth by 45% while SAHA reduced tumor growth by 34%.
  • the tumor growth inhibition effects of 1b and Enza are comparable to their effects at 10 mg/kg while the efficacy of compound 1d is slightly reduced (Not shown).
  • HDACi potently inhibit HDACs 1 and 6, isoforms suggested to be relevant for PCa growth, stable in human plasma and microsomes, target AR expressing PCa cells and robustly inhibit the growth of AR expressing PCa in xemograft mouse model.
  • Compounds 26-28 screened for their HDAC inhibition (against HDACs 1, 6 and 8), AR binding and antiproliferative activity against LNCaP cells. Surprisingly, the AR binding affinities of compounds 26-28 are approx. 12-30 and 25-60 folds stronger than that of 1b and Enza respectively. Compounds 26-28 maintained HDAC1/6 micromolar HDACinhibition activities.
  • the HDAC inhibitory activities of 26-28 are somewhat influenced by their aryl-fluorination pattern.
  • the influence of the aryl-fluorination is even much more pronounced on the antiproliferative activities of these compounds.
  • the un-fluorinated compound 26 and the ortho- fluorinated analog 27 inhibit LNCaP proliferation with mid-nanomolar IC 50 s (approx. 13- 25-fold more potent than the lead compound 1b) while the meta-fluorinated compound 28 has IC 50 is 1.5 ⁇ M (approx. 2-fold more potent than 1b).
  • compounds 26 and 27 are 11-27 times more selective for the AR(+)-LNCaP relative to the AR-independent DU-145 cell while the compound 1b is 4 times more selective for AR(+)-LNCaP.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The disclosure is directed to compounds and methods of using compounds of Formula II. The compounds and methods are effective as inhibitors of to hydantoin-derived histone deacetylase (HDAC), such as in proliferative disease states. The compounds can be formulated with pharmaceutically acceptable carriers and excipients, and can be administered as the compound or its pharmaceutically acceptable salts, prodrugs, or hydrates.

Description

HYDANTOIN-DERIVED HISTONE DEACETYLASE (HDAC) INHIBITORS AND METHODS OF MAKING AND USING THEREOF CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application, filed January 5, 2017, claims the benefit of U.S. Provisional Patent Application Ser. No. 62/275,164, filed January 5, 2016, entitled“HYDANTOIN- DERIVED HISTONE DEACETYLASE (HDAC) INHIBITORS AND METHODS OF MAKING AND USING THEREOF,” the entire contents and substance of which are hereby incorporated by reference as if fully set forth below.
STATEMENT OF FEDERALLY SPONSORED RESEARCH
[0002] This invention was made with Government support under Grant No. 1R43CA180508-01A1. The US Government has certain rights in the invention.
TECHNICAL FIELD
[0003] The various embodiments of the disclosure relate generally to hydantoin-derived histone deacetylase (HDAC) inhibitors that target proliferative and hyperporoliferative diseases, including prostate tumors. More particularly, the disclosure describes a series of hydantoin-derived HDAC inhibitors equipped with androgen receptor binding affinity to target androgen positive prostate. The disclosure describes the methods for making and using thereof.
BACKGROUND
[0004] Prostate cancer (PCa) is the most common form of cancer among all males in the US. Despite the tremendous advances in PCa screening and treatment, more than a quarter million men die globally from the disease every year due primarily to treatment-resistance and metastasis (primarily to the bone) (American Cancer Society. Cancer Facts & Figures 2015. Atlanta: American Cancer Society; 2015.SEER. (2014) Surveillance, Epidemiology, and End Results Program, NCI.). Common treatments include radical prostatectomy, external beam radiation therapy (RT) and interstitial RT (brachytherapy), cryotherapy, and androgen deprivation therapy (ADT). ADT starves tumor of growth-promoting androgen hormones such as testosterone (T). ADT drugs (called antiandrogens) in clinical use, either as monotherapy or combination therapy (with castration), include bicalutamide (Casodex®), nilutamide (Nilandron®), MDV3100 (Enzalutamide) and flutamide (Eulexin®). In the early stage, PCas respond well to ADT and other available therapies. However, within 2–3 years, tumors inevitably enter an antiandrogen-resistant (i.e. castration-resistant or hormone refractory) state and subsequently exhibit chemotherapy- 1 resistance. This castration-resistant state is incurable, and the median survival following this period is just 18–24 months (Eisenberger, M. A.; Blumenstein, B. A.; Crawford, E. D.; et al. Bilateral orchiectomy with or without flutamide for metastatic prostate cancer. N. Engl. J. Med. 1998, 339, 1036–1042.). Therefore, there is a need for increasingly selective and potent drugs to treat castration-resistant stage of PCa (CRPC) and the early stage as well.
[0005] The androgen receptor (AR) expression state is one of the key hallmarks of PCa sustenance and progression. All PCas (early-stage and CRPC) overexpress AR with CRPC showing higher expression than the androgen-dependent early-stage tumors or benign prostate hyperplasias (Linja, M. J.; Savinainen, K. J.; Saramaki, O. R.; Tammela, T. L.; et al. Amplification and Overexpression of Androgen Receptor Gene in Hormone-Refractory Prostate Cancer. Cancer Res 2001, 61, 3550–3555). In fact most mechanistic drivers of CRPC phenotype target the AR pathway. The validated drivers of the CRPC phenotype are (i) tumor expression of promiscuous mutant AR which are activated, in addition to the natural ligands testosterone (T) and dihydrotestoterone (DHT), by other steroid hormones such as progesterone, estradiol, and cortisol, (ii) tumor expression of constitutively active AR splice variants lacking the ligand binding domain, and (iii) adaptive intratumoral androgen biosynthesis powered by adrenal androgens such as DHEA-SO4, DHEA and 4- androstene-3,17-dione (∆4-AD) (Yuan, X.; Balk, S. P. Mechanisms Mediating Androgen Receptor Reactivation After Castration. Urol Oncol. 2009, 27, 36–41. Veldscholte, J.; Berrevoets, C. A.; Ris-Stalpers, C.; Kuiper, G. G.; et al. The androgen receptor in LNCaP cells contains a mutation in the ligand binding domain which affects steroid binding characteristics and response to antiandrogens. J Steroid Biochem Mol Biol. 1992, 41, 665- 669. Tan, J.; Sharief, Y.; Hamil, K. G.; Gregory, C. W.; Zang, D. Y.; Sar, M.; et al. Dehydroepiandrosterone activates mutant androgen receptors expressed in the androgen- dependent human prostate cancer xenograft CWR22 and LNCaP cells. Mol Endocrinol. 1997, 11, 450–459. Knudsen, K.; Scher, H. I. Starving the addiction: New opportunities for durable suppression of AR signaling in prostate cancer. Clin. Cancer Res. 2009, 15, 4792-4798. Knudsen, K.; Penning, T. M. Partners in crime: deregulation of AR activity and androgen synthesis in prostate cancer. Trends Endocrinol. Metab. 2010, 21, 315-324. Yu, Z.; Chen, S.; Sowalsky, A. G.; Voznesensky, O. S.; Mostaghel, E. A.; Nelson, P. S.; Cai, C.; Balk, S P. Rapid induction of androgen receptor splice variants by androgen deprivation in prostate cancer. Clin Cancer Res. 2014, 20, 1590-1600). Tumors harboring amplified AR gene expression and somatic mutations in AR are targeted with the strong antiandrogen (or super-AR antagonist) enzalutamide (Enza) (Scher, H. I.; Beer, T. M.; Higano, C. S.; Anand, A.; Taplin, M. E.; Efstathiou, E.; Rathkopf, D.; et al. Prostate Cancer Foundation/Department of Defense Prostate Cancer Clinical Trials Consortium. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study. Lancet 2010, 375, 1437-1446. Scher, H. I.; Fizazi, K.; Saad, F.; Taplin, M. E.; Sternberg, C. N.; Miller, K.; de Wit, R.; Mulders, P.; et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 2012, 367, 1187- 1197. Tran, C.; Ouk, S.; Clegg, N. J.; Chen, Y.; Watson, P. A.; Arora, V.; Wongvipat, J.; Smith-Jones, P. M.; et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 2009, 324, 787-790.). Enza is not well tolerated by some patients due to dose limiting CNS toxicities (Foster, W. R.; Car, B. D.; Shi, H.; Levesque, P. C.; Obermeier, M. T.; Gan, J.; Arezzo, J. C.; et al. Drug safety is a barrier to the discovery and development of new androgen receptor antagonists. Prostate 2011, 71, 480-488.); however, ARN-509, an Enza-derived next generation super-AR antagonist, is devoid of CNS toxicity and is in a phase III clinical trial (Clegg, N. J.; Wongvipat, J.; Joseph, J. D.; Tran, C.; Ouk, S.; Dilhas, A.; Chen, Y.; et al. ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Res. 2012, 72, 1494-1503.). The contribution of the AR splice variants to the CRPC phenotype is under active investigation and no drug exists yet to target the AR splice variants axis. A recent comprehensive study revealed that the splice variants remained minor transcripts (<1%) relative to the full- length AR (AR-FL) in resistant xenografts and CRPC clinical samples. (Yu, Z.; et al.) Moreover, a previous study has shown that the effects of AR splice variants are mediated through wild-type AR (AR-FL) and that the effect of AR splice variant could be blunted by LBD targeting antiandrogens such as Enza (Watson, P. A.; Chen, Y. F.; Balbas, M. D.; et al. Constitutively active androgen receptor splice variants expressed in castration resistant prostate cancer require full-length androgen receptor. PNAS 2010,107, 16759- 16765.). PCa with adaptive androgen biosynthesis is targeted with a combination of P450c17 (17α-hydroxylase/17,20-lyase) inhibitors, such as abiraterone acetate (Abi) and prednisone (Attard, G.; Reid, A. H. M.; Yap, T.A.; Raynaud, F.; Dowsett, M.; Settatree, S.; et al. Phase 1 clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. J. Clin. Oncol. 2008, 28, 4563-4571. Danila, D. C., Morris, M. J., de Bono, J. S., Ryan, C. J., Denmeade, S. R., Smith, M. R., Taplin, M. E., Bubley, G. J., Kheoh, T.; et al. Phase II multicenter study of abiraterone acetate plus prednisone therapy in patients with docetaxel-treated castration-resistant prostate cancer. J. Clin. Oncol. 2010, 28, 1496-1501. Reid, A. H.; Attard, G.; Danila, D. C.; Oommen, N. B.; Olmos, D.; Fong, P. C.; Molife, L. R.; Hunt, J.; Messiou, C.; Parker, C.; et al. Significant and sustained antitumor activity in post-docetaxel, castration-resistant prostate cancer with the CYP17 inhibitor abiraterone acetate. J. Clin. Oncol. 2010, 28, 1489-1495. Fizazi, K.; Scher, H. I.; Molina, A.; Logothetis, C. J.; Chi, K. N.; Jones, R. J.; Staffurth, J. N.; North, S.; Vogelzang, N. J.; Saad, F.; Mainwaring, P.; Harland, S.; Goodman, O. B. Jr.; Sternberg, C. N.; Li, J. H.; Kheoh, T.; Haqq, C. M.; de Bono, J. S.; COU-AA-301 Investigators. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COUAA- 301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012, 983-992. Attard, G.; Reid, A. H.; Auchus, R. J.; Hughes, B.A.; Cassidy, A.M.; Thompson, E.; Oommen, N.B.et al. Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. J. Clin. Endocrinol. Metab. 2012, 97, 507-516.). P450c17 inhibitors, such as Galeterone (TOK-001) (Vasaitis, T.; Belosay, A.; Schayowitz, A.; Khandelwal, A.; Chopra, P.; Gediya, L.K.; Guo, Z.; Fang, H. B.; Njar, V. C.; Brodie, A. M. Androgen receptor inactivation contributes to antitumor efficacy of 17α-hydroxylase/17,20-lyase inhibitor 3β-hydroxy-17-(1H-benzimidazole-1- yl)androsta-5,16-diene in prostate cancer. Mol. Cancer Therap. 2008, 7, 2348-2357. Yu, Z.; Cai, C.; Gao, S.; Simon, N. I.; Shen, H. C.; Balk, S. P. Galeterone prevents androgen receptor binding to chromatin and enhances degradation of mutant androgen receptor. Clin. Cancer Res. 2014, 20, 4075-4085.) and VT-464,( Rafferty, S. W.; Eisner, J. R.; Moore, W. R.; Schotzinger, R. J.; Hoekstra, W. J. Highly selective 4-(1,2,3-triazole)-based P450c 17α-hydroxylase/ 17,20-lyase inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, 2444- 2447. Toren PJ, Kim S, Pham S, Mangalji A, Adomat H, Guns ES, Zoubeidi A, Moore W, Gleave ME. Anticancer activity of a novel selective CYP17A1 inhibitor in preclinical models of castrate-resistant prostate cancer. Mol. Cancer Ther. 2015, 14, 59–69.) which selectively inhibit the 17,20-lyase activity of P450c17 are being developed as well. Observations from the use of these agents in the clinic, both alone and in combination, have clearly indicated the need for better agents able to treat CRPC.
BRIEF SUMMARY [0006] The various embodiments of the disclosure relate generally to compounds that inhibit histone deacetylase (HDAC) and methods of treatment using the same compound.
[0007] An embodiment of the disclosure can be a compound of Formula II,
Figure imgf000007_0001
[0008] An embodiment of the disclosure can be a method of treating a proliferative disorder by administering a compound of Formula II.
[0009] The disclosure for the compound of or method using Formula II can include W selected from O or S; X selected from C-H or N; Y selected from cyano or nitro; Z selected from trifluoromethyl or iodo; R1 and R2 independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group; R3 selected from hydrogen, fluorine, or CF3, preferably hydrogen or fluorine; and n as 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement.
[0010] The disclosure for the compound of or method using Formula II can include ZBG that is a Zinc Binding Group selected from
Figure imgf000008_0001
.
In the ZBG, m can be 0 to 7, and when m is greater than 0 represents a C1 to C7 group, optionally can contain one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement. R4 can be hydrogen or methyl. R5 can be hydrogen, fluoro, phenyl, or 4-pyridyl. R6 can be hydrogen or a salt counterion. R7 can be a C1 to C6 alkyl or acyl.
R8 and R9 can each independently be hydrogen or C1 to C6 alkyls.
[0011] The disclosure for the compound of or method using Formula II can include n as at least 1, or n equal to 1. In some embodiments of Formula II, Y can be is cyano, Z can be trifluoromethyl, or Y can be cyano and Z can be trifluoromethyl. In some embodiments of Formula II, X can be C-H or N, Y can be cyano, Z can be trifluoromethyl, and n can be greater than 1.
[0012] The disclosure for the compound of or method using Formula II can include ZBG as
Figure imgf000009_0001
Figure imgf000009_0002
In some embodiments, ZBG can include
Figure imgf000009_0003
.
In some embodiments, ZBG can include
Figure imgf000009_0004
[0013] The disclosure for the compound of or method using Formula II can include X as H or N; Y as cyano; Z as trifluoromethyl; n equal to 1; and ZBG is selected from
Figure imgf000010_0001
BRIEF DESCRIPTION OF THE DRAWINGS
[0014] Figure 1A illustrates three exemplary compounds, in accordance with exemplary embodiments of the disclosure.
[0015] Figure 1B illustrates intracellular activity of two compounds of Figure 1A, in accordance with exemplary embodiments of the disclosure.
[0016] Figure 1C illustrates an exemplary synthetic scheme for the three compounds of Fig.1A, in accordance with an exemplary embodiment of the disclosure.
[0017] Figure 2 illustrates exemplary structures 30-146, in accordance with exemplary embodiments of the disclosure.
[0018] Figures 3A-C illustrate comparative chemical compounds and activities, in accordance with an exemplary embodiment of the disclosure.
[0019] Figures 4A-C illustrate activity of comparative chemical compounds and intracellular target validation of comparative compounds, in accordance with an exemplary embodiment of the disclosure.
[0020] Figure 5 illustrates androgen receptor antagonist activity of comparative chemical compounds, in accordance with an exemplary embodiment of the disclosure.
[0021] Figure 6A illustrates the structures of comparative compounds and Figure 6B illustrates the stability and clearance of the compounds in Figure 6A, in accordance with an exemplary embodiment of the disclosure.
[0022] Figure 7 illustrates an in vivo efficacy study of comparative compounds, in accordance with an exemplary embodiment of the disclosure.
[0023] Figures 8A illustrates a toxicity study in healthy mice of comparative compounds and Figures 8B and 8C illustrate in vivo efficacy studies of comparative compounds, in accordance with an exemplary embodiment of the disclosure. DETAILED DESCRIPTION
[0024] Although preferred embodiments of the disclosure are explained in detail, it is to be understood that other embodiments are contemplated. Accordingly, it is not intended that the disclosure is limited in its scope to the details of construction and arrangement of components set forth in the following description or illustrated in the drawings. The disclosure is capable of other embodiments and of being practiced or carried out in various ways. Also, in describing the preferred embodiments, specific terminology will be resorted to for the sake of clarity.
[0025] It must also be noted that, as used in the specification and the appended claims, the singular forms“a,”“an” and“the” include plural referents unless the context clearly dictates otherwise.
[0026] Also, in describing the preferred embodiments, terminology will be resorted to for the sake of clarity. It is intended that each term contemplates its broadest meaning as understood by those skilled in the art and includes all technical equivalents which operate in a similar manner to accomplish a similar purpose.
[0027] Ranges can be expressed herein as from“about” or“approximately” one particular value and/or to“about” or“approximately” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value.
[0028] By“comprising” or“containing” or“including” is meant that at least the named compound, element, particle, or method step is present in the composition or article or method, but does not exclude the presence of other compounds, materials, particles, method steps, even if the other such compounds, material, particles, method steps have the same function as what is named.
[0029] It is also to be understood that the mention of one or more method steps does not preclude the presence of additional method steps or intervening method steps between those steps expressly identified. Similarly, it is also to be understood that the mention of one or more components in a device or system does not preclude the presence of additional components or intervening components between those components expressly identified.
[0030] Ar represents an aryl group.“Aryl”, as used herein, refers to 5-, 6- and 7- membered aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic, or bihetereocyclic ring system, optionally substituted by halogens, alkyl-, alkenyl-, and alkynyl-groups. Broadly defined,“Ar”, as used herein, includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as“aryl heterocycles” or “heteroaromatics”. The aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, alkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties,—CF3,—CN, or the like. The term“Ar” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are“fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocycles. Examples of heterocyclic ring include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2- dithiazinyl, dihydrofuro [2,3b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H- indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4- thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl and xanthenyl. [0031]“Zinc binding group” or“ZBG”, as used herein, refers to moieties capable of inhibiting zinc metalloenzymes activity including HDAC. Suitable examples include, but are not limited to, hydroxamates, N-formyl hydroxylamine (or retro-hydroxamate), carboxylates, thiols, dithiols, trithiocarbonates, thioesters, benzamide, keto, mercaptoacetamides, 2-ketoamides, epoxides, epoxyketones, trifluoromethyl ketones, hydroxypyridinones, pyrones, hydroxylpyridinethiones, and thiopyrones.
[0032]“Alkyl”, as used herein, refers to the radical of saturated or unsaturated aliphatic groups, including straight-chain alkyl, alkenyl, or alkynyl groups, branched-chain alkyl, alkenyl, or alkynyl groups, cycloalkyl, cycloalkenyl, or cycloalkynyl (alicyclic) groups, alkyl substituted cycloalkyl, cycloalkenyl, or cycloalkynyl groups, and cycloalkyl substituted alkyl, alkenyl, or alkynyl groups. Unless otherwise indicated, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone. For example, C1 to C4 or C6 linear alkyl. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure, and optionally contain or are substituted with heteroatoms, such as with 1, 2, 3 or more independently selected oxygen, nitrogen, or sulfur atoms.
[0033]“Alkylaryl”, as used herein, refers to an alkyl group substituted with an aryl group (e.g., an aromatic or hetero aromatic group).
[0034]“Acyl” or“acyl group” as used herein is intended to mean a—C(O)—R radical, where R is a suitable substituent (for example, an acetyl group, a propionyl group, a butyroyl group, a benzoyl group, or an alkylbenzoyl group).
[0035]“Heterocycle” or“heterocyclic”, as used herein, refers to a cyclic radical attached via a ring carbon or nitrogen of a monocyclic or bicyclic ring containing 3-10 ring atoms, and preferably from 5-6 ring atoms, consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y) wherein Y is absent or is H, O,
Figure imgf000013_0001
phenyl or benzyl, and optionally containing 1-3 double bonds and optionally substituted with one or more substituents. Examples of heterocyclic ring include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH- carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H- 1,5,2-dithiazinyl, dihydrofuro [2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H- indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4- thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl and xanthenyl.
[0036]“Heteroaryl”, as used herein, refers to a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and 1, 2, 3, or 4 heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y) where Y is absent or is H, O, (C1-C8) alkyl, phenyl or benzyl. Non-limiting examples of heteroaryl groups include furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyrazolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl, (or its N-oxide), thienyl, pyrimidinyl (or its N- oxide), indolyl, isoquinolyl (or its N-oxide), quinolyl (or its N-oxide) and the like. The term“heteroaryl” can include radicals of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto. Examples of heteroaryl can be furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyraxolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl (or its N-oxide), thientyl, pyrimidinyl (or its N-oxide), indolyl, isoquinolyl (or its N-oxide), quinolyl (or its N- oxide), and the like.
[0037]“Halogen”, as used herein, refers to fluorine, chlorine, bromine, or iodine.
[0038] The terms“alkenyl” and“alkynyl” refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. [0039] The terms ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
[0040]“Pharmaceutically acceptable salt”, as used herein, refer to derivatives of the compounds defined by Formula I wherein the parent compound is modified by making acid or base salts thereof. Example of pharmaceutically acceptable salts include but are not limited to mineral or organic acid salts of basic residues such as amines; and alkali or organic salts of acidic residues such as carboxylic acids. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. Such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric acids; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, naphthalenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic salts.
[0041] The pharmaceutically acceptable salts of the compounds can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 20th ed., Lippincott Williams & Wilkins, Baltimore, Md., 2000, p. 704; and“Handbook of Pharmaceutical Salts: Properties, Selection, and Use,” P. Heinrich Stahl and Camille G. Wermuth, Eds., Wiley-VCH, Weinheim, 2002.
[0042] As generally used herein “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. [0043]“Prodrug”, as used herein, refers to a pharmacological substance (drug) which is administered in an inactive (or significantly less active) form. Once administered, the prodrug is metabolized in the body (in vivo) into the active compound.
[0044]“Solvate”, as used herein, refers to a compound which is formed by the interaction of molecules of a solute with molecules of a solvent.
[0045]“Reverse ester”, as used herein, refers to interchange in the positions of the oxygen and carbon groups in a series of structurally related compounds.
[0046]“Reverse amide”, as used herein, refers to interchange in the positions of the nitrogen and carbon groups in a series of structurally related compounds.
[0047] AR pathway constitutes a viable target to develop new generation of novel, targeted agents against PCas even after the establishment of the CRPC phenotype. Two possible approaches toward new agents against PCas are: (i) development of anti- androgens with stronger AR binding affinities, (ii) redesign of anti-androgens, including but not limiting to the more potent diarylhydantions, to incorporate independent cancer inhibiting chemotype(s). The later approach explores the tumor AR expression state to affect a selective delivery of an independent anti-tumor chemotype. Histone deacetylase (HDAC) inhibiting pharmacophores ideally functions as independent cancer inhibiting chemotype(s) for incorporation to anti-androgen pharmacophores (US 9,139,565). HDAC inhibitors (HDACi) have stimulated much enthusiasm in oncology recently with over 500 cancer clinical trials initiated to date, resulting in five clinically approved drugs: SAHA and FK228, approved by the FDA for the treatment of cutaneous T cell lymphoma; Belinostat and Epidaza, approved for peripheral T cell lymphoma; and Panobinostat, approved for multiple myeloma. Despite their success in blood malignancies, current HDACi have serious limitations in solid tumors. Moreover, HDAC inhibition elicits a pleiotropic phenotype which is largely due to their nonselective inhibition of various cellular HDAC isoforms and possibly other non-HDAC targets as well. This broad HDAC inhibition is associated with reduced in vivo potency and toxic side effects. An approach that selectively target HDAC inhibitors to the diseased cells could improve their therapeutic indices.
[0048] Compounds effective in the HDAC inhibition have been developed. In an embodiment of the disclosure, the compounds of Formula I can be used in HDAC inhibition.
Figure imgf000017_0001
wherein,
Ar represents an aryl group,
W is O or S;
X is C-H or N;
Y is cyano or nitro;
Z is trifluoromethyl or iodo;
R1 and R2 are independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group;
n is 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement; and
ZBG is a Zinc Binding Group selected from
Figure imgf000018_0001
, wherein,
m is 0 to 7, and when m is greater than 0 represents a C1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
R4 is hydrogen or a C1 to C6 alkyl; and
R5 is hydrogen, fluoro, phenyl, or 4-pyridyl
R6 is hydrogen or a salt counterion
R7 is a C1 to C6 alkyl or acyl; and
R8 and R9 are each independently hydrogen or C1 to C6 alkyls.
[0049] As used herein,“Aryl” includes 5-, 6- and 7-membered aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic, or bihetereocyclic ring system, optionally substituted by halogens, alkyl-, alkenyl-, and alkynyl-groups. Broadly defined,“Ar”, as used herein, includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as“aryl heterocycles” or“heteroaromatics”. The aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, --CF3, --CN, or the like. The term “Ar” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are“fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocycles. Examples of heterocyclic ring include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H- 1,5,2-dithiazinyl, dihydrofuro[2,3 b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H- indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4- thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl and xanthenyl.
[0050] In some embodiments, the hydrogen atom positions of the alkene bond bridging the Ar and the ZBG can be isotopically modified to include one or two of deuterium and/or tritium. In some embodiments, both the hydrogen atom positions can be hydrogen. In some embodiments, at least one of the hydrogen atom positions can be isotopically modified to deuterium or tritium, preferably deuterium.
[0051] In another embodiment of the disclosure, the compounds of Formula II can be used in HDAC inhibition.
Figure imgf000020_0001
wherein,
W is O or S;
X is C-H or N;
Y is cyano or nitro;
Z is trifluoromethyl or iodo;
R1 and R2 are independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group;
R3 is hydrogen, fluorine, or CF3
n is 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement; and
ZBG is a Zinc Binding Group selected from
Figure imgf000021_0001
, wherein,
m is 0 to 7, and when m is greater than 0 represents a C1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
R4 is hydrogen or a C1 to C6 alkyl; and
R5 is hydrogen, fluoro, phenyl, or 4-pyridyl
R6 is hydrogen or a salt counterion
R7 is a C1 to C6 alkyl or acyl; and
R8 and R9 are each independently hydrogen or C1 to C6 alkyls.
[0052] As described above, W can be O or S. W can also be oxygen; or W can be sulfur. In some embodiment, X can be C-H or N. X can be C-H to establish the substituted phenyl ring. X can also be N, to establish the substituted pyridyl ring.
[0053] As described above, Y can be cyano or nitro, and Z can be trifluoromethyl or iodo. In various embodiments, Y can be cyano and Z can be trifluoromethyl; Y can be cyano and Z can be iodo; Y can be nitro and Z can be trifluoromethyl; or Y can be nitro and Z can be iodo.
[0054] As described above, R1 and R2 can be independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group. In some embodiments, R1 and R2 can both be a short chain C1 to C4 alkyl. R1 and R2 can both be methyl. In some embodiments, R1 and R2 can be taken together with the carbon they are attached to form a C3 to C6 cycloalkyl ring. R1 and R2 can be taken together with the carbon they are attached can for a cyclopropyl ring, a cyclobutyl ring, a cyclopentyl ring or a cyclohexyl ring. The cycloalkyl ring can be optionally substituted. In some embodiments, the cycloalkyl ring can be cyclopropyl, cyclobutyl, or cyclopentyl, preferably be cyclopropyl or cyclobutyl.
[0055] In some embodiments, R3 can be hydrogen, halo, or CF3; or hydrogen, fluorine or CF3. R3 can be preferably hydrogen or fluorine. R3can be at either of the 2 and 3 position of the aryl ring when R3is not hydrogen.
[0056] As described above, n is 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement. Preferably, n is at least 1, i.e. n is 1 to 6. In some embodiments, n can be 1.
[0057] In some embodiments, the hydrogen atom positions of the alkene bond bridging the Ar and the ZBG can be isotopically modified to include one or two of deuterium or tritium. In some embodiments, both the hydrogen atom positions be hydrogen. In some embodiments, at least one of the hydrogen atom positions can be isotopically modified to deuterium or tritium, preferably deuterium.
[0058] “Zinc binding group” or“ZBG”, as used herein, refers to moieties capable of inhibiting zinc metalloenzymes activity including HDAC and matrix metalloproteinase (MMP) activity. Suitable examples include, but are not limited to, hydroxamates, N- formyl hydroxylamine (or retro-hydroxamate), carboxylates, thiols, dithiols, trithiocarbonates, thioesters, benzamide, keto, mercaptoacetamides, 2-ketoamides, epoxides, epoxyketones, trifluoromethyl ketones, hydroxypyridinones, pyrones, hydroxylpyridinethiones, and thiopyrones. Thus examples of zinc binding groups include, but are not limited to, hydroxamic acids, thiol-hydroxamic acids, n-formyl hydroxylamines, carboxylates, nitro, thiols, dithiol mercaptoacetamies trithiocarbonates,thioesters, benzamides, epoxides, epoxyketones, trifluoromethyl ketones, ketones, and ketoamides. Exemplary ZBGs can also be found in U.S. Patent No. 9,139,565, incorporated herein by reference in its entirety.
[0059] As described above, ZBG can be a zinc binding group selected from
Figure imgf000023_0001
[0060] In the various ZBG noted above and throughout the specification, m can be 0 to 7, and when m is greater than 0, can represent a C1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement. Preferably, m can be 1 to 4, and can represent a C1 to C4 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement. In the various ZBG noted above and throughout the specification, R4 can be hydrogen or a C1 to C6 alkyl, hydrogen or C1 to C4 alkyl, or hydrogen and methyl. In the various ZBG noted above and throughout the specification, R5 can be hydrogen, fluoro, phenyl, or 4-pyridyl. In the various ZBG noted above and throughout the specification, R6 can be hydrogen or a salt counterion, e.g. a sodium salt, a potassium salt, an ammonium or a typical organic salt counterion, such as an alkyl ammonium salt with one to four carbon groups on the ammonium. In the various ZBG noted above and throughout the specification, R7 is a C1 to C6 alkyl or acyl. In the various ZBG noted above and throughout the specification, R8 and R9 are each independently hydrogen or C1 to C6 alkyls.
[0061] In some preferred embodiments, the ZBG can be
Figure imgf000024_0001
wherein R4, R5, R6, R7, R8, R9 and m are as noted above.
[0062] In some preferred embodiments, the ZBG can be
Figure imgf000024_0002
wherein R4, R5 and m are as noted above. [0063] In some embodiments, the ZBG can be
Figure imgf000025_0001
wherein R4, R5 and m are as noted above.
[0064] In another embodiment of the disclosure, the compounds of Formula III can be used.
Figure imgf000025_0002
wherein X, Y, Z, R1, R2, R3, n and ZBG are as defined above.
[0065] In some embodiments, the compound of Formula II can be
Figure imgf000025_0003
wherein X can be C-H or N; Y is cyano; Z is trifluoromethyl; n is greater than 1, and ZBG is selected from
Figure imgf000026_0001
[0066] The compound of Formula II can also include X as C-H or N; Y as cyano; Z as trifluoromethyl; and n equal to 1. Furthermore, ZBG can be selected from
Figure imgf000026_0002
Alternatively, the compound of Formula II can also include ZBG selected from
Figure imgf000027_0001
where m is 1 to 4.
[0067] The compounds of Formulas I, II, and III, as disclosed and fully described above, including all variations therein, can be used to treat and/or prevent proliferative or hyperproliferative disorders. The disclosure includes a method for treating these disorders administering a compound of Formula I, II or III, as disclosed and described above, and incorporated herein. The disorder can include prostate cancer, including hormone sensitive and hormone refractory prostate cancers. Compounds of this disclosure and related compounds in U.S. Patent No. 9,139,565, incorporated by reference herein in its entirety, have been demonstrated to effect inhibition in histone deacetylase (HDAC.) Cinnamoyl derivatives and aryl alkenyl (e.g. styrenyl) structures as disclosed herein have shown a particular and surprising efficacy.
[0068] The compounds of Formulas I, II, and III can be administered as a pharmaceutically acceptable salt, prodrug, or solvate. The compounds described herein can be formulated with a pharmaceutically acceptable carrier and, optionally one or more pharmaceutically acceptable excipients, for enteral or parenteral administration, as further detailed below.
[0069] The compositions of the disclosure can comprise a carrier and/or excipient. While it is possible to use a compound of the present disclosure for therapy as is, it may be preferable to administer it in a pharmaceutical formulation, e.g., in admixture with a suitable pharmaceutical excipient and/or carrier selected with regard to the intended route of administration and standard pharmaceutical practice. The excipient and/or carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Acceptable excipients and carriers for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington: The Science and Practice of Pharmacy. Lippincott Williams & Wilkins (A.R. Gennaro edit.2005). The choice of pharmaceutical excipient and carrier can be selected with regard to the intended route of administration and standard pharmaceutical practice. Oral formulations readily accommodate additional mixtures, such as, e.g., milk, yogurt, and infant formula. Solid dosage forms for oral administration can also be used and can include, e.g., capsules, tablets, caplets, pills, troches, lozenges, powders, and granules. Non-limiting examples of suitable excipients include, e.g., diluents, buffering agents (e.g., sodium bicarbonate, infant formula, sterilized human milk, or other agents which allow bacteria to survive and grow [e.g., survive in the acidic environment of the stomach and to grow in the intestinal environment]), preservatives, stabilizers, binders, compaction agents, lubricants, dispersion enhancers, disintegration agents, antioxidants, flavoring agents, sweeteners, and coloring agents, including hydroxypropyl-β-cyclodextrin (CD), Solutol HS 15 (a PEG15-hydroxystearate) & Cremophor RH 40 (a PEG-40 Hydrogenated Castor Oil). Those of relevant skill in the art are well able to prepare suitable solutions.
[0070] In one embodiment of any of the compositions of the disclosure, the composition is formulated for delivery by a route such as, e.g., oral, topical, rectal, mucosal, sublingual, nasal, naso/oro-gastric gavage, parenteral, intraperitoneal, intradermal, transdermal, intrathecal, nasal, and intracheal administration. In one embodiment of any of the compositions of the disclosure, the composition is in a form of a liquid, foam, cream, spray, powder, or gel. In one embodiment of any of the compositions of the disclosure, the composition comprises a buffering agent (e.g., sodium bicarbonate, infant formula or sterilized human milk). In a preferred embodiment, the composition is formulated for oral delivery.
[0071] Administration of the compounds and compositions in the methods of the disclosure can be accomplished by any method known in the art. Non-limiting examples of useful routes of delivery include oral, rectal, fecal (by enema), and via naso/oro-gastric gavage, as well as parenteral, intraperitoneal, intradermal, transdermal, intrathecal, nasal, and intracheal administration. The active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation. The carrier material should be non-toxic to the bacteria and the subject/patient.
[0072] Although there are no physical limitations to delivery of the formulations of the present disclosure, oral delivery is preferred for delivery to the digestive tract because of its ease and convenience, and because oral formulations readily accommodate additional mixtures and multiple pharmaceutically active ingredients. [0073] For oral administration, the active ingredient(s) can be administered in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions. The active component(s) can be encapsulated in gelatin capsules together with inactive ingredients and powdered carriers, such as glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium saccharin, talcum, magnesium carbonate. Examples of additional inactive ingredients that may be added to provide desirable color, taste, stability, buffering capacity, dispersion or other known desirable features are red iron oxide, silica gel, sodium lauryl sulfate, titanium dioxide, and edible white ink. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric-coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance. In a preferred embodiment, the active ingredient(s) are compounded and administered in a single tablet or capsule.
[0074] Formulations suitable for parenteral administration include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
[0075] Solutions or suspensions can include any of the following components, in any combination: a sterile diluent, including by way of example without limitation, water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent; antimicrobial agents, such as benzyl alcohol and methyl parabens; antioxidants, such as ascorbic acid and sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetates, citrates and phosphates; and agents for the adjustment of tonicity, such as sodium chloride or dextrose.
[0076] In instances in which the agents exhibit insufficient solubility, methods for solubilizing agents may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using co-solvents, such as, e.g., dimethylsulfoxide (DMSO), ethanol, dimethylacetamide or water, using surfactants, such as TWEEN®80, or dissolution in aqueous sodium bicarbonate. Pharmaceutically acceptable derivatives of the agents may also be used in formulating effective pharmaceutical compositions.
[0077] The composition can contain along with the active agent, for example and without limitation: a diluent such as lactose, sucrose, dicalcium phosphate, or carboxymethylcellulose; a lubricant, such as magnesium stearate, calcium stearate and talc; and a binder such as starch, natural gums, such as gum acacia gelatin, glucose, molasses, polyvinylpyrrolidone, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to those of skill in the art. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active agent as defined above and optional pharmaceutical adjuvants in a carrier, such as, by way of example and without limitation, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, such as, by way of example and without limitation, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art (e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975). The composition or formulation to be administered will, in any event, contain a quantity of the active agent in an amount sufficient to alleviate the symptoms of the treated subject.
[0078] The active agents or pharmaceutically acceptable derivatives may be prepared with carriers that protect the agent against rapid elimination from the body, such as time release formulations or coatings. The compositions may include other active agents to obtain desired combinations of properties.
[0079] Oral pharmaceutical dosage forms include, by way of example and without limitation, solid, gel and liquid. Solid dosage forms include tablets, capsules, granules, and bulk powders. Oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated. Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art. [0080] In certain embodiments, the formulations are solid dosage forms, such as capsules or tablets. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or agents of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
[0081] Examples of binders include, by way of example and without limitation, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose, and starch paste. Lubricants include, by way of example and without limitation, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid. Diluents include, by way of example and without limitation, lactose, sucrose, starch, kaolin, salt, mannitol, and dicalcium phosphate. Glidants include, by way of example and without limitation, colloidal silicon dioxide. Disintegrating agents include, by way of example and without limitation, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose. Coloring agents include, by way of example and without limitation, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate. Sweetening agents include, by way of example and without limitation, sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors. Flavoring agents include, by way of example and without limitation, natural flavors extracted from plants such as fruits and synthetic blends of agents which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate. Wetting agents include, by way of example and without limitation, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene laural ether. Emetic- coatings include, by way of example and without limitation, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates. Film coatings include, by way of example and without limitation, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
[0082] If oral administration is desired, the agent could be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active agent in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient. [0083] When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents. The agents can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may contain, in addition to the active agents, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. The active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics.
[0084] Pharmaceutically acceptable carriers included in tablets are binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, and wetting agents. Enteric-coated tablets, because of the enteric-coating, resist the action of stomach acid and dissolve or disintegrate in the neutral or alkaline intestines. Sugar-coated tablets are compressed tablets to which different layers of pharmaceutically acceptable substances are applied. Film-coated tablets are compressed tablets which have been coated with a polymer or other suitable coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned. Coloring agents may also be used in the above dosage forms. Flavoring and sweetening agents are used in compressed tablets, sugar-coated, multiple compressed and chewable tablets. Flavoring and sweetening agents are useful in the formation of chewable tablets and lozenges.
[0085] Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Aqueous solutions include, for example, elixirs and syrups. Emulsions are either oil-in-water or water-in-oil.
[0086] Elixirs are clear, sweetened, hydroalcoholic preparations. Pharmaceutically acceptable carriers used in elixirs include solvents. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative. An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid. Pharmaceutically acceptable carriers used in emulsions are non- aqueous liquids, emulsifying agents and preservatives. Suspensions use pharmaceutically acceptable suspending agents and preservatives. Pharmaceutically acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents. Pharmaceutically acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic acids and a source of carbon dioxide. Coloring and flavoring agents may be used in any of the above dosage forms.
[0087] Solvents include, by way of example and without limitation, glycerin, sorbitol, ethyl alcohol and syrup. Examples of preservatives include, without limitation, glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol. Non-aqueous liquids utilized in emulsions include, by way of example and without limitation, mineral oil and cottonseed oil. Emulsifying agents include, by way of example and without limitation, gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate. Suspending agents include, by way of example and without limitation, sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia. Diluents include, by way of example and without limitation, lactose and sucrose. Sweetening agents include, by way of example and without limitation, sucrose, syrups, glycerin and artificial sweetening agents such as saccharin. Wetting agents include, by way of example and without limitation, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Organic acids include, by way of example and without limitation, citric and tartaric acid. Sources of carbon dioxide include, by way of example and without limitation, sodium bicarbonate and sodium carbonate. Coloring agents include, by way of example and without limitation, any of the approved certified water soluble FD and C dyes, and mixtures thereof. Flavoring agents include, by way of example and without limitation, natural flavors extracted from plants, such as fruits, and synthetic blends of agents which produce a pleasant taste sensation.
[0088] For a solid dosage form, the solution or suspension, in for example propylene carbonate, vegetable oils or triglycerides, is encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in US 4,328,245, US 4,409,239, and US 4,410,545. For a liquid dosage form, the solution (e.g., in a polyethylene glycol) may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier (e.g., water) to be easily measured for administration.
[0089] Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active agent or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in US RE28819 and US 4,358,603. Briefly, such formulations include, but are not limited to, those containing an agent provided herein, a dialkylated mono- or poly-alkylene glycol, including, but not limited to, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
[0090] Other formulations include, but are not limited to, aqueous alcoholic solutions including a pharmaceutically acceptable acetal. Alcohols used in these formulations are any pharmaceutically acceptable water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol. Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes, such as acetaldehyde diethyl acetal.
[0091] Tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient. Thus, for example and without limitation, they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.
[0092] Parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously, is also contemplated herein. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients include, by way of example and without limitation, water, saline, dextrose, glycerol or ethanol. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as, for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins. Excipients can also include hydroxypropyl-β- cyclodextrin (CD), Solutol HS 15 (a PEG15-hydroxystearate) & Cremophor RH 40 (a PEG-40 Hydrogenated Castor Oil). [0093] Implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained (e.g., US 3,710,795) is also contemplated herein. The compounds can be dispersed in a solid inner matrix (e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross- linked partially hydrolyzed polyvinyl acetate) that is surrounded by an outer polymeric membrane (e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer) that is insoluble in body fluids. The agent diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active agent contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the agent and the needs of the subject.
[0094] Lyophilized powders can be reconstituted for administration as solutions, emulsions, and other mixtures or formulated as solids or gels. The sterile, lyophilized powder is prepared by dissolving an agent provided herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent. The solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder. Excipients that may be used include, but are not limited to, dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agents. The solvent may also contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. Generally, the resulting solution will be apportioned into vials for lyophilization. Each vial will contain, by way of example and without limitation, a single dosage (10-1000 mg, such as 100- 500 mg) or multiple dosages of the agent. The lyophilized powder can be stored under appropriate conditions, such as at about 4°C to room temperature. Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration. For reconstitution, about 1-50 mg, such as about 5-35 mg, for example, about 9-30 mg of lyophilized powder, is added per mL of sterile water or other suitable carriers. The precise amount depends upon the selected agent. Such amount can be empirically determined.
[0095] The disclosed composition or pharmaceutically acceptable derivatives thereof may be formulated as aerosols for application e.g., by inhalation or intranasally (e.g., as described in US 4,044,126, 4,414,209, and 4,364,923). These formulations can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation will, by way of example and without limitation, have diameters of less than about 50 microns, such as less than about 10 microns.
[0096] The agents may be also formulated for local or topical application, such as for application to the skin and mucous membranes (e.g., intranasally), in the form of nasal solutions, gels, creams, and lotions.
[0097] Other routes of administration, such as transdermal patches are also contemplated herein. Transdermal patches, including iontophoretic and electrophoretic devices, are well known to those of skill in the art. For example, such patches are disclosed in US 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010,715, 5,985,317, 5,983,134, 5,948,433, and 5,860,957.
EXAMPLES
[0098] Compound numberings, e.g., 1, 2, 3, etc. as used in this example and method of preparation sections of the present application are for reference within these sections only do not describe and are not to be confused with any similar numberings in other section of the present application.
[0099] Synthesis of Compounds 26-28. Exemplary compounds 26, 27, and 28 are shown in Figure 1A and are synthesized in accordance with the scheme in Figure 1C.
[0100] N3-(4-bromobenzyl)cyanonilutamide (22): Cyanonilutamide 10 (891 mg, 2.99 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (240 mg, 5.99 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromobenzyl bromide 19 (1.53 g, 5.99 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h. After that the reaction mixture was cooled to room temperature and was quenched by adding crushed-ice, and then it was extracted with ethyl acetate (50 mL). The organic layer was washed with water (2 x 10 ml), brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude was purified by column chromatography (SiO2, 35% ethyl acetate in hexane) to give the title compound 22 as white solid.1H NMR (400 MHz, CDCl3) δ 8.18 (d, J = 1.6 Hz, 1H), 8.03 (dd, J = 8.4, 2.1 Hz, 1H), 7.93 (d, J = 8.4 Hz, 1H), 7.54– 7.45 (m, 2H), 7.26 (m, 2H), 4.57 (s, 2H), 1.43 (s, 6H).
[0101] Cyanonilutamide-(N3-(4-benzyl-N-hydroxyacrylamide)) (26): Compound 22 (256 mg, 0.55 mmol), N-(trityloxy)acrylamide 25 (151 mg, 0.46 mmol), benzyltriethylammonium chloride (105 mg, 0.46 mmol), tributylamine (0.29 mmol), and palladium acetate (5.1 mg, 0.02 mmol) were mixed in 2 mL of degassed DMF, and the mixture was heated at 70°C for 3 h. After 3 h the reaction was cooled to room temperature and was diluted with ethyl acetate (50 mL), washed with water (20 mL), brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude was used in the next reaction withour further purification.
[0102] The crude was dissolved in 1.8 mL of DCM and to that 0.2 mL of TFA was added followed by dropwise addition of TIPS until the bright yellow solution was turned colorless. The resulting mixture was stirred at room temperature for 1 h and concentrated. The crude was purified by preparative chromatography (ethyl acetate as mobile phase and 15% MeOH in CH2Cl2 as eluent) to give target compound 26 as white solid.1H NMR (400 MHz, CD3OD) δ 8.13 (s, 1H), 8.00 (s, 2H), 7.41 (d, J = 29.1 Hz, 5H), 6.37 (s, 1H), 4.58 (s, 2H), 1.32 (s, 6H).
[0103] N3-(4-bromo-2-fluorobenzyl)cyanonilutamide (23): Cyanonilutamide 10 (998 mg, 3.36 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (268 mg, 6.72 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromo-2- fluorobenzyl bromide 20 (1.85 g, 6.72 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h. After that the reaction mixture was cooled to room temperature and was quenched by adding crushed-ice, and then it was extracted with ethyl acetate (50 mL). The organic layer was washed with water (2 x 10 ml), brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude was purified by column chromatography (SiO2, 40% ethyl acetate in hexane) to give the title compound 23 as white solid.1H NMR (400 MHz, CDCl3) δ 8.14 (d, J = 1.5 Hz, 1H), 8.00 (dd, J = 8.5, 2.0 Hz, 1H), 7.91 (d, J = 8.5 Hz, 1H), 7.39 (t, J = 8.2 Hz, 1H), 7.32– 7.27 (m, 2H), 4.61 (s, 2H), 1.44 (s, 6H)..
[0104] Cyanonilutamide-(N3-(4-(2-fluoro)benzyl-N-hydroxyacrylamide)) (27): Heck coupling of compound 23 (270 mg, 0.56) with N-(trityloxy)acrylamide 25 (153 mg, 0.46 mmol) followed by TBS removal as described for the synthesis of 26, gave target compound 27 as white solid. 1H NMR (400 MHz, CD3OD) δ 8.23 (s, 1H), 8.10 (s, 2H), 7.53 (s, 2H), 7.37 (s, 2H), 6.51 (s, 1H), 4.74 (s, 2H), 1.46 (s, 6H).
[0105] N3-(4-bromo-3-fluorobenzyl)cyanonilutamide (24): Cyanonilutamide 10 (771 mg, 2.59 mmol) was dissolved in anhydrous DMF (5 mL) and to that solution, sodium hydride (208 mg, 5.18 mmol, 60% in oil) was added portion-wise. The resulting suspension was stirred at room temperature for 2 h. Then a solution of 4-bromo-3- fluorobenzyl bromide 21 (1.42 g, 5.18 mmol) in anhydrous DMF (5 mL) was added to the suspension, and the resulting mixture was stirred at 55 °C for 5 h. After that the reaction mixture was cooled to room temperature and was quenched by adding crushed-ice, and then it was extracted with ethyl acetate (50 mL). The organic layer was washed with water (2 x 10 ml), brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude was purified by column chromatography (SiO2, 35% ethyl acetate in hexane) to give the title compound 24 as white solid. 1H NMR (400 MHz, CDCl3) δ 8.15 (d, J = 1.6 Hz, 1H), 8.01 (dd, J = 8.4, 2.0 Hz, 1H), 7.92 (d, J = 8.5 Hz, 1H), 7.59– 7.49 (m, 1H), 7.15 (dd, J = 9.0, 2.0 Hz, 1H), 7.03 (dt, J = 13.8, 6.9 Hz, 1H), 4.55 (s, 2H), 1.43 (s, 6H).
[0106] Cyanonilutamide-(N3-(4-(3-fluoro)benzyl-N-hydroxyacrylamide)) (28): Heck coupling of compound 24 (282 mg, 0.58) with N-(trityloxy)acrylamide 25 (160 mg, 0.49 mmol) followed by TBS removal as described for the synthesis of 26, gave target compound 28 as white solid.1H NMR (400 MHz, CD3OD) δ 8.13 (s, 1H), 7.99 (m, 2H), 7.52 (m, 2H), 7.18 (m, 2H), 6.46 (s, 1H), 4.57 (s, 2H), 1.34 (s, 6H).
[0107] The Heck coupling demonstrated in compounds 26-28 can thus install the alkenyl bond of the cinnamyl structural motif, and thereby allows creations of any ZBG to the cinnamyl structure via a similar connecting Heck coupling. Other alternative syntheses are possible as well, such as oelfin methatheses, Wittig, and other alkene bond constructions. Exemplary non-limiting analogous structures, where n can be 0 to 6 methylene groups, preferably at least 1, and m can be 1 to 4 methylene groups, are disclosed in Figure 2.
[0108] Validation and Testing of comparative compounds lacking the cinnamoyl structural motif
[0109] The antiandrogen equipped HDACi being developed have shown that appending antiandrogen moiety derived from Enza skeleton as a secondary pharmacophore to the surface recognition group of a prototypical hydroxamate-based HDACi resulted in antiandrogen equipped HDACi which potently inhibit HDAC isoforms– HDACs 1 and 6 – suggested to be relevant for PCa growth, and weakly active against the less relevant HDAC 8. Subsequent SAR study resulted in a series of compounds (aryl-nilutamide 1a-f, 3 and alkyl-nilutamide 2a-f and 4) with varying degree of HDAC inhibition potency which tracked well with the length of the methylene linker of the HDACi pharmacophore (Fig. 3a). Figures 3A-C show (a) a 1st generation antiandrogen equipped HDACi; Antiandrogen equipped HDACi is selectively cytotoxic to (b) AR+ LNCaP PCa cell and (c) a representative member (1d) has a 10-fold in vitro therapeutic index relative to non- transformed Vero cells. These compounds also maintained strong binding affinity for AR while devoid of binding interaction with the sex hormone binding globulin (SHBG). Because SHBG is not present in animals (mice) used in preclinical study, a lack of strong interaction with SHBG is an early indicator of the suitability of these compounds in these species for predictive absorption, distribution, metabolism, and excretion (ADME) analysis and in vivo efficacy studies. Moreover, targets (HDAC and AR) binding properties of these compounds translate into potent anticancer activity against hormone- dependent (AR+) LNCaP and to a lesser extent against hormone-independent (AR−) DU145 prostate cancer (Fig. 3b), while having greatly reduced toxicity in noncancerous cells (Fig. 3c). This data illustrates that engaging HDACi and AR with a single chemical probe can achieve both potent and cell-type selective responses.
[0110] HDACs and AR can serve as intracellular targets of antiandrogen equipped HDACi. The tubulin acetylation state is an important biomarker that is primarily associated with intracellular HDAC 6 inhibition, while anti-androgen induced perturbation of the AR intracellular localization is confirmatory of AR binding. As part of the study aimed at cellular target and mechanistic validation, the effect of these compounds on tubulin acetylation (data shown for compounds 1d, 2b and 2f in LNCaP) and AR localization (data shown for compounds 1a-f in HEK) was investigated. SAHA was as a positive control for HDAC inhibition and, Enza, bicalutamide and testosterone as positive controls for AR binding. We observed that 1d, 2f and SAHA induced a dose dependent tubulin hyper-acetylation while 2b moderately induced hyper-acetylation at 10 µM (Fig. 4a). Figure 4A-C show (a) Intracellular HDAC inhibition of representative compounds probed via α-tubulin acetylation; (b) Anti-androgen equipped HDACi induced nucleus translocation of AR; and (c) Correlation between HDAC1 inhibition activity and AR nuclear localization. Acetylation was more pronounced than SAHA for the most potent HDAC6 inhibitor 1d, agreeing with the cell-free HDACi assay. Similarly, 1d and 2f caused AR nuclear localization in addition to the positive controls while SAHA did not (Fig. 4b). Among the compounds with AR binding affinity stronger than that of bicalutamide or Enza (aryl-nilutamide 1a-f) there is a strong linear correlation (R2 = 0.9732) between HDAC1 inhibition activity and the extent to which these compounds induce nuclear localization of YFP-AR (Fig. 4c).31The observed correlation between AR binding affinity and HDAC1 inhibition could simply be due to the enhanced drug exposure to the cell nucleus localized HDAC1, the concomitant effect of AR-drug complex translocation into the nucleus. AR-dependent nuclear localization may contribute to improved inhibition of HDAC in the nucleus of AR containing cells, which we postulate to result in the observed cell-type-selective antiproliferative effects. This result provides strong evidence for the contribution of HDAC inhibition and AR binding to the anti-proliferative activity of these PCa targeted antiandrogen equipped HDACi.
[0111] AR binding and nuclear localization are not fully predictive of the effects of small molecules on AR pathway. Binding of small molecules to the AR may result in either the undesirable agonist or desirable antagonist activity. We therefore evaluated the effect of all synthesized compounds (1a-f, 2a-f, 3 and 4) on AR transcriptional activity to decipher the consequence of their AR interaction on AR activity. We used HEK 293T cells transfected with a mixture of three different plasmids pReceiverAR:pARLuc:pCMXβGal). The pReceiverAR expresses AR; pARLuc is the reporter plasmid which contains firefly luciferase downstream of AR response elements while pCMXβGal was used to express β- galactosidase as an internal control and to assess transfection efficiency. Overall, the aryl- nilutamide derivatives (1a−f and 3) were more potent antagonists than the alkyl- nilutamide compounds (2a−f and 4) (Fig. 5), correlating with the general trend seen with relative binding affinity. Figure 5 shows AR antagonist activity of antiandrogen equipped HDACi (% RLU for 10 µM). All compounds competed against 200 pM T. Note data was not presented for compounds 3 and 4. Interestingly, 1b showed a surprising ability to reduce activity lower than basal levels, a characteristic of relatively uncommon AR an inverse agonist.
[0112] On the strengths of their HDAC inhibition potency, AR binding and inhibition of AR transcriptional activity at lower concentrations than clinical antiandrogens, representative aryl-nilutamide compounds 1b, 1d, 1e and 3 , and one alkyl-nilutamide 4 were evaluated for their in vivo efficacy as part of phase I study.
[0113] We conducted preliminary in vitro PK studies, using human plasma and microsomes, on four compounds– structurally related aryl-nilutamide 1d and 3, and alkyl- nilutamide 4 to check for evidence of disparity in stability among these two classes. Figures 6A-B shows (a) structures of antiandrogen equipped HDACi studied in Phase I; and (b) stability and intrinsic clearance (CLint) of lead compounds. We used SAHA, a clinically validated HDACi as a benchmark agent. Relative to SAHA, we observed that all of our compounds have equal or slightly enhanced stability in human plasma. The stability of aryl-nilutamide 1d and 3 in human microsome is comparable to that of SAHA with nearly indistinguishable intrinsic clearance (CLint) (Fig. 6b). The alkyl-nilutamide 4, though as stable as 1d and 3 in human plasma, is metabolized 3-times faster in human microsome. Based on this preliminary in vitro PK results, we focused exclusively on the aryl-nilutamide compounds (1b, 1d, 1e and 3).
[0114] Efficacy and dose were studied in which the compounds were compared head-to- head with SAHA, a standard HDACi. and Enza, the newest antiandrogen approved by the FDA. For this dose-setting study, DMSO solution of the test agents was used. We first investigated the maximum tolerated dose (MTD) for representative lead compounds 1d and 3, and established that they are not toxic to healthy nude male BALB/c mice at concentration up 100 mg/kg and 50 mg/kg respectively. Based on these preliminary findings, we conducted preliminary efficacy study on 1d and 3 at 25 mg/kg in male nude mice (10 per group) bearing xenograft LNCaP model of PCa. We compared our compounds head-to-head with a treatment cohorts exposed to a combination therapy of SAHA (25 mg/kg), a standard HDACi and Enza (10 mg/kg), the newest antiandrogen approved by the FDA. Compounds 1d, 3 and SAHA were injected ip, q. d. in 100 % DMSO at the volume of 1 uL/gram of mouse body weight. Mice in SAHA treatment cohort were simultaneously administered Enza through oral gavage in 10% DMSO in hypromellose. We found that both 1d and 3 completely suppressed tumor growth at 25 mg/kg and are indistinguishable from the combination of SAHA and Enza (Fig. 7). Figure 7 shows in vivo efficacy evaluation of 1d and 3 in androgen sensitive LNCaP model of PCa
[0115] Compounds (1b, 1d, 1e and 3), SAHA, Enza and combination of SAHA and Enza were challenged in to mice bearing xenograft model of LNCaP PCa (10 per treatment group). Figure 8A shows a toxicity study in healthy nude male BALB/c mice and illustrates no significant weight loss in mice when dosed up to 67 mg/kg ip for 8 days. Figures 8B and 8C show the in vivo efficacy study, in which antiandrogen equipped HDACi are (a) not toxic to healthy nude male BALB/c mice and (b) more efficacious than a standard HDACi (SAHA) against xenograft model of AR(+) LNCaP in nude male BALB/c mice (data shown only for 1b to simply our presentation). Inset: 1b remained in the tumor 48 h post administration while undetectable in the serum. (c) A representative antiandrogen equipped HDACi (1e) showed a preliminary indication of potency enhancement with increased AR expression. In the first set of experiments, the treatment groups were administered lead compounds and SAHA at 10 mg/kg via ip route while Enza was administered (10 mg/kg) through oral gavage in 10% DMSO in hypromellose. Mice in SAHA/Enza combination treatment cohort were simultaneously administered SAHA at 10 mg/kg ip and Enza (10 mg/kg) through oral gavage in 10% DMSO in hypromellose. Animals were dosed once a day, five times a week. As expected based on literature data, SAHA only resulted in modest tumor growth retardation relative to the untreated control. Compounds 1b, 1d and 3 were much more potent than SAHA (Fig. 8b). Specifically, compounds 1b, 1d and 3 reduced tumor growth rate by nearly 76%, and their effect on tumor growth is nearly indistinguishable from those of Enza and Enza/SAHA combination. Compound 1e is less potent in this model; however it is more potent than SAHA, reducing tumor growth by 45% while SAHA reduced tumor growth by 34%. When administered at lower dose (5 mg/kg), the tumor growth inhibition effects of 1b and Enza are comparable to their effects at 10 mg/kg while the efficacy of compound 1d is slightly reduced (Not shown).
[0116] To obtain preliminary data about the effect of AR overexpression on efficacy, lead compounds 1b, 1d, 1e and 3, and Enza (10 mg/kg) were challenged with mice bearing xenograft model of LNCaP-AR, a parental LNCaP overexpressing wild-type AR (PCa model mimicking AR overexpression in CRPC). Compounds 1b, 1d, 1e, 3 and Enza nearly abrogated tumor growth. Intriguingly, compound 1e which is less potent in LNCaP model is more efficacious in this representative PCa model overexpressing AR (Fig.7c).
[0117] Efficacy of compounds with cinnamyl structural motif versus comparative compounds
[0118] The antiandrogen equipped HDACi potently inhibit HDACs 1 and 6, isoforms suggested to be relevant for PCa growth, stable in human plasma and microsomes, target AR expressing PCa cells and robustly inhibit the growth of AR expressing PCa in xemograft mouse model. Compounds 26-28 screened for their HDAC inhibition (against HDACs 1, 6 and 8), AR binding and antiproliferative activity against LNCaP cells. Surprisingly, the AR binding affinities of compounds 26-28 are approx. 12-30 and 25-60 folds stronger than that of 1b and Enza respectively. Compounds 26-28 maintained HDAC1/6 micromolar HDACinhibition activities. However, the HDAC inhibitory activities of 26-28 are somewhat influenced by their aryl-fluorination pattern. The influence of the aryl-fluorination is even much more pronounced on the antiproliferative activities of these compounds. Specifically, the un-fluorinated compound 26 and the ortho- fluorinated analog 27 inhibit LNCaP proliferation with mid-nanomolar IC50s (approx. 13- 25-fold more potent than the lead compound 1b) while the meta-fluorinated compound 28 has IC50 is 1.5 µM (approx. 2-fold more potent than 1b). Additionally, compounds 26 and 27 are 11-27 times more selective for the AR(+)-LNCaP relative to the AR-independent DU-145 cell while the compound 1b is 4 times more selective for AR(+)-LNCaP. These data showed that the morphing of antiandrogen and HDACi into a single molecular template led to synergistic cell growth inhibition of AR(+)-LNCaP as the resulting compounds (26 and 27) are 150-290 and 4-8 times more potent than Enza and SAHA, the benchmark antiandrogen and HDACi respectively (Table 2).
Table 2
Figure imgf000043_0001
[0119] Additionally, on-target effects were exemplified with 26 and 27, which showed intracellular inhibition of HDACs 1 and 6, evidenced by induction of histone 3 and tubulin hyper-acetylation, respectively (Fig 1B).
[0120] It is to be understood that the embodiments and claims disclosed herein are not limited in their application to the details of construction and arrangement of the components set forth in the description and illustrated in the drawings. Rather, the description and the drawings provide examples of the embodiments envisioned. The embodiments and claims disclosed herein are further capable of other embodiments and of being practiced and carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein are for the purposes of description and should not be regarded as limiting the claims.
[0121] Accordingly, those skilled in the art will appreciate that the conception upon which the application and claims are based can be readily utilized as a basis for the design of other structures, methods, and systems for carrying out the several purposes of the embodiments and claims presented in this application. It is important, therefore, that the claims be regarded as including such equivalent constructions.

Claims

CLAIMS We claim:
1. A compound of Formula II,
Figure imgf000045_0001
wherein
W is O or S;
X is C-H or N;
Y is cyano or nitro;
Z is trifluoromethyl or iodo;
R1 and R2 are independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group;
n is 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement;
R3 is hydrogen, fluorine, or CF3; and
ZBG is a Zinc Binding Group selected from
Figure imgf000046_0001
wherein
m is 0 to 7, and when m is greater than 0 represents a C1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
R4 is hydrogen or methyl; and
R5 is hydrogen, fluoro, phenyl, or 4-pyridyl
R6 is hydrogen or a salt counterion
R7 is a C1 to C6 alkyl or acyl; and
R8 and R9 are each independently hydrogen or C1 to C6 alkyls.
2. The compound of Claim 1, wherein n is at least 1.
3. The compound of Claim 1, wherein n is 1.
4. The compound of Claim 1, wherein Y is cyano.
5. The compound of Claim 1, wherein Z is trifluoromethyl.
6. The compound of Claim 1, wherein Y is cyano and Z is trifluoromethyl.
7. The compound of Claim 1, wherein
X is C-H or N;
Y is cyano;
Z is trifluoromethyl;
and n is greater than 1.
8. The compound of Claim 1, wherein ZBG is selected from
Figure imgf000047_0001
9. The compound of claim 8, wherein n is at least 1.
10. The compound of Claim 1, wherein ZBG is selected from
Figure imgf000048_0001
11. The compound of claim 10, wherein n is at least 1.
12. The com ound of Claim 1 wherein when ZBG is selected from
Figure imgf000048_0002
then m is 1 to 4.
13. The compound of claim 1, wherein
X is C-H or N;
Y is cyano;
Z is trifluoromethyl;
and n is 1; and
ZBG is selected from
Figure imgf000048_0003
14. A method of treating a proliferative disorder, comprising administering a compound of Formula II:
Figure imgf000049_0001
wherein
W is O or S;
X is C-H or N;
Y is cyano or nitro;
Z is trifluoromethyl or iodo;
R1 and R2 are independently selected from hydrogen, C1 to C8 alkyl, substituted alkyl including haloalkyl, or taken together with the carbon to which they are linked form a C3 to C6 cycloalkyl or substituted cycloalkyl group;
R3 is hydrogen, fluorine, or CF3;
n is 0 to 6, and when greater than 0 represents C1-6 group, optionally containing one or more heteroatoms, wherein the carbon atoms and/or heteroatoms are in a linear arrangement;
R3 is hydrogen, fluorine, or CF3; and
ZBG is a Zinc Binding Group selected from
Figure imgf000050_0001
wherein
m is 0 to 7, and when m is greater than 0 represents a C1-7 group, optionally containing one or more heteroatoms, with the carbon atoms and/or heteroatoms in a linear arrangement
R4 is hydrogen or methyl; and
R5 is hydrogen, fluoro, phenyl, or 4-pyridyl
R6 is hydrogen or a salt counterion
R7 is a C1 to C6 alkyl or acyl; and
R8 and R9 are each independently hydrogen or C1 to C6 alkyls.
15. The compound of Claim 1, wherein n is at least 1.
16. The compound of Claim 1, wherein n is 1.
17. The compound of Claim 1, wherein Y is cyano.
18. The compound of Claim 1, wherein Z is trifluoromethyl.
19. The compound of Claim 1, wherein Y is cyano and Z is trifluoromethyl.
20. The compound of Claim 1, wherein
X is C-H or N;
Y is cyano;
Z is trifluoromethyl;
and n is greater than 1.
21. The compound of Claim 1, wherein ZBG is selected from
Figure imgf000051_0001
Figure imgf000051_0002
22. The compound of claim 21, wherein n is at least 1.
23. The compound of Claim 1, wherein ZBG is selected from
Figure imgf000052_0001
24. The compound of claim 23, wherein n is at least 1.
25. The compound of Claim 1, wherein when ZBG is selected from
Figure imgf000052_0002
then m is 1 to 4.
26. The compound of claim 1, wherein
X is C-H or N;
Y is cyano;
Z is trifluoromethyl;
and n is 1; and
ZBG is selected from
Figure imgf000052_0003
27. The method of any of claims 14-26, wherein the proliferative disorder is prostate cancer.
PCT/US2017/012323 2016-01-05 2017-01-05 Hydantoin-derived histone deacetylase (hdac) inhibitors and methods of making and using thereof WO2017120328A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662275164P 2016-01-05 2016-01-05
US62/275,164 2016-01-05

Publications (1)

Publication Number Publication Date
WO2017120328A1 true WO2017120328A1 (en) 2017-07-13

Family

ID=59273977

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/012323 WO2017120328A1 (en) 2016-01-05 2017-01-05 Hydantoin-derived histone deacetylase (hdac) inhibitors and methods of making and using thereof

Country Status (1)

Country Link
WO (1) WO2017120328A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070004753A1 (en) * 2005-05-13 2007-01-04 The Regents Of The University Of California Diarylhydantoin compounds
US20090291965A1 (en) * 2005-11-03 2009-11-26 Joshua Close Histone Deacetylase Inhibitors With Aryl-Pyrazolyl-Motifs
US20130289085A1 (en) * 2010-09-28 2013-10-31 Adegboyega Oyelere Histone deacetylase (hdac) inhibitors targeting prostate tumors and methods of making and using thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070004753A1 (en) * 2005-05-13 2007-01-04 The Regents Of The University Of California Diarylhydantoin compounds
US20090291965A1 (en) * 2005-11-03 2009-11-26 Joshua Close Histone Deacetylase Inhibitors With Aryl-Pyrazolyl-Motifs
US20130289085A1 (en) * 2010-09-28 2013-10-31 Adegboyega Oyelere Histone deacetylase (hdac) inhibitors targeting prostate tumors and methods of making and using thereof

Similar Documents

Publication Publication Date Title
ES2568260T3 (en) Pyrimidine hydroxy amide compounds as protein deacetylase inhibitors and methods of use thereof
JP6701237B2 (en) Androgen receptor modulators for the treatment of prostate cancer and androgen receptor-related conditions
JP5769693B2 (en) (R) -N-methylnaltrexone, synthesis method thereof and pharmaceutical use thereof
TWI511732B (en) Reverse amide compounds as protein deacetylase inhibitors and methods of use thereof
JP6506248B2 (en) Bicyclic analgesic compound
HUE028490T2 (en) Zn 2+ -chelating motif-tethered short-chain fatty acids as a novel class of histone deacetylase inhibitors
BR112012000569B1 (en) composition comprising a conjugate of benzoatohydrocodone (bz-hc) and compositions for oral and intravenous administration comprising the same
US20120094963A1 (en) Targeting prodrugs and compositions for the treatment of gastrointestinal diseases
ES2771748T3 (en) Antiandrogens and selective modulators of non-steroidal androgen receptors with a pyridyl residue
ES2764490T3 (en) Derived from chromone oxime that penetrates the brain for therapy of levodopa-induced dyskinesia
CN112272552A (en) Solid forms and formulations comprising glucocorticoid receptor antagonists and uses thereof
JP2013528619A (en) Hydroxypyridone derivatives, pharmaceutical compositions thereof, and their therapeutic use for the treatment of proliferative diseases
US7834010B2 (en) Modulators of peripheral 5-HT receptors
EP1844792A1 (en) Therapeutic agent for constipation
US9475836B2 (en) Protopanoxadiol derivative, preparation method thereof and application thereof
US20010027203A1 (en) Chemical complex comprising a pyridine carboxy derivative and an H2 histamine receptor antagonist
WO2017120328A1 (en) Hydantoin-derived histone deacetylase (hdac) inhibitors and methods of making and using thereof
EP2070908A1 (en) Compound having benzamide skeleton and cyclooxygenase (cox-1)-selective inhibitory activity
WO2011163469A1 (en) Hydrated form of anti-inflammatory roflumilast-n-oxide
JP2008528677A (en) Tetracyclic monoamine reuptake inhibitors for the treatment of diseases and disorders of CNS
KR101072698B1 (en) New metronidazole prodrug compound and pharmaceutical composition comprising the same
CN113861194A (en) Pyrrolopyridine-containing bisamide imidazole compound and preparation method and application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17736328

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17736328

Country of ref document: EP

Kind code of ref document: A1