WO2004082638A2 - Histone deacetylase inhibitors - Google Patents

Histone deacetylase inhibitors Download PDF

Info

Publication number
WO2004082638A2
WO2004082638A2 PCT/US2004/008342 US2004008342W WO2004082638A2 WO 2004082638 A2 WO2004082638 A2 WO 2004082638A2 US 2004008342 W US2004008342 W US 2004008342W WO 2004082638 A2 WO2004082638 A2 WO 2004082638A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound according
phenyl
substituted
compound
Prior art date
Application number
PCT/US2004/008342
Other languages
French (fr)
Other versions
WO2004082638A3 (en
Inventor
Jerome C. Bressi
Jason W. Brown
Sheldon X. Cao
Anthony R. Gangloff
Andrew J. Jennings
Jeffrey A. Stafford
Phong H. Vu
Xiao-Yi Xiao
Original Assignee
Syrrx, 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 Syrrx, Inc. filed Critical Syrrx, Inc.
Priority to JP2006507336A priority Critical patent/JP2006520796A/en
Priority to EP04757631A priority patent/EP1608628A2/en
Priority to CA002518318A priority patent/CA2518318A1/en
Publication of WO2004082638A2 publication Critical patent/WO2004082638A2/en
Publication of WO2004082638A3 publication Critical patent/WO2004082638A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/18Benzimidazoles; Hydrogenated benzimidazoles with aryl radicals directly attached in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings

Definitions

  • the invention relates to compounds that may be used to inhibit deacetylases and, in one variation, histone deacetylases (HDACs), as well as compositions of matter and kits comprising these compounds.
  • HDACs histone deacetylases
  • the present invention also relates to methods for inhibiting deacetylases, such as HDAC, as well as treatment methods using compounds according to the present invention.
  • DNA in eukaryotic cells is tightly complexed with proteins (histones) to form chromatin.
  • Histones are small, positively charged proteins that are rich in basic amino acids (positively charged at physiological pH), which contact the phosphate groups (negatively charged at physiological pH) of DNA.
  • Histones There are five main classes of histones HI, H2A, H2B, H3, and H4.
  • the amino acid sequences of H2A, H2B, H3, and H4 show remarkable conservation between species, wherein HI varies somewhat and in some cases is replaced by another histone, e.g., H5.
  • H2A, H2B, H3 and H4 together form a disk- shaped octomeric protein core, around which DNA (about 140 base pairs) is wound to form a nucleosome.
  • Individual nucleosomes are connected by short stretches of linker DNA associated with another histone molecule to form a structure resembling a beaded string, which is itself arranged in a helical stack, known as a solenoid.
  • a small fraction of histones are enzymatically modified by post-translational addition of methyl, acetyl, or phosphate groups, neutralizing the positive charge of the side chain, or converting it to a negative charge.
  • lysine and arginine groups may be methylated
  • lysine groups may be acetylated
  • serine groups may be phosphorylated.
  • Methylation, acetylation, and phosphorylation of amino termini of histones that extend from the nucleosomal core affects chromatin structure and gene expression. Spencer and Davie 1999. Gene 240:1 1-12.
  • Acetylation and deacetylation of histones is associated with transcriptional events leading to cell proliferation and/or differentiation. Regulation of the function of transcriptional factors is also mediated through acetylation. Recent reviews on histone deacetylation include Kouzarides, et al., 1999. Curr. Opin. Genet. Dev.9: 1, 40-48 and Pazin, et al. 1997. 89:3 325-328.
  • acetylation status of histones and the transcription of genes has been known for quite some time.
  • Certain enzymes, specifically acetylases (e.g., histone acetyltransf erases (HAT)) and deacetylases (histone deacetylases or HDACs), which regulate the acetylation state of histones have been identified in many organisms and have been implicated in the regulation of numerous genes, confirming a link between acetylation and transcription.
  • HAT histone acetyltransf erases
  • deacetylases histone deacetylases or HDACs
  • HDACs histone deacetylases
  • Cancer lust. (Bethesda) 92, 1210-1215, 2000) such as leukemia (Lin et al. 1998. Nature 391: 811-814; Grignani, et al.1998. Nature 391: 815-818; Warrell et al. 1998. J. Natl. Cancer Inst. 90:1621-1625; Gelmetti et al. 1998. Mol. Cell Biol. 18:7185-7191; Wang et al. 1998. PNAS 951 0860- 10865), melanomas/squamous cell carcinomas (Gillenwater, et al., 1998, Int. J.
  • Histone deacetylase inhibitors are potent inducers of growth arrest, differentiation, or apoptotic cell death in a variety of transformed cells in culture and in tumor bearing animals (Histone deacetylase inhibitors as new cancer drugs, Marks, P.A., Richon, V.M., Breslow, R. and Rifkind, R.A., Current Opinions in Oncology, 2001, Nov. 13 (6): 477-83; Histone deacetylases and cancer: causes and therapies, Marks, P., Rifkind, R.A., Richon, V.M., Breslow, R., Miller, T. and Kelly, W.K., Nat. Rev. Cancer 2001 Dec.l (3): 194-202).
  • HDAC inhibitors are useful in the treatment or prevention of protozoal diseases (US Patent 5,922,837) and psoriasis (PCT Publication No. WO 02/26696).
  • PCT Publication No. WO 02/26696 PCT Publication No. WO 02/26696.
  • HDAC inhibitors can be found in Marks PA, et al., J. Natl. Cancer Inst.2000, 92: 1210-1216 & Weidle UH, et al., Antieancer Res.2000, 20: 1471-1486 and PCT Publication Nos. WO 02/26696, WO 02/062773, and WO 01/18171.
  • WO 02/26696 WO 02/062773
  • the present invention relates to compounds that have activity for inhibiting HDACs.
  • the present invention also provides compositions, articles of manufacture and kits comprising these compounds.
  • a pharmaceutical composition that comprises a HDAC inhibitor according to the present invention as an active ingredient.
  • Pharmaceutical compositions according to the invention may optionally comprise 0.001 %-100% of one or more HDAC inhibitors of this invention.
  • These pharmaceutical compositions may be administered or coadministered by a wide variety of routes, including for example, orally, parenterally, mtraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally.
  • the compositions may also be administered or coadministered in slow release dosage forms.
  • the invention is also directed to kits and other articles of manufacture for treating disease states associated with HDAC.
  • a kit comprising a composition comprising at least one HDAC inhibitor of the present invention in combination with instructions.
  • the instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also comprise packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • an article of manufacture is provided that comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • the compounds, compositions, kits and articles of manufacture are used to inhibit HDAC.
  • the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state.
  • a compound is administered to a subject wherein HDAC activity within the subject is altered, preferably reduced.
  • a prodrug of a compound is administered to a subj ect that is converted to the compound in vivo where it inhibits HDAC.
  • a method of inhibiting HDAC comprises contacting HDAC with a compound according to the present invention.
  • a method of inhibiting HDAC comprises causing a compound according to the present invention to be present in a subject in order to inhibit HDAC in vivo.
  • a method of inhibiting HDAC comprises administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo.
  • a therapeutic method comprises administering a compound according to the present invention.
  • a method of inhibiting cell proliferation comprises contacting a cell with an effective amount of a compound according to the present invention.
  • a method of inhibiting cell proliferation in a patient comprises administering to the patient a therapeutically effective amount of a compound according to the present invention.
  • a method of treating a condition in a patient which is known to be mediated by HDAC, or which is known to be treated by HDAC inhibitors comprising administering to the patient a therapeutically effective amount of a compound according to the present invention.
  • a method for using a compound according to the present invention in order to manufacture a medicament for use in the treatment of disease state which is known to be mediated by HDAC, or which is known to be treated by HDAC inhibitors.
  • HDAC possesses activity that contributes to the pathology and/or symptomology of d e disease state, the method comprising: causing a compound according to the present invention to be present in a subject in a therapeutically effective amount for the disease state.
  • HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a first compound to a subject that is converted in vivo to a second compound such that the second compound is present in the subject in a therapeutically effective amount for the disease state.
  • HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a compound according to the present invention to a subject such that the compound is present in the subject in a therapeutically effective amount for the disease state.
  • a method for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated with the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy.
  • combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
  • protozoal diseases and cell proliferative diseases and conditions such as leukemia, melanomas, squamous cell carcinomas, breast cancer, prostrate cancer, bladder cancer, lung cancer including non small-cell lung cancer and small-cell lung cancer, ovarian cancer, colon cancer, squamous cell carcinoma, astrocytoma, Kaposi's sarcoma,
  • the compound optionally comprises a substituent that is convertible in vivo to a different substituent, such as hydrogen.
  • a substituent such as hydrogen
  • an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound.
  • esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di- -toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like.
  • an inhibitor comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound.
  • Figure 1 illustrates a ribbon diagram overview of the structure of HDAC8, highlighting the secondary structural elements of the protein.
  • Figure 2A illustrates particular examples of substituent Ri that may be employed in the Z moiety.
  • Figure 2B illustrates particular examples of Z moieties that the compounds of the present invention may comprise.
  • FIG. 2C illustrates examples of moieties, Q, that the leader group may comprise to link the leader group (L) to the remainder of the compound.
  • Figure 2D illustrates particular examples of moieties that the leader groups may comprise.
  • the squiggle line is intended to indicate a bond to an adjacent moiety. It is also noted that the substituents shown may optionally be further substituted beyond what is shown. Further, one or more heteroatoms may optionally be substituted for the carbon atoms shown. In regard to Figure 2D, it is noted that the leader groups moieties may be inco ⁇ orated into the leader group in either possible orientation.
  • Figure 3 illustrates residues 1-482 of HDACl and a 6-histidine tag at the N- terminus (SEQ. I.D. No. 1).
  • Figure 4 illustrates the DNA sequence (SEQ. ID. No.2) that was used to encode
  • Figure 5 illustrates residues 1-488 of HDAC2 and a 6-histidine tag at the C- terminus (SEQ. ID. No. 3).
  • Figure 6 illustrates the DNA sequence (SEQ. ID. No.4) that was used to encode
  • Figure 7 illustrates residues 73-845 of HDAC6 and a 6-histidine tag at the C- terminus (SEQ. ID. No. 5).
  • Figure 8 illustrates the DNA sequence (SEQ. ID. No. 6) that was used to encode
  • Figure 9 illustrates residues 1-377 of HDAC8 and a 6-histidine tag at the N- terminus (SEQ. ID. No. 7).
  • Figure 10 illustrates the DNA sequence (SEQ. ID. No.8) that was used to encode
  • Alicyclic means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one or more double or triple bonds.
  • Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur.
  • alicyclic moieties include, but are not limited to moieties with C3 - C8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
  • Aliphatic means a moiety characterized by a. straight or branched chain arrangement of constituent carbon atoms and may be saturated or partially unsaturated with one or more double or triple bonds.
  • Alkenyl represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one double bond between adjacent carbon atoms.
  • Cx alkenyl and C ⁇ .y alkenyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 2 . 6 alkenyl includes alkenyls that have a chain of between 2 and 6 carbons.
  • Alkoxy means an oxygen moiety having a further alkyl substituent.
  • Alkyl represented by itself means a straight or branched, saturated or unsaturated, aliphatic radical having a chain of carbon atoms, optionally with oxygen (See
  • oxaalkyl or nitrogen atoms (See “aminoalkyl”) between the carbon atoms.
  • C x alkyl and C ⁇ . ⁇ alkyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 1-6 alkyl includes alkyls that have a chain of between 1 and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methylallyl, ethynyl, 1-propynyl, 2-propynyl, and the like).
  • 1 and 6 carbons e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methylallyl, ethynyl, 1-propynyl, 2-
  • Alkyl represented along with another radical means a straight or branched, saturated or unsaturated aliphatic divalent radical having the number of atoms indicated or when no atoms are indicated means a bond (e.g., (C 6 - ⁇ o)aryl(Co- 3 )alkyl includes phenyl, benzyl, phenethyl, 1-phenylethyl 3-phenylpropyl, and the like).
  • Alkylene unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical.
  • Cx alkylene and C ⁇ . ⁇ alkylene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • 2-methyltetramethylene (-CH 2 CH(CH 3 )CH 2 CH 2 -), pentamethylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -) and the like).
  • Cx alkylidene and C ⁇ . ⁇ alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • Alkynyl represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one triple bond between adjacent carbon atoms.
  • Cx alkynyl and C ⁇ . ⁇ alkynyl are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 2-6 alkynyl includes alkynyls that have a chain of between 2 and 6 carbons.
  • amino means a nitrogen moiety having two further substituents where each substituent has a hydrogen or carbon atom alpha bonded to the nitrogen. Unless indicated otherwise, the compounds of the invention containing amino moieties may include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Aminoalkyl means an alkyl, as defined above, except where one or more substituted or unsubstituted nitrogen atoms (-N-) are positioned between carbon atoms of the alkyl. For example, an (C 2-6 ) aminoalkyl refers to a chain comprising between 2 and 6 carbons and one or more nitrogen atoms positioned between the carbon atoms.
  • Animal includes humans, non-human mammals (e.g., dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like).
  • non-human mammals e.g., dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like
  • non-mammals e.g., birds, and the like.
  • Aromatic means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp 1 hybridized and the total number of pi electrons is equal to 4n+2.
  • An aromatic ring may be such that the ring atoms are only carbon atoms or may include carbon and non-carbon atoms (see Heteroaryl).
  • Aryl means a monocyclic or fused bicyclic ring assembly wherein each ring is aromatic or when fused with a second ring forms an aromatic ring assembly. If one or more ring atoms is not carbon (e.g., N, S), the aryl is a heteroaryl. Cx aryl and C ⁇ . ⁇ aryl are typically used where X and Y indicate the number of atoms in the ring.
  • Bicycloalkyl means a saturated or partially unsaturated fused bicyclic or bridged polycyclic ring assembly.
  • Bicycloaryl means a bicyclic ring assembly wherein the rings are linked by a single bond or fused and at least one of the rings comprising the assembly is aromatic.
  • Cx bicycloaryl and C ⁇ . ⁇ bicycloaryl are typically used where X and Y indicate the number of carbon atoms in the bicyclic ring assembly and directly attached to the ring.
  • Carbamoyl means the radical -OC(O)NR a Rb where R a and R b are each independently two further substituents where a hydrogen or carbon atom is alpha to the nitrogen. It is noted that carbamoyl moieties may include protected derivatives thereof.
  • Suitable protecting groups for carbamoyl moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, and the like. It is noted that both the unprotected and protected derivatives fall within the scope of the invention.
  • Carbocycle means a ring consisting of carbon atoms.
  • Carbocyclic ketone derivative means a carbocyclic derivative having a -C(O)- substituent.
  • Carbonyl means the radical -C(O)-. It is noted that the carbonyl radical may be further substituted with a variety of substituents to form different carbonyl groups including acids, acid halides, amides, esters, and ketones.
  • Carboxy means the radical -C(O)O-. It is noted that compounds of the invention containing carboxy moieties may include protected derivatives thereof, i.e., where the oxygen is substituted with a protecting group. Suitable protecting groups for carboxy moieties include benzyl, tert-butyl, and the like.
  • Cyano means the radical -CN.
  • Cycloalkyl means a non-aromatic, saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly.
  • C x cycloalkyl and C ⁇ . ⁇ cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
  • C -10 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, bicyclo[2.2.2]octyl, adamantan-1-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl, 2-oxobicyclo[2.2.1]hept-l-yl, and the like.
  • Cycloalkylene means a divalent saturated or partially unsaturated, monocyclic ring or bridged polycyclic ring assembly.
  • C cycloalkylene and C ⁇ . ⁇ cycloalkylene are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
  • Disease specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, i.e., the "side effects" of such therapy.
  • Halo means fluoro, chloro, bromo or iodo.
  • Halo-substituted alkyl as an isolated group or part of a larger group, means
  • alkyl substituted by one or more "halo" atoms, as such terms are defined in this Application.
  • Halo-substituted alkyl includes haloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like
  • halo-substituted (C 1-3 )alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
  • Heterobicycloalkyl means bicycloalkyl, as defined in this Application, provided that one or more of the atoms forming the ring is a heteroatom.
  • hetero(C 9-12 )bicycloalkyl as used to define Z in this application includes, but is not limited to,
  • Heterocycloalkylene means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms indicated, is replaced by a heteroatom.
  • Heteroaryl means an aryl ring, as defined in this Application, where one or more of the atoms forming the ring is a heteroatom.
  • Heterobicycloaryl means bicycloaryl, as defined in this Application, provided that one or more of die atoms forming die ring is a heteroatom.
  • hetero(Cg -1 o)bicycloaryl as used in this Application includes, but is not limited to, 2-amino-
  • Heterocycloalkyl means cycloalkyl, as defined in this Application, provided that one or more of the atoms forming the ring is a heteroatom.
  • Haldroxy means the radical -OH.
  • Imine derivative means a derivative comprising the moiety -C(NR)-, wherein R comprises a hydrogen or carbon atom alpha to the nitrogen.
  • Isomers mean any compound having an identical molecular formulae but differing in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed
  • stereoisomers Stereoisomers that are not mirror images of one another are termed
  • enantiomers or sometimes “optical isomers".
  • a carbon atom bonded to four nonidentical substituents is termed a "chiral center”.
  • a compound with one chiral center has two enantiomeric forms of opposite chirality.
  • a mixture of the two enantiomeric forms is termed a "racemic mixture”.
  • a compound that has more than one chiral center has 2 n ⁇ enantiomeric pairs, where n is the number of chiral centers.
  • Compounds with more than one chiral center may exist as ether an individual diastereomers or as a mixture of diastereomers, termed a "diastereomeric mixture”.
  • a stereoisomer When one chiral center is present a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog. Conventions for stereochemical nomenclature, methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (e.g., see “Advanced Organic Chemistry", 4th edition, March, Jerry, John Wiley & Sons, New York, 1992). [0091] "Nitro" means the radical -NO 2 .
  • Oxaalkyl means an alkyl, as defined above, except where one or more oxygen atoms (-O-) are positioned between carbon atoms of the alkyl.
  • an (C -6 )oxaalkyl refers to a chain comprising between 2 and 6 carbons and one or more oxygen atoms positioned between the carbon atoms.
  • 'Oxoalkyl means an alkyl, further substituted with a carbonyl group.
  • the carbonyl group may be an aldehyde, ketone, ester, amide, acid or acid chloride.
  • “Pharmaceutically acceptable” means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically acceptable salts” means salts of inhibitors of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity.
  • Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, efhanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid
  • Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases.
  • Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide.
  • Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like.
  • Prodrug means a compound that is convertible in vivo metabolically into an inhibitor according to the present invention.
  • the prodrug itself may or may not also have HDAC inhibitory activity.
  • an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound.
  • Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates and quinates.
  • Protected derivatives means derivatives of inhibitors in which a reactive site or sites are blocked with protecting groups. Protected derivatives are useful in the preparation of inhibitors or in themselves may be active as inhibitors. A comprehensive list of suitable protecting groups can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
  • Substituted or unsubstituted means that a given moiety may consist of only hydrogen substituents through available valencies (unsubstituted) or may further comprise one or more non-hydrogen substituents through available valencies (substituted) that are not otherwise specified by the name of the given moiety.
  • isopropyl is an example of an ethylene moiety that is substituted by -CH 3 .
  • a non-hydrogen substituent may be any substituent that may be bound to an atom of the given moiety that is specified to be substituted.
  • substituents include, but are not limited to, aldehyde, alicyclic, aliphatic, alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocycle, carboxy, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted.
  • Sulfinyl means the radical -S(O)-. It is noted that the sulfinyl radical may be further substituted with a variety of substituents to form different sulfinyl groups including sulfinic acids, sulfinamides, sulfinyl esters, and sulfoxides.
  • Sulfonyl means the radical -S(O)(O)-. It is noted that the sulfonyl radical may be further substituted with a variety of substituents to form different sulfonyl groups including sulfonic acids, sulfonamides, sulfonate esters, and sulfones.
  • “Therapeutically effective amount” means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
  • Thiocarbonyl means the radical -C(S)-. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including ti ioacids, thioamides, thioesters, and thioketones.
  • Treatment or “treating” means any administration of a compound of the present invention and includes:
  • Ci alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom.
  • a Ci alkyl comprises methyl (i.e., -CH 3 ) as well as -CR a RbR c where R a , R , and R c may each independently be hydrogen or any other substituent where the atom alpha to the carbon is a heteroatom or cyano.
  • CF 3 , CH 2 OH and CH 2 CN are all Ci alkyls.
  • the present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit histone deacetylases (referred to herein as HDACs).
  • HDACs histone deacetylases
  • the compounds may optionally be more particularly used as inhibitors of Class I HDACs such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • HDACs have been categorized into tiiree distinct classes based on their relative size and sequence homology. The different HDACs (Homo sapiens), HDAC classes, sequences and references describing the different HDACs are provided in Tables 1 - 3.
  • Class I HDACs are Class I HDACs. All Class I HDACs appear to be sensitive to inhibition by trichostatin A (TS A). Also of particular note is HDAC8, a protein whose crystal structure Applicants determined and used in conjunction with arriving at the present invention.
  • HDAC8 is a 377 residue, 42kDa protein localized to the nucleus of a wide array of tissues, as well as several human tumor cell lines.
  • the wild-type form of full length HDAC8 is described in GenBank Accession Number NP 060956; Buggy ,JJ., Sideris,M.L., Mak,P., Lorimer,D.D., Mclhtosh,B. and Clark, J.M., Cloning and characterization of a novel human histone deacetylase, HDAC8, Biochem. J. 350 Pt 1, 199-205 (2000).
  • Zn 2+ is likely native to the protein and required for HDAC8 activity.
  • FIG. 1 illustrates a ribbon diagram overview of the structure of HDAC8, highlighting the secondary structural elements of the protein.
  • HDAC8 was found to have a single domain structure belonging to the open ⁇ / ⁇ class of folds.
  • the structure consists of a central 8-stranded parallel ⁇ -sheet sandwiched between layers of ⁇ -helices.
  • the ligand binding clefts lie almost in the plane of the central ⁇ -sheet, and are formed primarily by loops emanating from the carboxy-terminal ends of the ⁇ -strands comprising the sheet.
  • Residues contained in the extension off the second ⁇ -strand form a globular "cap" over the core of the protein, play an important role in defining the shape of the ligand binding pockets, and are involved in a number of key interactions with the bound ligands.
  • HDAC inhibitors of the present invention are provided that comprise the formula
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen, and each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R , R 3 , R 4 and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • each X is independently selected from the group consisting of CR 1 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R 6 , R , R 8( and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aiyloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbon
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR ⁇ 2 ;
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R 6 , R , R 8> and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
  • HDAC inhibitors of die present invention comprise the formula:
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R 2 , R 3 , R 4 , R 5 , R 6 , R , R 8 , and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino,
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R 10 and R ⁇ are taken together to form a substituted or unsubstituted aromatic ring; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
  • R , R 3 , , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z-Q- is selected from the group consisting of
  • Ri is selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • R 2 , R 3 , R , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z-Q- is selected from the group consisting of
  • Ri is selected from the group consisting of (C 1-4 )alkyl, phenyl, l-piperidin-4-ylmethyl, 2-morpholi-4-yl-ethyl, 2-halo-phenyl, 2-halo-phen(C 1 - 4 )alkyl, 2-CF 3 O- phen(C 1-4 )alkyl, 3-CF 3 O-phen(C 1 )alkyl, 3-halo-phenyl, 4-halo-phenyl, 2-mefhoxy-phenyl, 3- methoxy-phenyl, 4-methoxy-phenyl, 4-phenoxy-phenyl, 4-benzyloxyphenyl, 4-pyrazol-l-yl- benzyl, 1-p-tolyl-eti yl, pyrrolidin-3-yl, l-(C 1-4 )alkyl-pyrrolidin-2-yl, l-(Ci )alkyl-pyrro ⁇ idin- 2-yl; 2-di
  • R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro;
  • M is selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise the formula:
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of die present invention comprise the formula:
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of the present invention comprise d e formula:
  • each V is independently selected from the group consisting of C(R 12 ) 2 and NR 12 where at least one V is NR 12 ; each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ; each R ⁇ 2 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention comprise the fonnula:
  • Z is selected from the group consisting of
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z is selected from the group consisting of
  • R 2 , R 3 , R 4 and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • R 6 , R , R 8> and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • R 6 , R , R 8 , and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R ⁇ is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbony
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • R 2 , R 3 , R , R 5 , R 6 , R7, R 8 , and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino,
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
  • HDAC inhibitors of die present invention comprise the formula:
  • Z is selected from the group consisting of
  • R 10 and R ⁇ are taken together to form a substituted or unsubstituted aromatic ring; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R ⁇ 2 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z-Q- is selected from the group consisting of
  • each V is independently selected from the group consisting of C(R 12 ) 2 and NR ⁇ 2 where at least one V is NR 12 ;
  • R , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 1 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aiyloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 2-10 atoms separation between M and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z-Q- is selected from the group consisting of
  • each V is independently selected from the group consisting of C(R 12 ) 2 and NR 12 where at least one V is NR ⁇ 2 ;
  • R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R ⁇ 2 is not halo, cyano, nitro, and thio in die case where the ring atom to which R 12 is bound is nitrogen;
  • M is selected from the group consisting of
  • HDAC inhibitors of the present invention comprise the formula:
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR ⁇ 2 ; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R ⁇ 2 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of d e present invention comprise the formula:
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ; each R 1 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso tiiat R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of the present invention comprise the formula:
  • Z is selected from the group consisting of wherein each X is independently selected from the group consisting of CR ⁇ and N; each Y is independently selected from the group consisting of O, S and NR 12 ; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 12 is bound is nitrogen;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR ⁇ 2 ;
  • R 2 , R 3 , R 4 and R 5 are each independently selected from die group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbony
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • R 6 , R 7 , R 8j and R are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl
  • Ri 4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ; R 6 , R , R 8 , and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryl
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and die ring.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ;
  • R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8j and R 9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino,
  • R1 4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z is selected from the group consisting of
  • each X is independently selected from the group consisting of CR 12 and N; each Y is independently selected from the group consisting of O, S and NR 12 ; R 10 and R ⁇ are taken together to form a substituted or unsubstituted aromatic ring; each R 12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R 12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R 1 is bound is nitrogen;
  • Ri 4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • Q is a substituted or unsubstituted aromatic ring
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
  • HDAC inhibitors of die present invention comprise the formula:
  • R 2 , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
  • HDAC inhibitors of the present invention are provided that comprise the formula:
  • Z-Q- is selected from the group consisting of
  • R , R 3 , R 4 , and R 5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion;
  • HDAC inhibitors of the present invention comprise the formula:
  • Z-Q- is selected from the group consisting of
  • R 2 , R 3 , R 4 , and R 5 are each independently selected from die group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro;
  • R 14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
  • M is selected from the group consisting of
  • Q is a substituted or unsubstituted phenyl ring. In another variation of any of the above embodiments comprising Q, Q is a substituted or unsubstituted heteroaryl.
  • Q is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzofhiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, benthiazole, and triazine.
  • At least one X in the six membered ring is a substituted carbon atom.
  • at least one X in the six membered ring is -CF.
  • any two adjacent X moieties may optionally be CRj 2 where the R 12 substituents are taken together to form a ring.
  • M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF 3 ), -NH- P(O)OH-CH 3 , sulfonamides (-SO 2 NH ), hydroxysulfonamides (-SO 2 NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R 13 wherein R 13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
  • M comprises a hydroxamic acid.
  • M is selected from the group consisting of:
  • QLM is
  • QLM is
  • R 2 , R 3 , R 4 ., and R 5 at least one of R 2 , R 3 , R 4 , or R 5 is fluorine.
  • R 6 , R 7 , R 8 or R 9 may be selected such that the phenyl ring linking the five membered ring and the L group comprise one or two fluorines as indicated in the structural subunit below:
  • substituted or unsubstituted aromatic ring formed when Rio and R ⁇ are taken together may optionally be a substituted or unsubstituted aryl or a heteroaryl.
  • each V is selected so that the ring is an unsubstituted or substituted piperdin-3-yl moiety.
  • R 14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
  • R 14 is a substituted or unsubstituted C 1-6 alkyl. In still another variation of any of the above embodiments and variations comprising R 14 , R 14 is a substituted or unsubstituted - C(O)C 1-6 alkyl.
  • R 14 is selected from the group consisting of hydrogen, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted.
  • R 14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
  • R 14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
  • a given alkyl, alkoxy, aryloxy, heteroaryloxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, amino, thio, or carbonyl group substituent may optionally be further substituted.
  • such two substituents may be taken together to form a ring.
  • Examples of further substituted alkyl groups include, but are not limited to, those selected from the group consisting of haloalkyl, cycloalkyl, aminoalkyl, oxaalkyl, heteroaralkyl, and aralkyl, each of which may optionally be further substituted.
  • Examples of further substituted alkoxy aryloxy, and heteroaryloxy groups include, but are not limited to, those selected from the group consisting of haloalkoxy, haloaryloxy, and haloheteroaryloxy, each of which may optionally be further substituted.
  • Examples of further substituted aminosulfonyl, alkylsulfonyl, arylsulfonyl, and heteroarylsulfonyl groups include, but are not limited to, those selected from the group consisting of alkylaminosulfonyl, arylaminosulfonyl, heteroarylaminosulfonyl, heteroaralkylsulfonyl, and aralkylsulfonyl, each of which may optionally be further substituted.
  • Examples of further substituted amino groups include, but are not limited to, those selected from the group consisting of alkylamino, arylarriino, and acylamino, each of which may optionally be further substituted.
  • Examples of further substituted thio groups include, but are not limited to, tiiose selected from die group consisting of alkylthio, arylthio, and heteroarylthio, each of which may optionally be further substituted.
  • Examples of further substituted carbonyl groups include, but are not limited to, acids, acid halides, amides, esters, and ketones.
  • the carbonyl groups may be an alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl, aminocarbonyl, alkoxycarbonyl, aralkoxycarbonyl, or heteroaralkoxycarbonyl, each of which may optionally be further substituted.
  • Figure 2A illustrates particular examples of moieties that may be used as a Ri substituent.
  • the below table also provides examples of different compounds having different Ri substituents.
  • Ri is a substituted alkyl where the carbon of Rj alpha to the ring atom is a tertiary carbon, i.e., in addition to the bond to the ring atom, the carbon atom has two non-hydrogen substituents.
  • R 2 , R 3 , R , R 5 , R 6 , R , R 8 and R 9 may each independently be selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted.
  • R 2 and R 3 ; R 3 and R 4 ; and R 4 and R 5 may each optionally be taken together to form a ring.
  • the ring formed may optionally be a 5 or 6 membered ring.
  • the ring formed is an aryl or heteroaryl ring.
  • R 6 and R 7 ; R and R 8 ; and R 8 and R 9 may each optionally be taken together to form a ring.
  • the ring formed may optionally be a 5 or 6 membered ring.
  • the ring formed is an aryl or heteroaryl ring.
  • Figure 2B illustrates particular examples of Z moieties that the compounds of the present invention may comprise.
  • the Z moiety is a substituted or unsubstituted benzimidazole or imidazole.
  • Figure 2B is intended only to be exemplary and that other Z substituents may be employed in the compounds according to the present invention consistent with the teachings herein.
  • Q may be a substituted or unsubstituted aromatic ring.
  • the substituents of the aromatic ring can vary widely and may optionally be such that one or more additional rings are fused to d e core aromatic ring of Q.
  • Q may optionally be a 5 or 6 membered aromatic ring.
  • moieties Z and L may be meta or para substituents relative to each other on the
  • moieties Z and L are meta substituted relative to each other.
  • die phenyl ring may have substituents R 6 ,
  • R , R 8 , and R 9 substituents may each optionally be independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted. It is noted that other substituents may additionally be appended to the phenyl ring without departing from the intended scope of the present invention.
  • Q is a 5 and 6 membered aromatic ring comprising heteroatoms, i.e., a heteroaryl.
  • the heteroaryl ring may optionally have the formula or
  • a, b, c, d and e are each independently nitrogen (N) or carbon (C), with a proviso that when a and c are both nitrogen, then c is carbon.
  • N nitrogen
  • C carbon
  • substituents include, but are not limited to members selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted. It is noted that other substituents may additionally be appended to the heteroaryl ring without departing from the intended scope of the present invention. [0176] Examples of rings comprising heteroatoms, including 5 and 6 membered aromatic rings comprising heteroatoms are illustrated in Figure 2C. It is noted that the rings shown in Figure 2C are unsubstituted and that further substitutions may optionally be added as has been specified.
  • the resulting inhibitors show improved biological activities over that of the corresponding para substituted aryl or heteroaryl groups.
  • the meta substituted aryl is a meta substituted phenyl moiety that is substituted or unsubstituted.
  • the meta substitution serves to direct the zinc complexing substituent M to a more favorable position so as to allow the zinc complexing substituent to interact with the zinc ion while the remainder of the compound maintains its interaction with hydrophobic regions in the binding pocket of the histone deacetylase.
  • substituent M may be a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion, and optionally more particularly a zinc ion since a zinc ion is known to be present in the catalytic site of deacetylases.
  • the M substituent may facilitate inhibitor binding by complexing with the zinc ion present in the catalytic site of deacetylases.
  • M is a substituent capable of complexing with a histone deacetylase catalytic site and/or a metal ion.
  • substituents capable of complexing with a zinc ion tiiat may be used as the M substituent include, but are not limited to trifluoroacetyl (-C(O)-CF 3 ), -NH-P(O)OH- CH 3 , sulfonamides (-SO 2 NH 2 ), hydroxysulfonamides (-SO 2 NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R 13 wherein R 13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkyloxy group.
  • substituents include:
  • M is a hydroxamic acid (-C(O)-NHOH), also shown above. It is noted that hydroxamic acids, such as trichostatin A, have been shown to be effective inhibitors against histone deacetylases by complexing with the zinc ion present in the catalytic site of histone deacetylases.
  • the leader group, L may be any substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
  • the number of atoms separating the M substituent and the remainder of the compound serves to extend the zinc complexing substituent, M, a sufficient distance away from the remainder of the compound so as to allow the zinc complexing substituent to interact with the zinc ion while the remainder of the compound interacts with hydrophobic regions in the binding pocket of the deacetylase.
  • the leader group, L provides between 1-10 atoms that extend from the M substituent to remainder of the compound, optionally 3-9 and optionally 4-8 atoms.
  • the number of atoms separating the M substituent from the remainder of the compound is 3, 4, 5, 6, 7, 8 or 9 atoms.
  • the atoms of the leader group extending between the M substituent and the remainder of the compound may consist only of carbon atoms.
  • the atoms of the leader group extending between the M substituent and the remainder of the compound may also comprise non-carbon atoms such as nitrogen, oxygen and sulfur.
  • the bonds between the atoms of the leader group extending between the M substituent and the remainder of the compound may be saturated, partially unsaturated, or fully unsaturated.
  • the atoms forming the backbone of the leader group, L may optionally comprise one or more members of the group consisting of: -(CH 2 )n-, where n is an integer from 1 to 10; -CH(CH 3 )-; -CH(CH 3 )CH 2 - and -CH 2 CH(CH 3 )-; -CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, and -CH 2 CH 2 CH (CH 3 )-; -CH(CH 3 )CH 2 CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 CH 2 -, - CH 2 CH 2 CH(CH 3 )CH 2 -, and -CH 2 CH 2 CH 2 CH(CH 3 )-; -CH(CH 3 )CH 2 CH 2 CHCH 2 -, - CH 2 CH(CH 3 )CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH(CH 3 )CH 2 -;
  • the atoms of the leader group forming the ring may be separated from each other by 0, 1, 2, 3, or 4 atoms.
  • the rings may be saturated or partially unsaturated (i.e., comprise one or two double bonds).
  • the rings may also be aromatic, referred to herein as aryl and heteroaryl rings.
  • the rings may optionally be further substituted. These further ring substituents may combine to form additional rings that are fused to the rings forming a portion of the backbone, e.g., bicycloaryl and bicycloheteroaryl.
  • cycloalkyl rings that may be formed by one or more leader group backbone atoms include, but are not limited to: cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, phenyl, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
  • heteroaryl rings that may be formed by one or more leader group backbone atoms include, but are not limited to: furan, thiofuran, pyrrole, isopyrrole, 3- isopyrrole, pyrazole, isoimidazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiofuran, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, and pyridopyridine.
  • the inhibitors may include one or more chiral centers.
  • the chiral centers may be either the R or S enantiomers, depending on the substituents.
  • d e compounds of the present invention may be present and optionally administered in the form of salts, hydrates and prodrugs that are converted in vivo into the compounds of the present invention.
  • the compounds of the present invention possess a free base form
  • the compounds can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, e.g., hydrohalides such as hydrochloride, hydrobromide, hydroiodide; otiier mineral acids and their corresponding salts such as sulfate, nitrate, phosphate, etc.; and alkyl- and monoarylsulfonates such as ethanesulfonate, toluenesulfonate and benzenesulfonate; and other organic acids and their corresponding salts such as acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate and ascorbate.
  • a pharmaceutically acceptable inorganic or organic acid e.g., hydrohalides such as hydrochloride, hydrobromide, hydroiodide
  • otiier mineral acids and their corresponding salts such
  • Further acid addition salts of the present invention include, but are not limited to: adipate, alginate, arginate, aspartate, benzenesulfonate (besylate), bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, ethanesulfonate, fumarate, galacterate (from mucic acid), galacturonate, glucoheptaoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, iodide, isethionat
  • a pharmaceutically acceptable base addition salt can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • bases include alkali metal hydroxides including potassium, sodium and lithium hydroxides; alkaline earth metal hydroxides such as barium and calcium hydroxides; alkali metal alkoxides, e.g. potassium ethanolate and sodium propanolate; and various organic bases such as ammonium hydroxide, piperidine, diethanolamine and N-methylglutamine.
  • aluminum salts of the compounds of the present invention are alkali metal hydroxides including potassium, sodium and lithium hydroxides; alkaline earth metal hydroxides such as barium and calcium hydroxides; alkali metal alkoxides, e.g. potassium ethanolate and sodium propanolate; and various organic bases such as ammonium hydroxide, piperidine, diethanolamine and N-methylglutamine.
  • aluminum salts of the compounds of the present invention are also included.
  • Organic base salts of the present invention include, but are not limited to: copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium and zinc salts.
  • Organic base salts include, but are not limited to, salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, e.g., arginine, betaine, caffeine, chloroprocaine, choline, N, N'-dibenzylethylenediamine (benzad ine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine,
  • N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art.
  • N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichlorornethane) at approximately 0 °C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, met ⁇ -chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichlorornethane
  • the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
  • Prodrug derivatives of compounds according to the present invention can be prepared by modifying substituents of compounds of the present invention tiiat are then converted in vivo to a different substituent. It is noted that in many instances, the prodrugs themselves also fall within the scope of the range of compounds according to the present invention.
  • prodrugs can be prepared by reacting a compound with a carbamylating agent (e.g., l,l-acyloxyalkylcarbonochloridate, ? ⁇ r ⁇ -nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et ⁇ /.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985.
  • Protected derivatives of compounds of the present invention can also be made. Examples of techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3 rd edition, John Wiley & Sons, Inc. 1999.
  • Compounds of the present invention may also be conveniently prepared, or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxane, tetrahydrofuran or methanol.
  • a "pharmaceutically acceptable salt”, as used herein, is intended to encompass any compound according to the present invention that is utilized in the form of a salt thereof, especially where the salt confers on the compound improved pharmacokinetic properties as compared to the free form of compound or a different salt form of the compound.
  • the pharmaceutically acceptable salt form may also initially confer desirable pharmacokinetic properties on the compound that it did not previously possess, and may even positively affect the pharmacodynamics of the compound with respect to its therapeutic activity in the body.
  • An example of a pharmacokinetic property that may be favorably affected is the manner in which the compound is transported across cell membranes, which in turn may directly and positively affect the absorption, distribution, biotransformation and excretion of the compound.
  • d e pharmaceutical composition While the route of administration of d e pharmaceutical composition is important, and various anatomical, physiological and pathological factors can critically affect bioa ailability, the solubility of the compound is usually dependent upon the character of d e particular salt form thereof, which it utilized.
  • aqueous solution of the compound will provide the most rapid absorption of the compound into the body of a subject being treated, while lipid solutions and suspensions, as well as solid dosage forms, will result in less rapid adsorption of the compound.
  • the diastereomers may then be separated in order to recover the optically pure enantiomers.
  • Dissociable complexes may also be used to resolve enantiomers (e.g., crystalline diastereoisomeric salts).
  • Diastereomers typically have sufficiently distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) that they can be readily separated by taking advantage of these dissimilarities.
  • diastereomers can typically be separated by chromatography or by separation/resolution techniques based upon differences in solubility.
  • a more detailed description of techniques that can be used to resolve stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
  • HDAC is believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of HDAC in a subject through inhibition may be used to therapeutically address these disease states. Examples of various diseases that may be treated using the HDAC inhibitors of the present invention are described herein. It is noted that additional diseases beyond those disclosed herein may be later identified as the biological roles that HDAC play in various pathways becomes more fully understood.
  • HDAC inhibitors of the present invention may be used to treat are those involving undesirable or uncontrolled cell proliferation.
  • indications include benign tumors, various types of cancers such as primary tumors and tumor metastasis, restenosis (e.g. coronary, carotid, and cerebral lesions), abnormal stimulation of endothelial cells (atherosclerosis), insults to body tissue due to surgery, abnormal wound healing, abnormal angiogenesis, diseases that produce fibrosis of tissue, repetitive motion disorders, disorders of tissues that are not highly vascularized, and proliferative responses associated with organ transplants.
  • restenosis e.g. coronary, carotid, and cerebral lesions
  • abnormal stimulation of endothelial cells atherosclerosis
  • insults to body tissue due to surgery abnormal wound healing
  • abnormal angiogenesis diseases that produce fibrosis of tissue, repetitive motion disorders, disorders of tissues that are not highly vascularized, and proliferative responses associated with organ transplants.
  • HDAC inhibitors include, but are not limited to prostate cancer, lung cancer, acute leukemia, multiple myeloma, bladder carcinoma, renal carcinoma, breast carcinoma, colorectal carcinoma, neuroblastoma and melanoma.
  • a method for treating diseases associated with undesired and uncontrolled cell proliferation. The method comprises administering to a subject suffering from uncontrolled cell proliferation a therapeutically effective amount of a HDAC inhibitor according to the present invention, such that said uncontrolled cell proliferation is reduced.
  • the particular dosage of the inhibitor to be used will depend on the severity of the disease state, the route of administration, and related factors that can be dete ⁇ nined by the attending physician.
  • HDAC inhibitors according to the present invention may also be used in conjunction with other agents to inhibit undesirable and uncontrolled cell proliferation.
  • anti-cell proliferation agents include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATL TM protein, ENDOSTATINTM protein, suramin, squalamine, tissue inhibitor of metalloproteinase-I, tissue inhibitor of metalloproteinase-2, plasminogen activator inhibitor- 1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulfate (clupeine), sulfated chitin derivatives (prepared from queen crab shells), sulfated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of
  • anti-angiogenesis agents include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: bFGF, aFGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-l/Ang-2.
  • bFGF vascular endothelial growth factor
  • aFGF vascular endothelial growth factor
  • FGF-5 vascular endothelial growth factor
  • VEGF isoforms VEGF-C
  • HGF/SF Ang-l/Ang-2.
  • Ferrara N. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
  • a benign tumor is usually localized and nonmetastatic.
  • Specific types of benign tumors that can be treated using HDAC inhibitors of the present invention include hemangiomas, hepatocellular adenoma, cavernous haemangioma, focal nodular hyperplasia, acoustic neuromas, neurofibroma, bile duct adenoma, bile duct cystanoma, fibroma, lipomas, leiomyomas, mesotheliomas, teratomas, myxomas, nodular regenerative hyperplasia, trachomas and pyogenic granulomas.
  • Malignant tumors In the case of malignant tumors, cells become undifferentiated, do not respond to the body' s growth control signals, and multiply in an uncontrolled manner. Malignant tumors are invasive and capable of spreading to distant sites (metastasizing). Malignant tumors are generally divided into two categories: primary and secondary. Primary tumors arise directly from the tissue in which they are found. Secondary tumors, or metastases, are tumors that originated elsewhere in the body but have now spread to distant organs. Common routes for metastasis are direct growth into adjacent structures, spread through the vascular or lymphatic systems, and tracking along tissue planes and body spaces (peritoneal fluid, cerebrospinal fluid, etc.).
  • Specific types of cancers or malignant tumors, either primary or secondary, that can be treated using the HDAC inhibitors of the present invention include, but are not limited to, leukemia, breast cancer, skin cancer, bone cancer, prostate cancer, liver cancer, lung cancer, brain cancer, cancer of the larynx, gallbladder, pancreas, rectum, parathyroid, thyroid, adrenal, neural tissue, head and neck, colon, stomach, bronchi, kidneys, basal cell carcinoma, squamous cell carcinoma of both ulcerating and papillary type, metastatic skin carcinoma, osteo sarcoma, Ewing's sarcoma, veticulum cell sarcoma, myeloma, giant cell tumor, small- cell lung tumor, gallstones, islet cell tumor, primary brain tumor, acute and chronic lymphocytic and granulocytic tumors, hairy-cell tumor, adenoma, hyperplasia, medullary carcinoma, pheochromocytoma, mucosal neurode
  • the HDAC inhibitors of the present invention may also be used to treat abnormal cell proliferation due to insults to body tissue during surgery. These insults may arise as a result of a variety of surgical procedures such as joint surgery, bowel surgery, and cheloid scarring. Diseases that produce fibrotic tissue include emphysema. Repetitive motion disorders that may be treated using the present invention include carpal tunnel syndrome. An example of a cell proliferative disorder that may be treated using the invention is a bone tumor.
  • Proliferative responses associated with organ transplantation that may be treated using HDAC inhibitors of the invention include proliferative responses contributing to potential organ rejections or associated complications. Specifically, these proliferative responses may occur during transplantation of the heart, lung, liver, kidney, and otiier body organs or organ systems.
  • Abnormal angiogenesis that may be may be treated using this invention include those abnormal angiogenesis accompanying rheumatoid arthritis, ischemic-reperfusion related brain edema and injury, cortical ischemia, ovarian hyperplasia and hypervascularity, (polycystic ovary syndrome), endometriosis, psoriasis, diabetic retinopathy, and other ocular angiogenic diseases such as retinopathy of prematurity (retrolental fibroplastic), macular degeneration, corneal graft rejection, neuroscular glaucoma and Oster Webber syndrome.
  • abnormal angiogenesis accompanying rheumatoid arthritis, ischemic-reperfusion related brain edema and injury, cortical ischemia, ovarian hyperplasia and hypervascularity, (polycystic ovary syndrome), endometriosis, psoriasis, diabetic retinopathy, and other ocular angiogenic diseases
  • Examples of diseases associated with uncontrolled angiogenesis include, but are not limited to retinal/choroidal neovascularization and corneal neovascularization.
  • Examples of retinal/choroidal neovascularization include, but are not limited to, Bests diseases, myopia, optic pits, Stargarts diseases, Pagets disease, vein occlusion, artery occlusion, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum carotid apo structive diseases, chronic uveitis/vitritis, mycobacterial infections, Lyme's disease, systemic lupus erythematosus, retinopathy of prematurity, Eales disease, diabetic retinopathy, macular degeneration, Bechets diseases, infections causing a retinitis or chroiditis, presumed ocular histoplasmosis, pars planitis, chronic retinal detachment,
  • corneal neovascularization examples include, but are not limited to, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, sjogrens, acne rosacea, phylectenulosis, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, Mooren ulcer, Terrien's marginal degeneration, marginal keratolysis, polyarteritis, Wegener sarcoidosis, Scleritis, periphigoid radial keratotomy, neovascular glaucoma and retrolental fibroplasia, syphilis, Mycobacteria infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections and Kaposi sarcoma
  • Chronic inflammatory diseases associated with uncontrolled angiogenesis may also be treated using HDAC inhibitors of the present invention.
  • Chronic inflammation depends on continuous formation of capillary sprouts to maintain an influx of inflammatory cells. The influx and presence of the inflammatory cells produce granulomas and thus maintains the chronic inflammatory state. Inhibition of angiogenesis using a HDAC inhibitor alone or in conjunction with other anti-inflammatory agents may prevent the formation of the granulosmas and thus alleviate the disease.
  • Examples of chronic inflammatory diseases include, but are not limited to, inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, psoriasis, sarcoidosis, and rheumatoid arthritis.
  • Inflammatory bowel diseases such as Crohn's disease and ulcerative colitis are characterized by chronic inflammation and angiogenesis at various sites in die gastrointestinal tract.
  • Crohn's disease occurs as a chronic transmural inflammatory disease that most commonly affects the distal ileum and colon but may also occur in any part of the gastrointestinal tract from the mouth to the anus and perianal area.
  • Patients with Crohn's disease generally have chronic diarrhea associated with abdominal pain, fever, anorexia, weight loss and abdominal swelling.
  • Ulcerative colitis is also a chronic, nonspecific, inflammatory and ulcerative disease arising in the colonic mucosa and is characterized by the presence of bloody diarrhea.
  • inflammatory bowel diseases are generally caused by chronic granulomatous inflammation throughout the gastrointestinal tract, involving new capillary sprouts surrounded by a cylinder of inflammatory cells. Inhibition of angiogenesis by these inhibitors should inhibit the formation of the sprouts and prevent the formation of granulomas. Inflammatory bowel diseases also exhibit extra intestinal manifectations, such as skin lesions. Such lesions are characterized by inflammation and angiogenesis and can occur at many sites other the gastrointestinal tract. Inhibition of angiogenesis by HDAC inhibitors according to the present invention can reduce the influx of inflammatory cells and prevent lesion formation.
  • Sarcoidosis another chronic inflammatory disease, is characterized as a multisystem granulomatous disorder.
  • the granulomas of this disease can form anywhere in the body. Thus, the symptoms depend on the site of the granulomas and whether the disease is active.
  • the granulomas are created by the angiogenic capillary sprouts providing a constant supply of inflammatory cells.
  • HDAC inhibitors according to the present invention to inhibit angionesis, such granulomas formation can be inhibited.
  • Psoriasis also a chronic and recurrent inflammatory disease, is characterized by papules and plaques of various sizes. Treatment using these inhibitors alone or in conjunction with other anti-inflammatory agents should prevent the formation of new blood vessels necessary to maintain the characteristic lesions and provide the patient relief from the symptoms.
  • Rheumatoid arthritis is also a chronic inflammatory disease characterized by non-specific inflammation of the peripheral joints. It is believed that the blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, the endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. The factors involved in angiogenesis may actively contribute to, and help maintain, the chronically inflamed state of rheumatoid arthritis. Treatment using HDAC inhibitors according to the present invention alone or in conjunction with other anti-RA agents may prevent the formation of new blood vessels necessary to maintain the chronic inflammation and provide the RA patient relief from the symptoms.
  • compositions and administration methods may be used in conjunction with the HDAC inhibitors of the present invention.
  • Such compositions may include, in addition to the HDAC inhibitors of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents.
  • the compositions may include active agents in addition to the HDAC inhibitors of the present invention. These additional active agents may include additional compounds according to the invention, or one or more other pharmaceutically active agents.
  • the compositions may be in gaseous, liquid, semi-liquid or solid form, formulated in a manner suitable for the route of administration to be used. For oral administration, capsules and tablets are typically used. For parenteral administration, reconstitution of a lyophilized powder, prepared as described herein, is typically used.
  • compositions comprising HDAC inhibitors of the present invention may be administered or coadministered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermaily, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally.
  • the compounds and/or compositions according to the invention may also be administered or coadministered in slow release dosage forms.
  • HDAC inhibitors and compositions comprising them may be administered or coadministered in any conventional dosage form.
  • Coadministration in the context of tiiis invention is intended to mean the administration of more than one therapeutic agents, one of which includes a HDAC inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome.
  • Such coadministration may also be coextensive, that is, occurring during overlapping periods of time.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application may optionally include one or more of the following components: a sterile diluent, such as 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; agents for the adjustment of tonicity such as sodium chloride or dextrose, and agents for adjusting the acidity or alkalinity of the composition, such as alkaline or acidifying agents or buffers like carbonates, bicarbonates, phosphates, hydrochloric acid, and organic acids like acetic and citric acid.
  • Parenteral preparations may optionally be enclosed in ampules
  • HDAC inhibitors according to the present invention exhibit insufficient solubility
  • methods for solubilizing the compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEN, or dissolution in aqueous sodium bicarbonate.
  • cosolvents such as dimethylsulfoxide (DMSO)
  • surfactants such as TWEEN
  • dissolution in aqueous sodium bicarbonate such as sodium bicarbonate
  • Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.
  • a solution, suspension, emulsion or the like may be formed.
  • the form of the resulting composition will depend upon a number of factors, including the intended mode of administration, and the solubility of the compound in the selected carrier or vehicle.
  • the effective concentration needed to ameliorate the disease being treated may be empirically determined.
  • compositions according to the present invention are optionally provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, dry powders for inhalers, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds, particularly the pharmaceutically acceptable salts, preferably the sodium salts, thereof.
  • the pharmaceutically tiierapeuticaliy active compounds and derivatives ti ereof are typically formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit- dose forms refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art.
  • Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.
  • unit-dose forms include ampoules and syringes individually packaged tablet or capsule.
  • Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form.
  • Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pint or gallons.
  • multiple dose form is a multiple of unit-doses that are not segregated in packaging.
  • the composition may comprise: 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 acaciagelatin, glucose, molasses, polvinylpyrrolidine, 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 acaciagelatin, glucose, molasses, polvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like
  • the pharmaceutical composition to be administered may also contain minor amounts of auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • composition or formulation to be administered will, in any event, contain a sufficient quantity of a HDAC inhibitor of the present invention to reduce HDAC activity in vivo, thereby treating the disease state of the subject.
  • Dosage forms or compositions may optionally comprise one or more HDAC inhibitors according to the present invention in the range of 0.005% to 100% (weight/weight) with the balance comprising additional substances such as those described herein.
  • a pharmaceutically acceptable composition may optionally comprise any one or more commonly employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum.
  • compositions include solutions, suspensions, tablets, capsules, powders, dry powders for inhalers and sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, and biodegradable, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others. Methods for preparing these formulations are known to those skilled in the art.
  • the compositions may optionally contain 0.01%-100% (weight/weight) of one or more HDAC inhibitors, optionally 0.1-95%, and optionally 1-95%.
  • Salts, preferably sodium salts, of the HDAC inhibitors may be prepared with carriers that protect the compound against rapid elimination from the body, such as time release formulations or coatings.
  • the formulations may further include other active compounds to obtain desired combinations of properties.
  • Oral pharmaceutical dosage forms may be as a solid, gel or liquid.
  • solid dosage forms include, but are not limited to tablets, capsules, granules, and bulk powders. More specific examples of oral tablets include compressed, chewable lozenges and tablets that may be enteric-coated, sugar-coated or film-coated.
  • capsules include hard or soft gelatin capsules. Granules and powders may be provided in non-effervescent or effervescent forms. Each may be combined with other ingredients known to those skilled in the art.
  • HDAC inhibitors according to the present invention are provided as solid dosage forms, preferably capsules or tablets.
  • the tablets, pills, capsules, troches and the like may optionally contain one or more of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
  • binders examples include, but are not limited to, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste.
  • lubricants examples include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • diluents examples include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • glidants examples include, but are not limited to, colloidal silicon dioxide.
  • disintegrating agents examples include, but are not limited to, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • coloring agents examples include, but are not limited to, 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 examples include, but are not limited to, sucrose, lactose, mannitol and artificial sweetening agents such as sodium cyclamate and saccharin, and any number of spray-dried flavors.
  • flavoring agents examples include, but are not limited to, natural flavors extracted from plants such as fruits and synthetic blends of compounds that produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
  • wetting agents examples include, but are not limited to, propylene glycol monost ⁇ arate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylen ⁇ lauryl ether.
  • anti-emetic coatings examples include, but are not limited to, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
  • film coatings examples include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyetiiylene glycol 4000 and cellulose acetate phthalate.
  • the salt of d e compound may optionally 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 compound in the intestine.
  • the composition may also be formulated in combination with an antacid or other such ingredient.
  • dosage unit form When the dosage unit form is a capsule, it may optionally additionally comprise a liquid carrier such as a fatty oil.
  • dosage unit forms may optionally additionally comprise various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • Compounds according to the present invention may also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like.
  • a syrup may optionally comprise, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the HDAC inhibitors of the present invention may also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics.
  • active materials such as antacids, H2 blockers, and diuretics.
  • Examples of pharmaceutically acceptable carriers that may be included in tablets comprising HDAC inhibitors of the present invention include, but are not limited to 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 may be compressed tablets to which different layers of pharmaceutically acceptable substances are applied.
  • Film-coated tablets may be compressed tablets that have been coated with polymers or other suitable coating. Multiple compressed tablets may be compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned.
  • Coloring agents may also be used in tablets. Flavoring and sweetening agents may be used in tablets, and are especially useful in the formation of chewable tablets and lozenges.
  • liquid oral dosage forms tiiat may be used include, but are not limited to, aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • aqueous solutions examples include, but are not limited to, elixirs and syrups.
  • elixirs refer to clear, sweetened, hydroalcoholic preparations.
  • pharmaceutically acceptable carriers examples include, but are not limited to solvents.
  • solvents Particular examples include glycerin, sorbitol, ethyl alcohol and syrup.
  • syrups refer to concentrated aqueous solutions of a sugar, for example, sucrose. Syrups may optionally further comprise a preservative.
  • Emulsions refer to two-phase systems in which one liquid is dispersed in the form of small globules throughout another liquid.
  • Emulsions may optionally be oil-in-water or water-in-oil emulsions.
  • pharmaceutically acceptable carriers that may be used in emulsions include, but are not limited to non-aqueous liquids, emulsifying agents and preservatives.
  • Examples of pharmaceutically acceptable substances that may be used in non- effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents.
  • Examples of pharmaceutically acceptable substances that may be used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic adds and a source of carbon dioxide.
  • Coloring and flavoring agents may optionally be used in all of the above dosage forms.
  • preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia.
  • Diluents include lactose and sucrose.
  • Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as sodium cyclamate and saccharin.
  • wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • organic acids that may be used include citric and tartaric acid.
  • Sources of carbon dioxide that may be used in effervescent compositions include sodium bicarbonate and sodium carbonate.
  • Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof.
  • Particular examples of flavoring agents that may be used include natural flavors extracted from plants such fruits, and synthetic blends of compounds that produce a pleasant taste sensation.
  • the solution or suspension in for example propylene carbonate, vegetable oils or triglycerides, is preferably encapsulated in a gelatin capsule.
  • a gelatin capsule Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the solution, egg. for example, 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.
  • a pharmaceutically acceptable liquid carrier e.g. water
  • liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound 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 U.S. Pat. Nos. Re 28,819 and 4,358,603.
  • compositions designed to administer the HDAC inhibitors of the present invention by parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously.
  • injectables may be prepared in any conventional form, for example as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • excipients that may be used in conjunction with inj ectables according to the present invention include, but are not limited to water, saline, dextrose, glycerol or ethanol.
  • the injectable compositions may also optionally comprise 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. Implantation of a slow- release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Pat. No. 3,710,795) is also contemplated herein.
  • the percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject.
  • Parenteral administration of the formulations includes intravenous, subcutaneous and intramuscular administrations.
  • Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as the lyophilized powders described herein, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions.
  • the solutions may be either aqueous or nonaqueous.
  • suitable carriers include, but are not limited to physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • PBS physiological saline or phosphate buffered saline
  • Example of pharmaceutically acceptable carriers that may optionally be used in parenteral preparations include, but are not limited to aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
  • aqueous vehicles tiiat may optionally be used include Sodium Chloride
  • nonaqueous parenteral vehicles examples include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
  • Antimicrobial agents in bacteriostatic or fungistatic concentrations may be added to parenteral preparations, particularly when the preparations are packaged in multiple-dose containers and thus designed to be stored and multiple aliquots to be removed.
  • antimicrobial agents include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
  • Examples of isotonic agents that may be used include sodium chloride and dextrose.
  • Examples of buffers that may be used include phosphate and citrate.
  • antioxidants that may be used include sodium bisulfate.
  • Examples of local anesthetics that may be used include procaine hydrochloride.
  • Examples of suspending and dispersing agents that may be used include sodium carboxymethylcellulose, hydroxypropyl methylcellulose and polyvinylpyrrolidone.
  • Examples of emulsifying agents that may be used include Polysorbate 80 (Tween 80).
  • a sequestering or chelating agent of metal ions include EDTA.
  • Pharmaceutical carriers may also optionally include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • the concentration of a HDAC inhibitor in the parenteral formulation may be adjusted so that an injection administers a pharmaceutically effective amount sufficient to produce the desired pharmacological effect.
  • concentration of a HDAC inhibitor and/or dosage to be used will ultimately depend on the age, weight and condition of the patient or animal as is known in the art.
  • Unit-dose parenteral preparations may be packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration should be sterile, as is know and practiced in the art.
  • Injectables may be designed for local and systemic administration. Typically a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1 % w/w up to about 90% w/w or more, preferably more than 1% w/w of the HDAC inhibitor to the treated tissue(s).
  • the HDAC inhibitor may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment will be a function of d e location of where the composition is parenterally administered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data.
  • concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens may need to be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. Hence, the concentration ranges set forth herein are intended to be exemplary and are not intended to limit the scope or practice of the claimed formulations.
  • the HDAC inhibitor may optionally be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease state and may be empirically determined.
  • the HDAC inhibitors of the present invention may also be prepared as lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures.
  • the lyophilized powders may also be formulated as solids or gels.
  • Sterile, lyophilized powder may be prepared by dissolving the sodium salt in a sodium phosphate buffer solution containing dextrose or other suitable excipient. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation.
  • the lyophilized powder may optionally be prepared by dissolving dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, about 1-20%, preferably about 5 to 15%, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in die art at, typically, about neutral pH.
  • a suitable buffer such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in die art at, typically, about neutral pH.
  • a HDAC inhibitor is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35 °C, and stirred until it dissolves.
  • the resulting mixture is diluted by adding more buffer to a desired concentration.
  • the resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization.
  • Each vial may contain a single
  • the HDAC inhibitors of the present invention may also be administered as topical mixtures.
  • Topical mixtures may be used for local and systemic administration.
  • the resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
  • the HDAC inhibitors may be formulated as aerosols for topical application, such as by inhalation (see, U.S. Pat. Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma).
  • These formulations for administration to the respiratory tract 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 typically diameters of less than 50 microns, preferably less than 10 microns.
  • the HDAC inhibitors may also be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the HDAC inhibitor alone or in combination with other pharmaceutically acceptable excipients can also be administered.
  • rectal administration Depending upon the disease state being treated, otiier routes of administration, such as topical application, transdermal patches, a rectal administration, may also be used.
  • pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect.
  • Rectal suppositories are used herein mean solid bodies for insertion into the rectum that melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients.
  • Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point.
  • bases examples include cocoa butter (theobroma oil), glycerin-gelatin, carbowax, (polyoxyetiiylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used.
  • Agents to raise the melting point of suppositories include spermaceti and wax.
  • Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm. Tablets and capsules for rectal administration may be manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
  • Citric Acid Monohydrate 1.05 mg
  • the invention is also directed to kits and other articles of manufacture for treating diseases associated with HDAC. It is noted that diseases are intended to cover all conditions for which the HDAC possesses activity that contributes to the pathology and/or symptomology of the condition.
  • kits comprising a composition comprising at least one HDAC inhibitor of the present invention in combination with instructions.
  • the instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also comprise packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • an article of manufacture comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with packaging materials.
  • the packaging material may comprise a container for housing the composition.
  • the container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition.
  • the kit may also optionally comprise additional components, such as syringes for administration of the composition.
  • the kit may comprise the composition in single or multiple dose forms.
  • the packaging material used in kits and articles of manufacture according to the present invention may form a plurality of divided containers such as a divided bottle or a divided foil packet.
  • the container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • a pharmaceutically acceptable material for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • the container that is employed will depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle that is in
  • the Idt form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, topical, transdermal and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • different dosage forms e.g., oral, topical, transdermal and parenteral
  • Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed.
  • the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet.
  • the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
  • kits are a dispenser designed to dispense the daily doses one at a time in the order of their intended use.
  • the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen.
  • a memory-aid is a mechanical counter that indicates the number of daily doses that has been dispensed.
  • a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
  • a wide variety therapeutic agents may have a therapeutic additive or synergistic effect with HDAC inhibitors according to the present invention.
  • Such therapeutic agents may additively or synergistically combine with the HDAC inhibitors to inhibit undesirable cell growth, such as inappropriate cell growti resulting in undesirable benign conditions or tumor growth.
  • a method for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated witi the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy.
  • combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
  • Examples of therapeutic agents that may be used in combination with HDAC inhibitors include, but are not limited to, antieancer agents, alkylating agents, antibiotic agents, antimetabolic agents, hormonal agents, plant-derived agents, and biologic agents.
  • Alkylating agents are polyfunctional compounds that have the ability to substitute alkyl groups for hydrogen ions. Examples of alkylating agents include, but are not limited to, bischloroethylamines (nitrogen mustards, e.g. chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, uracil mustard), aziridines (e.g.
  • alkyl alkone sulfonates e.g. busulfan
  • nitrosoureas e.g. carmustine, lomustine, streptozocin
  • nonclassic alkylating agents altretamine, dacarbazine, and procarbazine
  • platinum compounds carboplastin and cisplatin.
  • HDAC inhibitor and an alkylating agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
  • Antibiotic agents are a group of drugs that produced in a manner similar to antibiotics as a modification of natural products.
  • antibiotic agents include, but are not limited to, anthracyclines (e.g. doxorubicin, daunorubicin, epirabicin, idarubicin and anthraeenedione), mitomycin C, bleomycin, dactinomycin, plicatomycin.
  • anthracyclines e.g. doxorubicin, daunorubicin, epirabicin, idarubicin and anthraeenedione
  • mitomycin C e.g. doxorubicin, daunorubicin, epirabicin, idarubicin and anthraeenedione
  • mitomycin C e.g. doxorubicin, daunorubicin, epirabicin, idarubicin and anthraeenedione
  • mitomycin C e.
  • Bleomycin is generally believed to chelate iron and forms an activated complex, which then binds to bases of DNA, causing strand scissions and cell death.
  • Combination therapy including a HDAC inhibitor and an antibiotic agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotiierapeutic agents.
  • Antimetabolic agents are a group of drugs that interfere with metabolic processes vital to the physiology and proliferation of cancer cells. Actively proliferating cancer cells require continuous synthesis of large quantities of nucleic acids, proteins, lipids, and other vital cellular constituents. Many of the antimetabolites inhibit the synthesis of purine or pyrimidine nucleosides or inhibit the enzymes of DNA replication.
  • antimetabolites also interfere with the synthesis of ribonucleosides and RNA and/or amino acid metabolism and protein synthesis as well. By interfering with the synthesis of vital cellular constituents, antimetabolites can delay or arrest the growth of cancer cells.
  • antimetabolic agents include, but are not limited to, fluorouracil (5-FU), floxuridine (5-FUdR), methotrexate, leucovorin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6-MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, and gemcitabine.
  • Combination therapy including a HDAC inhibitor and a antimetabolic agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
  • Hormonal agents are a group of drug that regulate the growth and development of their target organs. Most of the hormonal agents are sex steroids and their derivatives and analogs thereof, such as estrogens, androgens, and progestins. These hormonal agents may serve as antagonists of receptors for the sex steroids to down regulate receptor expression and transcription of vital genes. Examples of such hormonal agents are synthetic estrogens (e.g. diethylstibestrol), antiestrogens (e.g.
  • tamoxifen toremifene, fluoxymesterol and raloxifene
  • antiandrogens bicalutamide, nilutamide, flutamide
  • aromatase inhibitors e.g., aminoglutethimide, anastrozole and tetrazole
  • ketoconazole goserelin acetate, leuprolide, megestrol acetate and mifepristone.
  • Combination tiierapy including a HDAC inhibitor and a hormonal agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with tiiese chemotherapeutic agents.
  • Plant-derived agents are a group of drugs that are derived from plants or modified based on the molecular structure of the agents.
  • plant-derived agents include, but are not limited to, vinca alkaloids (e.g., vincristine, vinblastine, vindesine, vinzolidine and vinorelbine), podophyllotoxins (e.g., etoposide (VP-16) and teniposide (VM-26)), taxanes (e.g., paclitaxel and docetaxel).
  • vinca alkaloids e.g., vincristine, vinblastine, vindesine, vinzolidine and vinorelbine
  • podophyllotoxins e.g., etoposide (VP-16) and teniposide (VM-26)
  • taxanes e.g., paclitaxel and docetaxel.
  • Podophyllotoxins such as etoposide are believed to interfere with DNA synthesis by interacting with topoisomerase ⁇ , leading to DNA strand scission.
  • Combination therapy including a HDAC inhibitor and a plant-derived agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
  • Biologic agents are a group of biomolecules that elicit cancer/tumor regression when used alone or in combination with chemotherapy and/or radiotherapy.
  • biologic agents include, but are not limited to, immuno-modulating proteins such as cytokines, monoclonal antibodies against tumor antigens, tumor suppressor genes, and cancer vaccines.
  • Combination therapy including a HDAC inhibitor and a biologic agent may have therapeutic synergistic effects on cancer, enhance the patient's immune responses to tumorigenic signals, and reduce potential sides affects associated with this chemotherapeutic agent.
  • Cytokines possess profound immunomodulatory activity.
  • Interleukin-2 interleukin-2
  • interleukin-2 aldesleukin
  • interferon have demonstrated antitumor activity and have been approved for the treatment of patients with metastatic renal cell carcinoma and metastatic malignant melanoma.
  • IL-2 is a T-cell growth factor that is central to T-cell-mediated immune responses. The selective antitumor effects of IL-2 on some patients are believed to be the result of a cell-mediated immune response that discriminate between self and nonself.
  • Examples of interleukins that may be used in conjunction with HDAC inhibitor include, but are not limited to, interleukin 2 (IL-2), and interleukin 4 (TL-4), interleukin 12 (IL-12).
  • Interferon include more than 23 related subtypes with overlapping activities, all of the IFN subtypes within the scope of the present invention. IFN has demonstrated activity against many solid and hematologic malignancies, the later appearing to be particularly sensitive.
  • immuno-modulating agents include, but are not limited to bacillus Calmette-Guerin, levamisole, and octreotide, a long-acting octapeptide that mimics the effects of the naturally occurring hormone somatostatin.
  • Monoclonal antibodies against tumor antigens are antibodies elicited against antigens expressed by tumors, preferably tumor-specific antigens.
  • monoclonal antibody HERCEPTIN® (Trastruzumab) is raised against human epidermal growth factor receptor2 (HER2) that is overexpressed in some breast tumors including metastatic breast cancer. Overexpression of HER2 protein is associated with more aggressive disease and poorer prognosis in the clinic.
  • HERCEPTIN® is used as a single agent for the treatment of patients with metastatic breast cancer whose tumors over express the HER2 protein.
  • Combination therapy including HDAC inhibitor and HERCEPTIN® may have therapeutic synergistic effects on tumors, especially on metastatic cancers.
  • RITUXAN® (Rituximab) that is raised against CD20 on lymphoma cells and selectively deplete normal and malignant CD20 + pre-B and mature B cells.
  • RITUXAN® is used as single agent for the treatment of patients with relapsed or refractory low-grade or follicular, CD20+, B cell non- Hodgkin' s lymphoma.
  • Combination therapy including HDAC inhibitor and RITUXAN® may have therapeutic synergistic effects not only on lymphoma, but also on other forms or types of malignant tumors.
  • Tumor suppressor genes are genes that function to inhibit the cell growth and division cycles, thus preventing the development of neoplasia. Mutations in tumor suppressor genes cause the cell to ignore one or more of the components of the network of inhibitory signals, overcoming the cell cycle check points and resulting in a higher rate of controlled cell growth — cancer. Examples of the tumor suppressor genes include, but are not limited to, DPC-4, NF-1, NF-2, RB, p53, WT1, BRCA1 and BRCA2.
  • DPC-4 is involved in pancreatic cancer and participates in a cytoplasmic pathway that inhibits cell division.
  • NF-1 codes for a protein that inhibits Ras, a cytoplasmic inhibitory protein.
  • NF-1 is involved in neurofibroma and pheochromocytomas of the nervous system and myeloid leukemia.
  • NF-2 encodes a nuclear protein that is involved in meningioma, schwanoma, and ependymoma of the nervous system.
  • RB codes for the pRB protein, a nuclear protein that is a major inhibitor of cell cycle. RB is involved in retinoblastoma as well as bone, bladder, small cell lung and breast cancer.
  • P53 codes for p53 protein that regulates cell division and can induce apoptosis. Mutation and/or inaction of p53 is found in a wide ranges of cancers. WT1 is involved in Wilm's tumor of the kidneys. BRCA1 is involved in breast and ovarian cancer, and BRCA2 is involved in breast cancer. The tumor suppressor gene can be transferred into the tumor cells where it exerts its tumor suppressing functions. Combination therapy including a HDAC inhibitor and a tumor suppressor may have therapeutic synergistic effects on patients suffering from various forms of cancers. [0310] Cancer vaccines are a group of agents that induce the body's specific immune response to tumors. Most of cancer vaccines under research and development and clinical trials are tumor-associated antigens (TAAs).
  • TAAs tumor-associated antigens
  • TAA are structures (i.e. proteins, enzymes or carbohydrates) which are present on tumor cells and relatively absent or diminished on normal cells. By virtue of being fairly unique to the tumor cell, TAAs provide targets for the immune system to recognize and cause their destruction.
  • Example of TAAs include, but are not limited to gangliosides (GM2), prostate specific antigen (PSA), alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA) (produced by colon cancers and other adenocarcinomas, e.g.
  • breast, lung, gastric, and pancreas cancer s melanoma associated antigens
  • MART-1 gplOO, MAGE 1,3 tyrosinase
  • papillomavirus E6 and E7 fragments whole cells or portions/lysates of antologous tumor cells and allogeneic tumor cells.
  • An adjuvant may be used to augment the immune response to TAAs.
  • adjuvants include, but are not limited to, bacillus Calmette-Guerin (BCG), endotoxin lipopolysaccharides, keyhole limpet hemocyanin (GKLH), interleukin-2 (IL-2), granulocyte- macrophage colony-stimulating factor (GM-CSF) and cytoxan, a chemotherapeutic agent which is believe to reduce tumor-induced suppression when given in low doses.
  • BCG Bacillus Calmette-Guerin
  • GKLH keyhole limpet hemocyanin
  • IL-2 interleukin-2
  • GM-CSF granulocyte- macrophage colony-stimulating factor
  • cytoxan a chemotherapeutic agent which is believe to reduce tumor-induced suppression when given in low doses.
  • Compound s according to the present invention may be screened for activity against one or more HDACs.
  • HDACs Provided in tiiis example are assays for activity against HDACl, HDAC2, HDAC6 and HDAC8.
  • HDACl DNA encoding residues 1-482 of the full-length sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Xbal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the N-terminus.
  • SEQ. ID. No. 1 corresponds to residues 1-482 with the N-terminal 6-histidine tag and SEQ. ID. No. 2 is the DNA sequence that was used to encode SEQ. ID. No. 1.
  • DNA encoding residues 1-488 of the full-length sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Smal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the C-terminus.
  • SEQ. ID. No. 3 corresponds to residues 1-488 with the C-terminal 6-histidine tag and SEQ. ID. No. 4 is the DNA sequence that was used to encode SEQ. ID. No. 3.
  • DNA encoding residues 73-845 of the human enzyme may be amplified by PCR and cloned into the Smal site of pFastbac (Invitrogen), which incorporates a 6xHistidine tag at the C-terminus.
  • SEQ. I.D. No.5 corresponds to residues 73-845 with the C- terminal 6-histidine tag and SEQ. ID. No. 6 is the DNA sequence that was used to encode SEQ. ID. No. 5.
  • DNA encoding residues 1 -377 corresponding to the entire sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Smal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the N-terminus.
  • SEQ. ID. No.7 corresponds to residues 1-377 with the N-terminal 6-histidine tag and SEQ. ID. No. 8 is the DNA sequence that was used to encode SEQ. ID. No. 7.
  • Recombinant baculo virus incorporating the HDAC constructs may be generated by transposition using the Bac-to-Bac system (Invitrogen).
  • High-titer viral stocks may be generated by infection of Spodoptera frugiperda Sf9 cells; the expression of recombinant protein may be carried out by infection of Spodoptera frugiperda Sf9 or Trichoplusia ni Hi5 cells (Invitrogen) in 10L Wave Bioreactors (Wave Biotech).
  • Recombinant protein may be isolated from cellular extracts by passage over ProBond resin (Invitrogen). HDAC 1 and HDAC6 may then be treated with TEV protease for the removal of the N-terminal 6XHistidine affinity tag (residual uncleaved protein may be removed through a second passage over Probond Resin). Partially purified extracts of all HDACs may then be further purified by high pressure liquid chromatography over a BioSep S3000 gel filtration resin. The purity of HDAC proteins may be determined on denaturing SDS-PAGE gel.
  • Purified HDACs may then be concentrated to a final concentration of 4.0 mg/ml for HDACl , 10 mg/ml for HDAC2, 4.0 mg/ml for HDAC6, and 3 mg/ml for HDAC8.
  • the proteins may be either stored at -78 °C in a buffer containing 25mM TRIS-HCl pH 7.6, 150mM NaCI, O.lmM EDTA and 0.25 mM TCEP or at -20 °C in the presence of glycerol (final concentration of glycerol at 50%)
  • the inhibitory properties of compounds relative to HDACl , HDAC2, HDAC6 and HDAC8 may be determined using a white or black 384- well-plate format under the following reaction conditions: 25 mM Tris pH 8.0, 100 mM NaCI, 50 mM KC1, 0.1 mM EDTA, 0.01% Brij35, 0.1 mM TCEP. 50 uM tBoc-Lys(Ac)-AMC, 2% DMSO.
  • Reaction product may be determined quantitatively by fluorescence intensity using a Fluorescence plate reader (Molecular Devices Gemini) with an excitation wavelength at 370 nm and emission at 480 nm (for white plates) or 465 nm (for black plates).
  • the assay reaction may be initiated as follows: 5 ul of 150 uM tBoc-Lys(Ac)AMC was added to each well of the plate, followed by the addition of 5 ul of inhibitor (2 fold serial dilutions for 11 data points for each inhibitor) containing 6% DMSO. 5 ul of either HDAC 1 , HDAC2, HDAC6 or HDAC8 solution may be added to initiate the reaction (final enzyme concentrations were 2.5 nM for HDACl, 1 nM for HDAC2, 2.5 nM for HDAC6 and 10 nM for HDAC8).
  • the reaction mixture may then be incubated at room temperature for 60 min, and quenched and developed by addition of 5 ul of 10 mM phenanthroline and 4 mg/ml trypsin (final concentration of phenanthroline is 2.5 mM, and trypsin is 1 mg/ml). Fluorescence intensities of the resulting reaction mixtures may be measured after a 30 minute incubation at room temperature.
  • IC50 values may be calculated by non-linear curve fitting of the compound concentrations and fluorescence intensities to the standard IC50 equation.
  • SAHA suberanilohydroxamic acid
  • HDAC inhibitors according to the present invention may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided herein in the examples. Other reaction schemes could be readily devised by those skilled in the art.
  • N-substituted-2-nitrophenylamine (1.10 mmol) was dissolved in MeOH/AcOH (4: 1, 5.0 mL) and heated to 100 °C. Zinc dust (5.50 mmol) was added to the reaction portion wise until frothing ceased. The reaction was cooled to ambient temperature, filtered, and evaporated to dryness. The resulting N ⁇ substituted-phenyl-l ⁇ -diamine (2) was used without further purification.
  • the product is obtained in salt form with two 10-camphorsulfonic acid (CSA).
  • the product is obtained in salt form with two 10-camphorsulfonic acid (CSA).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • Communicable Diseases (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Surgery (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Compounds that may be used to inhibit histone deacetylase having the formula (I) or formula (II) wherein M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound; and Z and Q are as defined herein.

Description

HISTONE DEACETYLASE INHIBITORS
FIELD OF THE INVENTION
[0001] The invention relates to compounds that may be used to inhibit deacetylases and, in one variation, histone deacetylases (HDACs), as well as compositions of matter and kits comprising these compounds. The present invention also relates to methods for inhibiting deacetylases, such as HDAC, as well as treatment methods using compounds according to the present invention.
DESCRIPTION OF RELATED ART
[0002] DNA in eukaryotic cells is tightly complexed with proteins (histones) to form chromatin. Histones are small, positively charged proteins that are rich in basic amino acids (positively charged at physiological pH), which contact the phosphate groups (negatively charged at physiological pH) of DNA. There are five main classes of histones HI, H2A, H2B, H3, and H4. The amino acid sequences of H2A, H2B, H3, and H4 show remarkable conservation between species, wherein HI varies somewhat and in some cases is replaced by another histone, e.g., H5. Four pairs of each of H2A, H2B, H3 and H4 together form a disk- shaped octomeric protein core, around which DNA (about 140 base pairs) is wound to form a nucleosome. Individual nucleosomes are connected by short stretches of linker DNA associated with another histone molecule to form a structure resembling a beaded string, which is itself arranged in a helical stack, known as a solenoid.
[0003] The majority of histones are synthesized during the S phase of the cell cycle, and newly synthesized histones quickly enter the nucleus to become associated with DNA. Within minutes of its synthesis, new DNA becomes associated with histones in nucleosomal structures.
[0004] A small fraction of histones, more specifically, the amino acid side chains thereof, are enzymatically modified by post-translational addition of methyl, acetyl, or phosphate groups, neutralizing the positive charge of the side chain, or converting it to a negative charge. For example, lysine and arginine groups may be methylated, lysine groups may be acetylated, and serine groups may be phosphorylated. For lysine, the -(CH2) 4-NH2 sidechain may be acetylated, for example by an acetyltransferase enzyme to give the amide -(CH2) - NHC(=O)CH3. Methylation, acetylation, and phosphorylation of amino termini of histones that extend from the nucleosomal core affects chromatin structure and gene expression. Spencer and Davie 1999. Gene 240:1 1-12.
[0005] Acetylation and deacetylation of histones is associated with transcriptional events leading to cell proliferation and/or differentiation. Regulation of the function of transcriptional factors is also mediated through acetylation. Recent reviews on histone deacetylation include Kouzarides, et al., 1999. Curr. Opin. Genet. Dev.9: 1, 40-48 and Pazin, et al. 1997. 89:3 325-328.
[0006] The correlation between acetylation status of histones and the transcription of genes has been known for quite some time. Certain enzymes, specifically acetylases (e.g., histone acetyltransf erases (HAT)) and deacetylases (histone deacetylases or HDACs), which regulate the acetylation state of histones have been identified in many organisms and have been implicated in the regulation of numerous genes, confirming a link between acetylation and transcription. In general, histone acetylation is believed to correlate with transcriptional activation, whereas histone deacetylation is believed to be associated with gene repression. [0007] A growing number of histone deacetylases (HDACs) have been identified. HDACs function as part of large multiprotein complexes, which are tethered to the promoter and repress transcription. Well characterized transcriptional repressors such as MAD, nuclear receptors and YY1 associate with HDAC complexes to exert their represser function. [0008] Studies of HDAC inhibitors have shown that these enzymes play an important role in cell proliferation and differentiation. HDACs are believed to be associated with a variety of different disease states including, but not limited to cell proliferative diseases and conditions (Marks, P.A., Richon, V.M., Breslow, R. and Rifkind, R.A., J. Natl. Cancer lust. (Bethesda) 92, 1210-1215, 2000) such as leukemia (Lin et al. 1998. Nature 391: 811-814; Grignani, et al.1998. Nature 391: 815-818; Warrell et al. 1998. J. Natl. Cancer Inst. 90:1621-1625; Gelmetti et al. 1998. Mol. Cell Biol. 18:7185-7191; Wang et al. 1998. PNAS 951 0860- 10865), melanomas/squamous cell carcinomas (Gillenwater, et al., 1998, Int. J. Cancer 75217- 224; Saunders, et al., 1999, Cancer Res.59:399-404), breast cancer, prostrate cancer, bladder cancer (Gelmetti et al. 1998. Mol. Cell Biol. 18:7185-7191; Wang et al. 1998. PNAS 951 0860-10865), lung cancer, ovarian cancer and colon cancer (Hassig, et al., 1997, Chem. Biol. 4:783-789; Archer, et al., 1998, PNAS, 956791-6796; Swendeman, et al., 1999, Proc. Amer. Assoc. Cancer Res. 40, Abstract #3836).
[0009] Histone deacetylase inhibitors are potent inducers of growth arrest, differentiation, or apoptotic cell death in a variety of transformed cells in culture and in tumor bearing animals (Histone deacetylase inhibitors as new cancer drugs, Marks, P.A., Richon, V.M., Breslow, R. and Rifkind, R.A., Current Opinions in Oncology, 2001, Nov. 13 (6): 477-83; Histone deacetylases and cancer: causes and therapies, Marks, P., Rifkind, R.A., Richon, V.M., Breslow, R., Miller, T. and Kelly, W.K., Nat. Rev. Cancer 2001 Dec.l (3): 194-202). In addition, HDAC inhibitors are useful in the treatment or prevention of protozoal diseases (US Patent 5,922,837) and psoriasis (PCT Publication No. WO 02/26696). [0010] A variety of inhibitors of HDAC have been reported. Some of these inhibitors are described in the following table:
Figure imgf000005_0001
[0011] Additional examples of HDAC inhibitors can be found in Marks PA, et al., J. Natl. Cancer Inst.2000, 92: 1210-1216 & Weidle UH, et al., Antieancer Res.2000, 20: 1471-1486 and PCT Publication Nos. WO 02/26696, WO 02/062773, and WO 01/18171. [0012] Despite the various HDAC inhibitors that have been reported to date, a need continues to exist for new and more effective inhibitors of HDACs.
SUMMARY OF THE INVENTION
[0013] The present invention relates to compounds that have activity for inhibiting HDACs.
[0014] The present invention also provides compositions, articles of manufacture and kits comprising these compounds.
[0015] In one embodiment, a pharmaceutical composition is provided that comprises a HDAC inhibitor according to the present invention as an active ingredient. Pharmaceutical compositions according to the invention may optionally comprise 0.001 %-100% of one or more HDAC inhibitors of this invention. These pharmaceutical compositions may be administered or coadministered by a wide variety of routes, including for example, orally, parenterally, mtraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The compositions may also be administered or coadministered in slow release dosage forms.
[0016] The invention is also directed to kits and other articles of manufacture for treating disease states associated with HDAC.
[0017] In one embodiment, a kit is provided that comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with instructions. The instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also comprise packaging materials. The packaging material may comprise a container for housing the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms. [0018] In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with packaging materials. The packaging material may comprise a container for housing the composition. The container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.
[0019] Also provided are methods for preparing compounds, compositions and kits according to the present invention. For example, several synthetic schemes are provided herein for synthesizing compounds according to the present invention.
[0020] Also provided are methods for using compounds, compositions, kits and articles of manufacture according to the present invention.
[0021] In one embodiment, the compounds, compositions, kits and articles of manufacture are used to inhibit HDAC.
[0022] In one embodiment, the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state.
[0023] In another embodiment, a compound is administered to a subject wherein HDAC activity within the subject is altered, preferably reduced.
[0024] In another embodiment, a prodrug of a compound is administered to a subj ect that is converted to the compound in vivo where it inhibits HDAC.
[0025] In another embodiment, a method of inhibiting HDAC is provided that comprises contacting HDAC with a compound according to the present invention.
[0026] In another embodiment, a method of inhibiting HDAC is provided that comprises causing a compound according to the present invention to be present in a subject in order to inhibit HDAC in vivo.
[0027] In another embodiment, a method of inhibiting HDAC is provided that comprises administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo. [0028] In another embodiment, a therapeutic method is provided that comprises administering a compound according to the present invention.
[0029] In another embodiment, a method of inhibiting cell proliferation is provided that comprises contacting a cell with an effective amount of a compound according to the present invention.
[0030] In another embodiment, a method of inhibiting cell proliferation in a patient is provided that comprises administering to the patient a therapeutically effective amount of a compound according to the present invention.
[0031] In another embodiment, a method of treating a condition in a patient which is known to be mediated by HDAC, or which is known to be treated by HDAC inhibitors, comprising administering to the patient a therapeutically effective amount of a compound according to the present invention.
[0032] In another embodiment, a method is provided for using a compound according to the present invention in order to manufacture a medicament for use in the treatment of disease state which is known to be mediated by HDAC, or which is known to be treated by HDAC inhibitors.
[0033] In another embodiment, a method is provided for treating a disease state for which
HDAC possesses activity that contributes to the pathology and/or symptomology of d e disease state, the method comprising: causing a compound according to the present invention to be present in a subject in a therapeutically effective amount for the disease state.
[0034] In another embodiment, a method is provided for treating a disease state for which
HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a first compound to a subject that is converted in vivo to a second compound such that the second compound is present in the subject in a therapeutically effective amount for the disease state.
[0035] In another embodiment, a method is provided for treating a disease state for which
HDAC possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering a compound according to the present invention to a subject such that the compound is present in the subject in a therapeutically effective amount for the disease state. [0036] In another embodiment, a method is provided for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated with the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy. It is noted that treatment of one agent before another is referred to herein as sequential therapy, even if the agents are also administered together. It is noted that combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
[0037] Examples of diseases that may be treated by administration of compounds and compositions according to the present invention include, but are not limited to protozoal diseases and cell proliferative diseases and conditions such as leukemia, melanomas, squamous cell carcinomas, breast cancer, prostrate cancer, bladder cancer, lung cancer including non small-cell lung cancer and small-cell lung cancer, ovarian cancer, colon cancer, squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, bladder cancer, head and neck cancer, glioma, colorectal cancer, genitourinary cancer and gastrointestinal cancer. [0038] It is noted in regard to all of the above embodiments ti at the present invention is intended to encompass pharmaceutically acceptable salts, biohydrolyzable esters, biohydrolyzable amides, biohydrolyzable carbamates, and solvates (e.g., hydrates) of the compounds, regardless of whether such salts, esters, amides, carbamates and solvates are specified since it is well know in the art to administer pharmaceutical agents in a salt, ester, amide, carbamate or solvated form. It is further noted that prodrugs may also be administered which are altered in vivo and become a compound according to the present invention. For example, the compound optionally comprises a substituent that is convertible in vivo to a different substituent, such as hydrogen. Accordingly, for example, an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di- -toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like. Similarly, an inhibitor comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound.
[0039] The various methods of using the compounds of the present invention are intended, regardless of whether prodrug delivery is specified, to encompass the administration of a prodrug that is converted in vivo into a compound according to the present invention.
BRIEF DESCRIPTION OF THE FIGURES
[0040] Figure 1 illustrates a ribbon diagram overview of the structure of HDAC8, highlighting the secondary structural elements of the protein.
[0041] Figure 2A illustrates particular examples of substituent Ri that may be employed in the Z moiety.
[0042] Figure 2B illustrates particular examples of Z moieties that the compounds of the present invention may comprise.
[0043] Figure 2C illustrates examples of moieties, Q, that the leader group may comprise to link the leader group (L) to the remainder of the compound.
[0044] Figure 2D illustrates particular examples of moieties that the leader groups may comprise.
[0045] It is noted in regard to Figures 2A-2D that the squiggle line is intended to indicate a bond to an adjacent moiety. It is also noted that the substituents shown may optionally be further substituted beyond what is shown. Further, one or more heteroatoms may optionally be substituted for the carbon atoms shown. In regard to Figure 2D, it is noted that the leader groups moieties may be incoφorated into the leader group in either possible orientation.
[0046] Figure 3 illustrates residues 1-482 of HDACl and a 6-histidine tag at the N- terminus (SEQ. I.D. No. 1).
[0047] Figure 4 illustrates the DNA sequence (SEQ. ID. No.2) that was used to encode
SEQ. ID. No. 1.
[0048] Figure 5 illustrates residues 1-488 of HDAC2 and a 6-histidine tag at the C- terminus (SEQ. ID. No. 3).
[0049] Figure 6 illustrates the DNA sequence (SEQ. ID. No.4) that was used to encode
SEQ. I.D. No. 3. [0050] Figure 7 illustrates residues 73-845 of HDAC6 and a 6-histidine tag at the C- terminus (SEQ. ID. No. 5).
[0051] Figure 8 illustrates the DNA sequence (SEQ. ID. No. 6) that was used to encode
SEQ. ID. No. 5.
[0052] Figure 9 illustrates residues 1-377 of HDAC8 and a 6-histidine tag at the N- terminus (SEQ. ID. No. 7).
[0053] Figure 10 illustrates the DNA sequence (SEQ. ID. No.8) that was used to encode
SEQ. ID. No. 7.
DEFINITIONS
[0054] Unless otherwise stated, the following terms used in the specification and claims shall have the following meanings for the puφoses of this Application.
[0055] "Alicyclic" means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one or more double or triple bonds.
Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. Examples of alicyclic moieties include, but are not limited to moieties with C3 - C8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
[0056] "Aliphatic" means a moiety characterized by a. straight or branched chain arrangement of constituent carbon atoms and may be saturated or partially unsaturated with one or more double or triple bonds.
[0057] "Alkenyl" represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one double bond between adjacent carbon atoms. Cx alkenyl and Cχ.y alkenyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2.6 alkenyl includes alkenyls that have a chain of between 2 and 6 carbons.
[0058] "Alkoxy" means an oxygen moiety having a further alkyl substituent.
[0059] "Alkyl" represented by itself means a straight or branched, saturated or unsaturated, aliphatic radical having a chain of carbon atoms, optionally with oxygen (See
"oxaalkyl") or nitrogen atoms (See "aminoalkyl") between the carbon atoms. Cx alkyl and Cχ.γ alkyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C1-6 alkyl includes alkyls that have a chain of between 1 and 6 carbons (e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methylallyl, ethynyl, 1-propynyl, 2-propynyl, and the like). Alkyl represented along with another radical (e.g., as in arylalkyl) means a straight or branched, saturated or unsaturated aliphatic divalent radical having the number of atoms indicated or when no atoms are indicated means a bond (e.g., (C6-ιo)aryl(Co-3)alkyl includes phenyl, benzyl, phenethyl, 1-phenylethyl 3-phenylpropyl, and the like). [0060] "Alkylene" , unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical. Cx alkylene and Cχ.γ alkylene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C1-6 alkylene includes methylene (-CH2-), ethylene (-CH2CH2-), trimethylene (-CH2CH2CH2-), tetramethylene (-CH2CH2CH2CH2-) 2-butenylene (-CH2CH=CHCH2-),
2-methyltetramethylene (-CH2CH(CH3)CH2CH2-), pentamethylene (-CH2CH2CH2CH2CH2-) and the like).
[0061] " Alkylidene" means a straight or branched unsaturated, aliphatic, divalent radical having a general formula =CRaRb. Cx alkylidene and Cχ.γ alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C1-6 alkylidene includes methylidene (=CH2), ethylidene (=CHCH3), isopropylidene (=C(CH3)2), propylidene (=CHCH2CH3), allylidene (=CH-CH=CH2), and the like).
[0062] " Alkynyl" represented by itself means a straight or branched, unsaturated, aliphatic radical having a chain of carbon atoms having at least one triple bond between adjacent carbon atoms. Cx alkynyl and Cχ.γ alkynyl are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C2-6 alkynyl includes alkynyls that have a chain of between 2 and 6 carbons.
[0063] "Amino" means a nitrogen moiety having two further substituents where each substituent has a hydrogen or carbon atom alpha bonded to the nitrogen. Unless indicated otherwise, the compounds of the invention containing amino moieties may include protected derivatives thereof. Suitable protecting groups for amino moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, and the like. [0064] "Aminoalkyl" means an alkyl, as defined above, except where one or more substituted or unsubstituted nitrogen atoms (-N-) are positioned between carbon atoms of the alkyl. For example, an (C2-6) aminoalkyl refers to a chain comprising between 2 and 6 carbons and one or more nitrogen atoms positioned between the carbon atoms.
[0065] "Animal" includes humans, non-human mammals (e.g., dogs, cats, rabbits, cattle, horses, sheep, goats, swine, deer, and the like) and non-mammals (e.g., birds, and the like).
[0066] "Aromatic" means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp1 hybridized and the total number of pi electrons is equal to 4n+2. An aromatic ring may be such that the ring atoms are only carbon atoms or may include carbon and non-carbon atoms (see Heteroaryl).
[0067] "Aryl" means a monocyclic or fused bicyclic ring assembly wherein each ring is aromatic or when fused with a second ring forms an aromatic ring assembly. If one or more ring atoms is not carbon (e.g., N, S), the aryl is a heteroaryl. Cx aryl and Cχ.γ aryl are typically used where X and Y indicate the number of atoms in the ring.
[0068] "Bicycloalkyl" means a saturated or partially unsaturated fused bicyclic or bridged polycyclic ring assembly.
[0069] "Bicycloaryl" means a bicyclic ring assembly wherein the rings are linked by a single bond or fused and at least one of the rings comprising the assembly is aromatic. Cx bicycloaryl and Cχ.γ bicycloaryl are typically used where X and Y indicate the number of carbon atoms in the bicyclic ring assembly and directly attached to the ring.
[0070] "Carbamoyl" means the radical -OC(O)NRaRb where Ra and Rb are each independently two further substituents where a hydrogen or carbon atom is alpha to the nitrogen. It is noted that carbamoyl moieties may include protected derivatives thereof.
Examples of suitable protecting groups for carbamoyl moieties include acetyl, tert-butoxycarbonyl, benzyloxycarbonyl, and the like. It is noted that both the unprotected and protected derivatives fall within the scope of the invention.
[0071] "Carbocycle" means a ring consisting of carbon atoms.
[0072] "Carbocyclic ketone derivative" means a carbocyclic derivative having a -C(O)- substituent. [0073] "Carbonyl" means the radical -C(O)-. It is noted that the carbonyl radical may be further substituted with a variety of substituents to form different carbonyl groups including acids, acid halides, amides, esters, and ketones.
[0074] "Carboxy" means the radical -C(O)O-. It is noted that compounds of the invention containing carboxy moieties may include protected derivatives thereof, i.e., where the oxygen is substituted with a protecting group. Suitable protecting groups for carboxy moieties include benzyl, tert-butyl, and the like.
[0075] "Cyano" means the radical -CN.
[0076] "Cycloalkyl" means a non-aromatic, saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly. Cx cycloalkyl and Cχ.γ cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly. For example, C -10 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,5-cyclohexadienyl, bicyclo[2.2.2]octyl, adamantan-1-yl, decahydronaphthyl, oxocyclohexyl, dioxocyclohexyl, thiocyclohexyl, 2-oxobicyclo[2.2.1]hept-l-yl, and the like.
[0077] "Cycloalkylene" means a divalent saturated or partially unsaturated, monocyclic ring or bridged polycyclic ring assembly. C cycloalkylene and Cχ.γ cycloalkylene are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
[0078] "Disease" specifically includes any unhealthy condition of an animal or part thereof and includes an unhealthy condition that may be caused by, or incident to, medical or veterinary therapy applied to that animal, i.e., the "side effects" of such therapy.
[0079] "Halo" means fluoro, chloro, bromo or iodo.
[0080] "Halo-substituted alkyl", as an isolated group or part of a larger group, means
"alkyl" substituted by one or more "halo" atoms, as such terms are defined in this Application.
Halo-substituted alkyl includes haloalkyl, dihaloalkyl, trihaloalkyl, perhaloalkyl and the like
(e.g. halo-substituted (C1-3)alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-l,l-dichloroethyl, and the like).
[0081] "Heteroatom" refers to an atom that is not a carbon atom. Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, sulfur and halogens. [0082] "Heteroatom moiety" includes a moiety where the atom by which the moiety is attached is not a carbon. Examples of heteroatom moieties include -N=, -NRo-, -N+(O")=, -O-,
-S- or -S(O)2-, wherein Rc is further substituent.
[0083] "Heterobicycloalkyl" means bicycloalkyl, as defined in this Application, provided that one or more of the atoms forming the ring is a heteroatom. For example hetero(C9-12)bicycloalkyl as used to define Z in this application includes, but is not limited to,
3-aza-bicyclo[4.1.0]hept-3-yl, 2-aza-bicyclo[3.1.0]hex-2-yl , 3-aza-bicyclo[3.1.0]hex-3-yl, and the like.
[0084] "Heterocycloalkylene" means cycloalkylene, as defined in this Application, provided that one or more of the ring member carbon atoms indicated, is replaced by a heteroatom.
[0085] "Heteroaryl" means an aryl ring, as defined in this Application, where one or more of the atoms forming the ring is a heteroatom.
[0086] "Heterobicycloaryl" means bicycloaryl, as defined in this Application, provided that one or more of die atoms forming die ring is a heteroatom. For example, hetero(Cg-1o)bicycloaryl as used in this Application includes, but is not limited to, 2-amino-
4-0X0-3 ,4-dihydropteridin-6-yl, and the like.
[0087] "Heterocycloalkyl" means cycloalkyl, as defined in this Application, provided that one or more of the atoms forming the ring is a heteroatom.
[0088] "Hydroxy" means the radical -OH.
[0089] "Imine derivative" means a derivative comprising the moiety -C(NR)-, wherein R comprises a hydrogen or carbon atom alpha to the nitrogen.
[0090] "Isomers" mean any compound having an identical molecular formulae but differing in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed
"stereoisomers". Stereoisomers that are not mirror images of one another are termed
"diastereomers" and stereoisomers that are nonsuperimposable mirror images are termed
"enantiomers" or sometimes "optical isomers". A carbon atom bonded to four nonidentical substituents is termed a "chiral center". A compound with one chiral center has two enantiomeric forms of opposite chirality. A mixture of the two enantiomeric forms is termed a "racemic mixture". A compound that has more than one chiral center has 2 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as ether an individual diastereomers or as a mixture of diastereomers, termed a "diastereomeric mixture". When one chiral center is present a stereoisomer may be characterized by the absolute configuration of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. Enantiomers are characterized by the absolute configuration of their chiral centers and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog. Conventions for stereochemical nomenclature, methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (e.g., see "Advanced Organic Chemistry", 4th edition, March, Jerry, John Wiley & Sons, New York, 1992). [0091] "Nitro" means the radical -NO2.
[0092] "Oxaalkyl" means an alkyl, as defined above, except where one or more oxygen atoms (-O-) are positioned between carbon atoms of the alkyl. For example, an (C -6)oxaalkyl refers to a chain comprising between 2 and 6 carbons and one or more oxygen atoms positioned between the carbon atoms.
[0093] 'Oxoalkyl" means an alkyl, further substituted with a carbonyl group. The carbonyl group may be an aldehyde, ketone, ester, amide, acid or acid chloride. [0094] "Pharmaceutically acceptable" means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
[0095] "Pharmaceutically acceptable salts" means salts of inhibitors of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, efhanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like. [0096] Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like.
[0097] "Prodrug" means a compound that is convertible in vivo metabolically into an inhibitor according to the present invention. The prodrug itself may or may not also have HDAC inhibitory activity. For example, an inhibitor comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-b-hydroxynaphthoates, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates and quinates.
[0098] "Protected derivatives" means derivatives of inhibitors in which a reactive site or sites are blocked with protecting groups. Protected derivatives are useful in the preparation of inhibitors or in themselves may be active as inhibitors. A comprehensive list of suitable protecting groups can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999.
[0099] "Substituted or unsubstituted" means that a given moiety may consist of only hydrogen substituents through available valencies (unsubstituted) or may further comprise one or more non-hydrogen substituents through available valencies (substituted) that are not otherwise specified by the name of the given moiety. For example, isopropyl is an example of an ethylene moiety that is substituted by -CH3. In general, a non-hydrogen substituent may be any substituent that may be bound to an atom of the given moiety that is specified to be substituted. Examples of substituents include, but are not limited to, aldehyde, alicyclic, aliphatic, alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocycle, carboxy, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted.
[0100] "Sulfinyl" means the radical -S(O)-. It is noted that the sulfinyl radical may be further substituted with a variety of substituents to form different sulfinyl groups including sulfinic acids, sulfinamides, sulfinyl esters, and sulfoxides.
[0101] "Sulfonyl" means the radical -S(O)(O)-. It is noted that the sulfonyl radical may be further substituted with a variety of substituents to form different sulfonyl groups including sulfonic acids, sulfonamides, sulfonate esters, and sulfones.
[0102] "Therapeutically effective amount" means that amount which, when administered to an animal for treating a disease, is sufficient to effect such treatment for the disease.
[0103] "Thiocarbonyl" means the radical -C(S)-. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including ti ioacids, thioamides, thioesters, and thioketones.
[0104] "Treatment" or "treating" means any administration of a compound of the present invention and includes:
( 1 ) preventing the disease from occurring in an animal which may be predisposed to the disease but does not yet experience or display the pathology or symptomatology of the disease,
(2) inhibiting the disease in an animal that is experiencing or displaying the pathology or symptomatology of the disease (i.e., arresting further development of the pati ology and/or symptomatology), or
(3) ameliorating the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., reversing the pathology and/or symptomatology) .
[0105] It is noted in regard to all of the definitions provided herein that the definitions should be inteφreted as being open ended in the sense that further substituents beyond those specified may be included. Hence, a Ci alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom. Hence, a Ci alkyl comprises methyl (i.e., -CH3) as well as -CRaRbRc where Ra, R , and Rc may each independently be hydrogen or any other substituent where the atom alpha to the carbon is a heteroatom or cyano. Hence, CF3, CH2OH and CH2CN are all Ci alkyls.
DETAILED DESCRIPTION OF THE INVENTION
[0106] The present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit histone deacetylases (referred to herein as HDACs). The compounds may optionally be more particularly used as inhibitors of Class I HDACs such as HDAC1, HDAC2, HDAC6 and HDAC8.
[0107] At least seventeen human genes that encode proven or putative HDACs have been identified to date, some of which are described in Johnstone, R. W., "Histone-Deacetylase Inhibitors: Novel Drugs for the Treatment of Cancer", Nature Reviews, Volume I, pp. 287- 299, (2002) and PCT Publication Nos. 00/10583, 01/18045, 01/42437 and 02/08273. [0108] HDACs have been categorized into tiiree distinct classes based on their relative size and sequence homology. The different HDACs (Homo sapiens), HDAC classes, sequences and references describing the different HDACs are provided in Tables 1 - 3.
TABLE 1: CLASS I HDACs
Figure imgf000019_0001
Figure imgf000020_0001
TABLE 2: CLASS II HDACs
Figure imgf000020_0002
TABLE 3: CLASS III HDACs
Figure imgf000020_0003
Figure imgf000021_0001
[0109] Of particular note are Class I HDACs. All Class I HDACs appear to be sensitive to inhibition by trichostatin A (TS A). Also of particular note is HDAC8, a protein whose crystal structure Applicants determined and used in conjunction with arriving at the present invention.
[0110] HDAC8 is a 377 residue, 42kDa protein localized to the nucleus of a wide array of tissues, as well as several human tumor cell lines. The wild-type form of full length HDAC8 is described in GenBank Accession Number NP 060956; Buggy ,JJ., Sideris,M.L., Mak,P., Lorimer,D.D., Mclhtosh,B. and Clark, J.M., Cloning and characterization of a novel human histone deacetylase, HDAC8, Biochem. J. 350 Pt 1, 199-205 (2000). Zn2+ is likely native to the protein and required for HDAC8 activity.
1. CRYSTAL STRUCTURE FOR HDAC
[0111] Syrrx, Inc. in San Diego, California recently solved the crystal structure for HDAC8. Knowledge of the crystal structure was used to guide the design of the HDAC inhibitors provided herein.
[0112] Figure 1 illustrates a ribbon diagram overview of the structure of HDAC8, highlighting the secondary structural elements of the protein. HDAC8 was found to have a single domain structure belonging to the open α/β class of folds. The structure consists of a central 8-stranded parallel β-sheet sandwiched between layers of α-helices. The ligand binding clefts lie almost in the plane of the central β-sheet, and are formed primarily by loops emanating from the carboxy-terminal ends of the β-strands comprising the sheet. There are two large structural extensions, which occur beyond the core of the α/β motif, off the second and last β-strands of the central β-sheet. Residues contained in the extension off the second β-strand form a globular "cap" over the core of the protein, play an important role in defining the shape of the ligand binding pockets, and are involved in a number of key interactions with the bound ligands.
2. HDAC INHIBITORS
[0113] In one embodiment, HDAC inhibitors of the present invention are provided that comprise the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000023_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen, and each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0114] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000024_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R , R3, R4 and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0115] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000025_0001
wherein each X is independently selected from the group consisting of CR1 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R6, R , R8( and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aiyloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
[0116] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000026_0001
wherein
each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R6, R , R8> and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
[0117] In another embodiment, HDAC inhibitors of die present invention are provided that comprise the formula:
Figure imgf000028_0001
wherein
each X is independently selected from the group consisting of CRι2 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R2, R3, R4, R5, R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
Lis a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0118] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000029_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R10 and Rπ are taken together to form a substituted or unsubstituted aromatic ring; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0119] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000030_0001
wherein
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R , R3, , and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound. [0120] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000031_0001
Ri is selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
R2, R3, R , and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
[0121] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein Z-Q- is selected from the group consisting of
Figure imgf000032_0001
Ri is selected from the group consisting of (C1-4)alkyl, phenyl, l-piperidin-4-ylmethyl, 2-morpholi-4-yl-ethyl, 2-halo-phenyl, 2-halo-phen(C1-4)alkyl,
Figure imgf000032_0002
2-CF3O- phen(C1-4)alkyl, 3-CF3O-phen(C1 )alkyl, 3-halo-phenyl, 4-halo-phenyl, 2-mefhoxy-phenyl, 3- methoxy-phenyl, 4-methoxy-phenyl, 4-phenoxy-phenyl, 4-benzyloxyphenyl, 4-pyrazol-l-yl- benzyl, 1-p-tolyl-eti yl, pyrrolidin-3-yl, l-(C1-4)alkyl-pyrrolidin-2-yl, l-(Ci )alkyl-pyrroιidin- 2-yl; 2-di(C1-4)alkylamino-ethyl, 2-di(C1- )alkylamino-l -methyl-ethyl, 2-di(Cι- )alkylamino- ethyl, 2-hydroxy-2-phenyl-ethyl, 2-pyridin-2-yl-ethyl, 2-pyridin-3-yl-ethyl, 2-pyridin-4-yl- ethyl, 2-(lH-indol-3-yl)-elhyl, 3-indolyl(C1-4)alkyl, l-indan-2-yl,
Figure imgf000032_0003
4)alkyl, S-α-(HOCH2)-phen(C1-4)alkyl, S-β-(HOCH2)-phen(C1-4)alkyl, i?-β-(CH3)-phen(Ci. 4)alk3 , 6-propylsulfanyl, trøns-4-hydroxy-cyclohexyl, l-aza-bicyclo[2.2.2]oct-2-yl, l-(Cι. 4)alkyl-piperidin-3-yl, l-(2,2-difluoro-ethyl)-piperidin-3-yl, (2-di(C1-4)alkylamino-2-phenyl- ethyl), l-benzyl-piperidin-3-yl, l-allyl-piperidin-3-yl, l-acetyl-piperidin-3-yl, piperidin-3-yl, and phen(C1-4)alkyl;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro;
M is selected from the group consisting of:
Figure imgf000032_0004
and L is E, Z or mixtures of E/Z -CH2=CH2- [0122] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000033_0001
wherein
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR1 ; each X is independently selected from the group consisting of CR1 and N; each Y is independently selected from the group consisting of O, S and NRι2; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring. [0123] In another embodiment, HDAC inhibitors of die present invention are provided that comprise the formula:
Z L M
wherein
Z is selected from the group consisting of
Figure imgf000034_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring. [0124] In another embodiment, HDAC inhibitors of the present invention are provided that comprise d e formula:
Figure imgf000034_0002
wherein
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; each Rι2 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0125] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the fonnula:
Z L M
wherein
Z is selected from the group consisting of
Figure imgf000035_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0126] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z L M
wherein
Z is selected from the group consisting of
Figure imgf000036_0001
Figure imgf000036_0002
Figure imgf000037_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NR12; each X is independently selected from the group consisting of CRι2 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R4 and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with d e proviso that R1 is not halo, cyano, nitro, and thio in the case where d e ring atom to which R1 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0127] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000038_0001
wherein
each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R6, R , R8> and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
[0128] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000039_0001
wherein
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each Rι is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen; M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
[0129] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000040_0001
wherein
each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NR1 ; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R , R5, R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
[0130] In another embodiment, HDAC inhibitors of die present invention are provided tiiat comprise the formula:
TL <Q> M
wherein
Z is selected from the group consisting of
Figure imgf000042_0001
Figure imgf000042_0002
wherein each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NRι2; each X is independently selected from the group consisting of CRι and N; each Y is independently selected from the group consisting of O, S and NR12;
R10 and Rπ are taken together to form a substituted or unsubstituted aromatic ring; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0131] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000043_0001
wherein
each V is independently selected from the group consisting of C(R12)2 andNR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each Rι2 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι is not halo, cyano, nitro, and thio in d e case where the ring atom to which R1 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
[0132] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein Z-Q- is selected from the group consisting of
Figure imgf000044_0001
each V is independently selected from the group consisting of C(R12)2 and NRι2 where at least one V is NR12;
R , R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R1 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aiyloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 2-10 atoms separation between M and the Q substituent.
[0133] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000045_0001
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NRι2;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in die case where the ring atom to which R12 is bound is nitrogen;
M is selected from the group consisting of
Figure imgf000045_0002
and L is E, Z or mixtures of E/Z -CH =CH2-. [0134] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000045_0003
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring. [0135] In another embodiment, HDAC inhibitors of d e present invention are provided tiiat comprise the formula:
Z L M
wherein
Z is selected from the group consisting of
Figure imgf000047_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; each R1 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso tiiat R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring. [0136] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000048_0001
wherein each X is independently selected from the group consisting of CRι and N; each Y is independently selected from the group consisting of O, S and NR12; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0137] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein Z is selected from the group consisting of
Figure imgf000049_0001
Figure imgf000049_0002
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
R2, R3, R4 and R5 are each independently selected from die group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0138] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
M
Figure imgf000050_0001
wherein
each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R6, R7, R8j and R are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Ri4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
[0139] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000051_0001
wherein
each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and die ring.
[0140] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Figure imgf000053_0001
wherein
each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R4, R5, R6, R7, R8j and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
[0141] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000054_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; R10 and Rπ are taken together to form a substituted or unsubstituted aromatic ring; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen;
Ri4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
[0142] In another embodiment, HDAC inhibitors of die present invention are provided that comprise the formula:
Figure imgf000055_0001
wherein
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
[0143] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000056_0001
wherein
R , R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 2-10 atoms separation between the M substituent and the Q substituent. [0144] In another embodiment, HDAC inhibitors of the present invention are provided that comprise the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000057_0001
R2, R3, R4, and R5 are each independently selected from die group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is selected from the group consisting of
Figure imgf000057_0002
and L is E, Z or mixtures of E/Z -CH2=CH2-. [0145] In one variation of any of the above embodiments comprising Q, Q is a substituted or unsubstituted phenyl ring. In another variation of any of the above embodiments comprising Q, Q is a substituted or unsubstituted heteroaryl. In still another variation of any of the above embodiments comprising Q, Q is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzofhiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, benthiazole, and triazine.
[0146] In one variation of any of the above embodiments and variations comprising X, at least one X in the six membered ring is a substituted carbon atom. In another variation of any of the above embodiments and variations comprising X, at least one X in the six membered ring is -CF.
[0147] In one variation of any of the above embodiments and variations comprising X, any two adjacent X moieties may optionally be CRj2 where the R12 substituents are taken together to form a ring.
[0148] In one variation of any of the above embodiments and variations comprising M, M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH- P(O)OH-CH3, sulfonamides (-SO2NH ), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group. In another variation of any of the above embodiments and variations comprising M, M comprises a hydroxamic acid. In yet another variation of any of the above embodiments and variations comprising M, M is selected from the group consisting of:
Figure imgf000058_0001
[0149] In one variation of any of the above embodiments and variations comprising QLM, QLM is
Figure imgf000059_0001
[0150] In another variation of any of the above embodiments and variations comprising Q- LM, QLM is
[0151] In one variation of any of the above embodiments and variations comprising LM, L-
M is
Figure imgf000059_0003
[0152] In another variation of any of the above embodiments and variations comprising L- M, LM is
Figure imgf000059_0004
[0153] In one variation of any of the above embodiments and variations comprising R2, R3, R4., and R5, at least one of R2, R3, R4, or R5 is fluorine.
[0154] In one variation of any of the above embodiments and variations comprising R6, R7, R8, and R9, at least one of R6, R7, R8, or R9 is fluorine. [0155] In one variation of any of the above embodiments and variations comprising Re, R ,
R8, and R9, it is noted that R6, R7, R8 or R9 may be selected such that the phenyl ring linking the five membered ring and the L group comprise one or two fluorines as indicated in the structural subunit below:
Figure imgf000060_0001
[0156] In one variation of any of the above embodiments and variations comprising R6, R , Rs, and R9, it is also noted in regard to the R6, R , R8 and R9 substituents that any two adjacent substituents may be taken together to form a ring.
[0157] In one variation of any of the above embodiments and variations comprising R10 and Rπ, it is noted that the substituted or unsubstituted aromatic ring formed when Rio and Rπ are taken together may optionally be a substituted or unsubstituted aryl or a heteroaryl. [0158] In one variation of any of the above embodiments and variations comprising V, each V is selected so that the ring is an unsubstituted or substituted piperdin-3-yl moiety. [0159] In one variation of any of the above embodiments and variations comprising R14, R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
[0160] In another variation of any of the above embodiments and variations comprising Rι4, R14 is a substituted or unsubstituted C1-6 alkyl. In still another variation of any of the above embodiments and variations comprising R14, R14 is a substituted or unsubstituted - C(O)C1-6 alkyl. In a further variation of any of the above embodiments and variations comprising R14, R14 is selected from the group consisting of hydrogen, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted. In yet another variation of any of the above embodiments and variations comprising R14, R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC. [0161] It is noted in regard to each of the above embodiments that a given alkyl, alkoxy, aryloxy, heteroaryloxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, amino, thio, or carbonyl group substituent may optionally be further substituted. As also noted, such two substituents may be taken together to form a ring. Examples of further substituted alkyl groups include, but are not limited to, those selected from the group consisting of haloalkyl, cycloalkyl, aminoalkyl, oxaalkyl, heteroaralkyl, and aralkyl, each of which may optionally be further substituted. Examples of further substituted alkoxy aryloxy, and heteroaryloxy groups include, but are not limited to, those selected from the group consisting of haloalkoxy, haloaryloxy, and haloheteroaryloxy, each of which may optionally be further substituted. Examples of further substituted aminosulfonyl, alkylsulfonyl, arylsulfonyl, and heteroarylsulfonyl groups include, but are not limited to, those selected from the group consisting of alkylaminosulfonyl, arylaminosulfonyl, heteroarylaminosulfonyl, heteroaralkylsulfonyl, and aralkylsulfonyl, each of which may optionally be further substituted. Examples of further substituted amino groups include, but are not limited to, those selected from the group consisting of alkylamino, arylarriino, and acylamino, each of which may optionally be further substituted. Examples of further substituted thio groups include, but are not limited to, tiiose selected from die group consisting of alkylthio, arylthio, and heteroarylthio, each of which may optionally be further substituted. Examples of further substituted carbonyl groups include, but are not limited to, acids, acid halides, amides, esters, and ketones. For example, the carbonyl groups may be an alkylcarbonyl, arylcarbonyl, heteroarylcarbonyl, aminocarbonyl, alkoxycarbonyl, aralkoxycarbonyl, or heteroaralkoxycarbonyl, each of which may optionally be further substituted.
[0162] It is noted that the preceding lists of examples are not intended to be limiting as other forms of alkyl, alkoxy, aryloxy, heteroaryloxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, amino, and thio groups may also be formed with the addition of other substituents to the base group, some of which are described herein and all of which are intended to fall within the scope of the present invention. Substituent Ri
[0163] Figure 2A illustrates particular examples of moieties that may be used as a Ri substituent. The below table also provides examples of different compounds having different Ri substituents.
Figure imgf000062_0001
Ri Substituents
Figure imgf000062_0002
Figure imgf000063_0001
[0164] It should be recognized that the compounds described in the above table where the Ri substituent is varied may each be further substituted by replacing one or more of the hydrogens implicitly depicted in the structure with non-hydrogen substituents. Such further substituents may optionally form additional fused rings, as is also taught herein. [0165] In one variation, Ri is a substituted alkyl where the carbon of Rj alpha to the ring atom is a tertiary carbon, i.e., in addition to the bond to the ring atom, the carbon atom has two non-hydrogen substituents. It is believed that substitution of the carbon alpha to die ring atom in this manner may reduce oxidation of that alpha carbon, particularly when the ring atom is nitrogen, thus adding to the stability of the compound. Substituentg R Ra, R., Rs, Rfe Rr. s and R»
[0166] R2, R3, R , R5, R6, R , R8 and R9 may each independently be selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted.
[0167] It is noted that R2 and R3; R3 and R4; and R4 and R5 may each optionally be taken together to form a ring. The ring formed may optionally be a 5 or 6 membered ring. In one variation, the ring formed is an aryl or heteroaryl ring.
[0168] It is also noted that R6 and R7; R and R8; and R8 and R9 may each optionally be taken together to form a ring. The ring formed may optionally be a 5 or 6 membered ring. In one variation, the ring formed is an aryl or heteroaryl ring. Substituent Z
[0169] Figure 2B illustrates particular examples of Z moieties that the compounds of the present invention may comprise. In one particular embodiment, the Z moiety is a substituted or unsubstituted benzimidazole or imidazole.
[0170] It is noted that the examples of Z moieties shown in Figure 2B may optionally be further substituted as has been specified herein. For example, the various Ri substituents that may be appended to the ring are not specified in Figure 2B.
[0171] Also, it is noted that Figure 2B is intended only to be exemplary and that other Z substituents may be employed in the compounds according to the present invention consistent with the teachings herein.
Substituent O
[0172] As noted above, Q may be a substituted or unsubstituted aromatic ring. The substituents of the aromatic ring can vary widely and may optionally be such that one or more additional rings are fused to d e core aromatic ring of Q.
[0173] Q may optionally be a 5 or 6 membered aromatic ring. When Q is a 6 membered aromatic ring, moieties Z and L may be meta or para substituents relative to each other on the
6 membered aromatic ring. In one variation, moieties Z and L are meta substituted relative to each other.
[0174] In one variation where Q is a phenyl ring, die phenyl ring may have substituents R6,
R , R8, and R9. As indicated above, these substituents may each optionally be independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted. It is noted that other substituents may additionally be appended to the phenyl ring without departing from the intended scope of the present invention.
[0175] In another variation, Q is a 5 and 6 membered aromatic ring comprising heteroatoms, i.e., a heteroaryl. For example, the heteroaryl ring may optionally have the formula
Figure imgf000065_0001
or
Figure imgf000065_0002
where a, b, c, d and e are each independently nitrogen (N) or carbon (C), with a proviso that when a and c are both nitrogen, then c is carbon. When a, b, c, d and/or e are carbon, the given carbon atom may be substituted. Examples of substituents include, but are not limited to members selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted. It is noted that other substituents may additionally be appended to the heteroaryl ring without departing from the intended scope of the present invention. [0176] Examples of rings comprising heteroatoms, including 5 and 6 membered aromatic rings comprising heteroatoms are illustrated in Figure 2C. It is noted that the rings shown in Figure 2C are unsubstituted and that further substitutions may optionally be added as has been specified.
[0177] Further particular examples of rings that may be comprised in the Q substituent include, but are not limited to furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, benthiazole, and triazine.
[0178] Surprisingly, it was determined that when group Q is a meta substituted aryl or heteroaryl group, the resulting inhibitors show improved biological activities over that of the corresponding para substituted aryl or heteroaryl groups. Preferably, the meta substituted aryl is a meta substituted phenyl moiety that is substituted or unsubstituted. Without being bound by any particular theory, it is believed that the meta substitution serves to direct the zinc complexing substituent M to a more favorable position so as to allow the zinc complexing substituent to interact with the zinc ion while the remainder of the compound maintains its interaction with hydrophobic regions in the binding pocket of the histone deacetylase.
Metal Ion Complexing Substituent. M
[0179] In regard to each of the above embodiments, substituent M may be a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion, and optionally more particularly a zinc ion since a zinc ion is known to be present in the catalytic site of deacetylases. Hence, the M substituent may facilitate inhibitor binding by complexing with the zinc ion present in the catalytic site of deacetylases. In one particular variation, M is a substituent capable of complexing with a histone deacetylase catalytic site and/or a metal ion. [0180] Examples of substituents capable of complexing with a zinc ion tiiat may be used as the M substituent include, but are not limited to trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH- CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkyloxy group. Particular examples of such substituents include:
Figure imgf000066_0001
[0181] In one particular variation, M is a hydroxamic acid (-C(O)-NHOH), also shown above. It is noted that hydroxamic acids, such as trichostatin A, have been shown to be effective inhibitors against histone deacetylases by complexing with the zinc ion present in the catalytic site of histone deacetylases.
Leader group, L
[0182] In regard to each of the above embodiments, the leader group, L, may be any substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound. The number of atoms separating the M substituent and the remainder of the compound serves to extend the zinc complexing substituent, M, a sufficient distance away from the remainder of the compound so as to allow the zinc complexing substituent to interact with the zinc ion while the remainder of the compound interacts with hydrophobic regions in the binding pocket of the deacetylase.
[0183] In one embodiment, the leader group, L, provides between 1-10 atoms that extend from the M substituent to remainder of the compound, optionally 3-9 and optionally 4-8 atoms. In one variation, the number of atoms separating the M substituent from the remainder of the compound is 3, 4, 5, 6, 7, 8 or 9 atoms.
[0184] It is noted that the atoms of the leader group extending between the M substituent and the remainder of the compound may consist only of carbon atoms. Alternatively, the atoms of the leader group extending between the M substituent and the remainder of the compound may also comprise non-carbon atoms such as nitrogen, oxygen and sulfur. [0185] It is also noted that the bonds between the atoms of the leader group extending between the M substituent and the remainder of the compound may be saturated, partially unsaturated, or fully unsaturated. For example, the leader group may comprise one or more alkene (-CH=CH-) or alkyne (-C≡C-) bonds.
[0186] A variety of different moieties may be incorporated into the leader groups of the HDAC inhibitors of die present invention. Examples of such moieties are shown in Figure 2D.
[0187] The atoms forming the backbone of the leader group, L, may optionally comprise one or more members of the group consisting of: -(CH2)n-, where n is an integer from 1 to 10; -CH(CH3)-; -CH(CH3)CH2- and -CH2CH(CH3)-; -CH(CH3)CH2CH2-, -CH2CH(CH3)CH2-, and -CH2CH2CH (CH3)-; -CH(CH3)CH2CH2CH2-, -CH2CH(CH3)CH2CH2-, - CH2CH2CH(CH3)CH2-, and -CH2CH2CH2CH(CH3)-; -CH(CH3)CH2CH2CHCH2-, - CH2CH(CH3)CH2CH2CH2-, -CH2CH2CH(CH3)CH2CH2-, -CH2CH2CH2CH(CH3)CH2-, and - CH2CH2CH2CH2CH(CH3)-; -CH(CH2CH3)-; -CH(CH2CH3)CH2- and -CH2CH(CH2CH3)-; - CH(CH2CH3)CH2CH2-, -CH2CH(CH2CH3)CH2-, and -CH2CH2CH(CH2CH3)-; - CH(CH2CH3)CH2CH2CH2-, -CH2CH(CH2CH3)CH2CH2-, -CH2CH2CH(CH2CH3)CH2-, and - CH2CH2CH2CH(CH2CH3)-; -CH2CH2CH(CH2CH3)CH2CH2-,
CH2CH2CH2CH(CH2CH3)CH2-, and -CH(CH2CH3)CH2CH2CH2CH2-,
CH2CH(CH2CH3)CH2CH2CH2-, -CH2CH2CH2CHCH(CH2CH3); -CH=CH-; -CH=CHCH2- and -CH2CH=CH-; -CH=CHCHCH2-, -CH2CH=CHCH2-, and -CH2CH2CH=CH-; - CH=CHCH2CH2CH2-, -CH2CH=CHCH2CH2-, -CH2CH2CH=CHCH2-, and
-H2CH2CH2CH=CH-; -CH=CHCHCH2CH2CH2-, -CH2CH=CHCH2CH2CH2-,
-CH2CH2CH=CHCH2CH2-, -CH2CH2CH2CH=CHCH2-, and -CH2CH2CH2CHCH=CH-; -C(CH3)=CH- and -CH=C(CH3)-; -C(CH3)=CHCH2-, -CH=C(CH3)CH2-, and - CH=CHCH(CH3)-; -CH(CH3)CH=CH-, -CH2C(CH3)=CH-, and -CH2CH=C(CH3)-; - CH=CHCH=CH-; -CH=CHCH=CHCH2-, -CH2CH=CHCH=CH-, and-CH=CHCH2CH=CH-; -CH=CHCH=CHCH2CH2-, -CH=CHCH2CH=CHCH2-, and -CH=CHCH2CH2CH=CH-, -CH2CH=CHCH=CHCH2-, -CH2CH=CHCH2CH=CH-, and -CH2CH2CH=CHCH=CH-; -C(CH3)=CHCH=CH-, -CH=C(CH3)CH =CH-, -CH=CHC(CH3)=CH-, and - CH=CHCH=C(CH3)- ; -C≡C-; -C≡CCH2-, -CH2C≡C-; -C≡CCH(CH3)-, and -CH(CH3)C≡C-; - C=CCH2CH2-, -CH2C-CCH2-, and -CH2CH2C=C-; -C≡CCH(CH3)CH2- and - C≡CCH2CH(CH3)-; -CH(CH3)C=CCH2- and -CH2C≡CCH(CH3)-; -CH(CH3)CH2C≡C- and- CH2CH(CH3)C≡C-; -C≡CCH=CH-, -CH=CHC≡C-, and -C≡CC≡C-; -C≡CCH2CH2CH2- and - CH2CH2CH2C≡C-; -C≡CCH2CH2CH2CH2- and -CH2CH2CH2CH2C≡C-;
Figure imgf000068_0001
C(CH3)=CHC≡C-, -CH=C(CH3)C≡C-, -C≡CC(CH3)=CH-, and -C≡CCH=C(CH3). L may also be E, Z or mixtures of E/Z -CH =CH2-. It is noted that the hydrogen atoms of above possible portions of the leader group may optionally be substituted with further substituents. [0188] It is also noted that the leader group may comprise one or more substituents extending from one or more atoms of the leader group backbone. In one variation, two substituents extending from the atoms extending between the carbon alpha to the leader group and the M substituent to form one or more three, four, five, six, seven, eight or nine membered rings. The atoms of the leader group forming the ring may be separated from each other by 0, 1, 2, 3, or 4 atoms.
[0189] The rings may be saturated or partially unsaturated (i.e., comprise one or two double bonds). The rings may also be aromatic, referred to herein as aryl and heteroaryl rings. The rings may optionally be further substituted. These further ring substituents may combine to form additional rings that are fused to the rings forming a portion of the backbone, e.g., bicycloaryl and bicycloheteroaryl.
[0190] Examples of cycloalkyl rings that may be formed by one or more leader group backbone atoms include, but are not limited to: cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, phenyl, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene. [0191] Examples of heteroaryl rings that may be formed by one or more leader group backbone atoms include, but are not limited to: furan, thiofuran, pyrrole, isopyrrole, 3- isopyrrole, pyrazole, isoimidazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiofuran, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, and pyridopyridine.
[0192] It is noted that the inhibitors may include one or more chiral centers. The chiral centers may be either the R or S enantiomers, depending on the substituents. [0193] Synthetic scheme for synthesizing compounds according to these various embodiments are provided in the Examples. Particular examples of HDAC inhibitors according to these embodiments are provided in the examples.
A. Salts, Hydrates^ and Prodrugs of HDAC Inhibitors
[0194] It should be recognized that d e compounds of the present invention may be present and optionally administered in the form of salts, hydrates and prodrugs that are converted in vivo into the compounds of the present invention. For example, it is within the scope of the present invention to convert the compounds of the present invention into and use them in the form of their pharmaceutically acceptable salts derived from various organic and inorganic acids and bases in accordance with procedures well known in the art.
[0195] When the compounds of the present invention possess a free base form, the compounds can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, e.g., hydrohalides such as hydrochloride, hydrobromide, hydroiodide; otiier mineral acids and their corresponding salts such as sulfate, nitrate, phosphate, etc.; and alkyl- and monoarylsulfonates such as ethanesulfonate, toluenesulfonate and benzenesulfonate; and other organic acids and their corresponding salts such as acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate and ascorbate. Further acid addition salts of the present invention include, but are not limited to: adipate, alginate, arginate, aspartate, benzenesulfonate (besylate), bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, ethanesulfonate, fumarate, galacterate (from mucic acid), galacturonate, glucoheptaoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, iodide, isethionate, iso-butyrate, lactate, lactobionate, malate, malonate, mandelate, metaphosphate, methanesulfonate, methylbenzoate, monohydrogenphosphate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, oleate, pamoate, pectinate, persulfate, phenylacetate, 3-phenylpropionate, phosphate, phosphonate and phthalate. It should be recognized that the free acid forms will typically differ from their respective salt forms somewhat in physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid forms for the purposes of the present invention.
[0196] When the compounds of the present invention possess a free base form, a pharmaceutically acceptable base addition salt can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Examples of such bases are alkali metal hydroxides including potassium, sodium and lithium hydroxides; alkaline earth metal hydroxides such as barium and calcium hydroxides; alkali metal alkoxides, e.g. potassium ethanolate and sodium propanolate; and various organic bases such as ammonium hydroxide, piperidine, diethanolamine and N-methylglutamine. Also included are the aluminum salts of the compounds of the present invention. Further base salts of the present invention include, but are not limited to: copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium and zinc salts. Organic base salts include, but are not limited to, salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, e.g., arginine, betaine, caffeine, chloroprocaine, choline, N, N'-dibenzylethylenediamine (benzad ine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropylamine and tris-(hyckoxymethyl)-methylamine (tromethamine). It should be recognized that the free base forms will typically differ from their respective salt forms somewhat in physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base forms for the purposes of the present invention. [0197] Compounds of the present invention, which comprise basic nitrogen-containing groups, may be quaternized with such agents as (C1-4) alkyl halides, e.g., methyl, ethyl, isopropyl and tert-butyl chlorides, bromides and iodides; di (C1-4) alkyl sulfates, e.g., dimethyl, diethyl and diamyl sulfates; (Cio-is) alkyl halides, e.g., decyl, dodecyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aryl (Cι-4) alkyl halides, e.g., benzyl chloride and phenethyl bromide. Such salts permit the preparation of both water-soluble and oil-soluble compounds of the present invention.
[0198] N-oxides of compounds according to the present invention can be prepared by methods known to those of ordinary skill in the art. For example, N-oxides can be prepared by treating an unoxidized form of the compound with an oxidizing agent (e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, metα-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichlorornethane) at approximately 0 °C. Alternatively, the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
[0199] Prodrug derivatives of compounds according to the present invention can be prepared by modifying substituents of compounds of the present invention tiiat are then converted in vivo to a different substituent. It is noted that in many instances, the prodrugs themselves also fall within the scope of the range of compounds according to the present invention. For example, prodrugs can be prepared by reacting a compound with a carbamylating agent (e.g., l,l-acyloxyalkylcarbonochloridate, ?αrα-nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et α/.(1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985.
[0200] Protected derivatives of compounds of the present invention can also be made. Examples of techniques applicable to the creation of protecting groups and their removal can be found in T.W. Greene, Protecting Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, Inc. 1999. [0201] Compounds of the present invention may also be conveniently prepared, or formed during the process of the invention, as solvates (e.g. hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxane, tetrahydrofuran or methanol. [0202] A "pharmaceutically acceptable salt", as used herein, is intended to encompass any compound according to the present invention that is utilized in the form of a salt thereof, especially where the salt confers on the compound improved pharmacokinetic properties as compared to the free form of compound or a different salt form of the compound. The pharmaceutically acceptable salt form may also initially confer desirable pharmacokinetic properties on the compound that it did not previously possess, and may even positively affect the pharmacodynamics of the compound with respect to its therapeutic activity in the body. An example of a pharmacokinetic property that may be favorably affected is the manner in which the compound is transported across cell membranes, which in turn may directly and positively affect the absorption, distribution, biotransformation and excretion of the compound. While the route of administration of d e pharmaceutical composition is important, and various anatomical, physiological and pathological factors can critically affect bioa ailability, the solubility of the compound is usually dependent upon the character of d e particular salt form thereof, which it utilized. One of skill in the art will appreciate that an aqueous solution of the compound will provide the most rapid absorption of the compound into the body of a subject being treated, while lipid solutions and suspensions, as well as solid dosage forms, will result in less rapid adsorption of the compound.
3. PREPARATION OF HDAC INHIBITORS
[0203] Various methods may be developed for synthesizing compounds according to the present invention. Representative methods for synthesizing these compounds are provided in the Examples. It is noted, however, that the compounds of the present invention may also be synthesized by other synthetic routes that others may devise.
[0204] It will be readily recognized that certain compounds according to the present invention have atoms with linkages to other atoms that confer a particular stereochemistry to the compound (e.g., chiral centers). It is recognized that synthesis of compounds according to the present invention may result in the creation of mixtures of different stereoisomers (enantiomers, diastereomers). Unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all of the different possible stereoisomers. [0205] Various methods for separating mixtures of different stereoisomers are known in the art. For example, a racemic mixture of a compound may be reacted with an optically active resolving agent to form a pair of diastereoisomeric compounds. The diastereomers may then be separated in order to recover the optically pure enantiomers. Dissociable complexes may also be used to resolve enantiomers (e.g., crystalline diastereoisomeric salts). Diastereomers typically have sufficiently distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) that they can be readily separated by taking advantage of these dissimilarities. For example, diastereomers can typically be separated by chromatography or by separation/resolution techniques based upon differences in solubility. A more detailed description of techniques that can be used to resolve stereoisomers of compounds from their racemic mixture can be found in Jean Jacques Andre Collet, Samuel H. Wilen, Enantiomers, Racemates and Resolutions, John Wiley & Sons, Inc. (1981).
4. INDICATIONS FOR USE OF HDAC INHIBITORS
[0206] HDAC is believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of HDAC in a subject through inhibition may be used to therapeutically address these disease states. Examples of various diseases that may be treated using the HDAC inhibitors of the present invention are described herein. It is noted that additional diseases beyond those disclosed herein may be later identified as the biological roles that HDAC play in various pathways becomes more fully understood.
A. Undesirable or Uncontrolled Cell Proliferation [0207] One set of indications that HDAC inhibitors of the present invention may be used to treat are those involving undesirable or uncontrolled cell proliferation. Such indications include benign tumors, various types of cancers such as primary tumors and tumor metastasis, restenosis (e.g. coronary, carotid, and cerebral lesions), abnormal stimulation of endothelial cells (atherosclerosis), insults to body tissue due to surgery, abnormal wound healing, abnormal angiogenesis, diseases that produce fibrosis of tissue, repetitive motion disorders, disorders of tissues that are not highly vascularized, and proliferative responses associated with organ transplants. More specific indications for HDAC inhibitors include, but are not limited to prostate cancer, lung cancer, acute leukemia, multiple myeloma, bladder carcinoma, renal carcinoma, breast carcinoma, colorectal carcinoma, neuroblastoma and melanoma. [0208] In one embodiment, a method is provided for treating diseases associated with undesired and uncontrolled cell proliferation. The method comprises administering to a subject suffering from uncontrolled cell proliferation a therapeutically effective amount of a HDAC inhibitor according to the present invention, such that said uncontrolled cell proliferation is reduced. The particular dosage of the inhibitor to be used will depend on the severity of the disease state, the route of administration, and related factors that can be deteπnined by the attending physician. Generally, acceptable and effective daily doses are amounts sufficient to effectively slow or eliminate uncontrolled cell proliferation. [0209] HDAC inhibitors according to the present invention may also be used in conjunction with other agents to inhibit undesirable and uncontrolled cell proliferation. Examples of other anti-cell proliferation agents that may be used in conjunction with the HDAC inhibitors of the present invention include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATL ™ protein, ENDOSTATIN™ protein, suramin, squalamine, tissue inhibitor of metalloproteinase-I, tissue inhibitor of metalloproteinase-2, plasminogen activator inhibitor- 1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulfate (clupeine), sulfated chitin derivatives (prepared from queen crab shells), sulfated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA), cishydroxyproline, d,l-3,4- dehydroproline, thiaproline), beta.-aminopropionitrile fumarate, 4-propyl-5-(4-pyridinyl)- 2(3H)-oxazolone; methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta.-cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d-penicillamine (CDPT), beta.-1-anticollagenase-serum, alpha.2- antiplasmin, bisantrene, lobenzarit disodium, n-(2-carboxyphenyl-4-chloroanthronilic acid disodium or "CCA", thalidomide; angostatic steroid, carboxyaminoimidazole; metalloproteinase inhibitors such as BB94. Other anti-angiogenesis agents that may be used include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: bFGF, aFGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-l/Ang-2. Ferrara N. and Alitalo, K. "Clinical application of angiogenic growth factors and their inhibitors" (1999) Nature Medicine 5:1359-1364.
[0210] Generally, cells in benign tumors retain their differentiated features and do not divide in a completely uncontrolled manner. A benign tumor is usually localized and nonmetastatic. Specific types of benign tumors that can be treated using HDAC inhibitors of the present invention include hemangiomas, hepatocellular adenoma, cavernous haemangioma, focal nodular hyperplasia, acoustic neuromas, neurofibroma, bile duct adenoma, bile duct cystanoma, fibroma, lipomas, leiomyomas, mesotheliomas, teratomas, myxomas, nodular regenerative hyperplasia, trachomas and pyogenic granulomas. [0211] In the case of malignant tumors, cells become undifferentiated, do not respond to the body' s growth control signals, and multiply in an uncontrolled manner. Malignant tumors are invasive and capable of spreading to distant sites (metastasizing). Malignant tumors are generally divided into two categories: primary and secondary. Primary tumors arise directly from the tissue in which they are found. Secondary tumors, or metastases, are tumors that originated elsewhere in the body but have now spread to distant organs. Common routes for metastasis are direct growth into adjacent structures, spread through the vascular or lymphatic systems, and tracking along tissue planes and body spaces (peritoneal fluid, cerebrospinal fluid, etc.).
[0212] Specific types of cancers or malignant tumors, either primary or secondary, that can be treated using the HDAC inhibitors of the present invention include, but are not limited to, leukemia, breast cancer, skin cancer, bone cancer, prostate cancer, liver cancer, lung cancer, brain cancer, cancer of the larynx, gallbladder, pancreas, rectum, parathyroid, thyroid, adrenal, neural tissue, head and neck, colon, stomach, bronchi, kidneys, basal cell carcinoma, squamous cell carcinoma of both ulcerating and papillary type, metastatic skin carcinoma, osteo sarcoma, Ewing's sarcoma, veticulum cell sarcoma, myeloma, giant cell tumor, small- cell lung tumor, gallstones, islet cell tumor, primary brain tumor, acute and chronic lymphocytic and granulocytic tumors, hairy-cell tumor, adenoma, hyperplasia, medullary carcinoma, pheochromocytoma, mucosal neuronms, intestinal ganglloneuromas, hyperplastic corneal nerve tumor, marfanoid habitus tumor, Wilm's tumor, seminoma, ovarian tumor, leiomyomater tumor, cervical dysplasia and in situ carcinoma, neuroblastoma, retinoblastoma, soft tissue sarcoma, malignant carcinoid, topical skin lesion, mycosis fungoide, rhabdomyosarcoma, Kaposi's sarcoma, osteogenic and other sarcoma, malignant hypercalcemia, renal cell tumor, polycythermia vera, adenocarcinoma, glioblastoma multiforma, leukemias, lymphomas, malignant melanomas, epidermoid carcinomas, and other carcinomas and sarcomas.
[0213] The HDAC inhibitors of the present invention may also be used to treat abnormal cell proliferation due to insults to body tissue during surgery. These insults may arise as a result of a variety of surgical procedures such as joint surgery, bowel surgery, and cheloid scarring. Diseases that produce fibrotic tissue include emphysema. Repetitive motion disorders that may be treated using the present invention include carpal tunnel syndrome. An example of a cell proliferative disorder that may be treated using the invention is a bone tumor.
[0214] Proliferative responses associated with organ transplantation that may be treated using HDAC inhibitors of the invention include proliferative responses contributing to potential organ rejections or associated complications. Specifically, these proliferative responses may occur during transplantation of the heart, lung, liver, kidney, and otiier body organs or organ systems.
[0215] Abnormal angiogenesis that may be may be treated using this invention include those abnormal angiogenesis accompanying rheumatoid arthritis, ischemic-reperfusion related brain edema and injury, cortical ischemia, ovarian hyperplasia and hypervascularity, (polycystic ovary syndrome), endometriosis, psoriasis, diabetic retinopathy, and other ocular angiogenic diseases such as retinopathy of prematurity (retrolental fibroplastic), macular degeneration, corneal graft rejection, neuroscular glaucoma and Oster Webber syndrome. [0216] Examples of diseases associated with uncontrolled angiogenesis that may be treated according to the present invention include, but are not limited to retinal/choroidal neovascularization and corneal neovascularization. Examples of retinal/choroidal neovascularization include, but are not limited to, Bests diseases, myopia, optic pits, Stargarts diseases, Pagets disease, vein occlusion, artery occlusion, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum carotid apo structive diseases, chronic uveitis/vitritis, mycobacterial infections, Lyme's disease, systemic lupus erythematosus, retinopathy of prematurity, Eales disease, diabetic retinopathy, macular degeneration, Bechets diseases, infections causing a retinitis or chroiditis, presumed ocular histoplasmosis, pars planitis, chronic retinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma and post-laser complications, diseases associated with rubesis (neovascularization of the angle) and diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy. Examples of corneal neovascularization include, but are not limited to, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, sjogrens, acne rosacea, phylectenulosis, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, Mooren ulcer, Terrien's marginal degeneration, marginal keratolysis, polyarteritis, Wegener sarcoidosis, Scleritis, periphigoid radial keratotomy, neovascular glaucoma and retrolental fibroplasia, syphilis, Mycobacteria infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infections, Herpes zoster infections, protozoan infections and Kaposi sarcoma.
[0217] Chronic inflammatory diseases associated with uncontrolled angiogenesis may also be treated using HDAC inhibitors of the present invention. Chronic inflammation depends on continuous formation of capillary sprouts to maintain an influx of inflammatory cells. The influx and presence of the inflammatory cells produce granulomas and thus maintains the chronic inflammatory state. Inhibition of angiogenesis using a HDAC inhibitor alone or in conjunction with other anti-inflammatory agents may prevent the formation of the granulosmas and thus alleviate the disease. Examples of chronic inflammatory diseases include, but are not limited to, inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, psoriasis, sarcoidosis, and rheumatoid arthritis.
[0218] Inflammatory bowel diseases such as Crohn's disease and ulcerative colitis are characterized by chronic inflammation and angiogenesis at various sites in die gastrointestinal tract. For example, Crohn's disease occurs as a chronic transmural inflammatory disease that most commonly affects the distal ileum and colon but may also occur in any part of the gastrointestinal tract from the mouth to the anus and perianal area. Patients with Crohn's disease generally have chronic diarrhea associated with abdominal pain, fever, anorexia, weight loss and abdominal swelling. Ulcerative colitis is also a chronic, nonspecific, inflammatory and ulcerative disease arising in the colonic mucosa and is characterized by the presence of bloody diarrhea. These inflammatory bowel diseases are generally caused by chronic granulomatous inflammation throughout the gastrointestinal tract, involving new capillary sprouts surrounded by a cylinder of inflammatory cells. Inhibition of angiogenesis by these inhibitors should inhibit the formation of the sprouts and prevent the formation of granulomas. Inflammatory bowel diseases also exhibit extra intestinal manifectations, such as skin lesions. Such lesions are characterized by inflammation and angiogenesis and can occur at many sites other the gastrointestinal tract. Inhibition of angiogenesis by HDAC inhibitors according to the present invention can reduce the influx of inflammatory cells and prevent lesion formation.
[0219] Sarcoidosis, another chronic inflammatory disease, is characterized as a multisystem granulomatous disorder. The granulomas of this disease can form anywhere in the body. Thus, the symptoms depend on the site of the granulomas and whether the disease is active. The granulomas are created by the angiogenic capillary sprouts providing a constant supply of inflammatory cells. By using HDAC inhibitors according to the present invention to inhibit angionesis, such granulomas formation can be inhibited. Psoriasis, also a chronic and recurrent inflammatory disease, is characterized by papules and plaques of various sizes. Treatment using these inhibitors alone or in conjunction with other anti-inflammatory agents should prevent the formation of new blood vessels necessary to maintain the characteristic lesions and provide the patient relief from the symptoms.
[0220] Rheumatoid arthritis (RA) is also a chronic inflammatory disease characterized by non-specific inflammation of the peripheral joints. It is believed that the blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, the endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. The factors involved in angiogenesis may actively contribute to, and help maintain, the chronically inflamed state of rheumatoid arthritis. Treatment using HDAC inhibitors according to the present invention alone or in conjunction with other anti-RA agents may prevent the formation of new blood vessels necessary to maintain the chronic inflammation and provide the RA patient relief from the symptoms.
5. COMPOSITIONS COMPRISING HDAC INHIBITORS
[0221] A wide variety of compositions and administration methods may be used in conjunction with the HDAC inhibitors of the present invention. Such compositions may include, in addition to the HDAC inhibitors of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents. Additionally, the compositions may include active agents in addition to the HDAC inhibitors of the present invention. These additional active agents may include additional compounds according to the invention, or one or more other pharmaceutically active agents. [0222] The compositions may be in gaseous, liquid, semi-liquid or solid form, formulated in a manner suitable for the route of administration to be used. For oral administration, capsules and tablets are typically used. For parenteral administration, reconstitution of a lyophilized powder, prepared as described herein, is typically used. [0223] Compositions comprising HDAC inhibitors of the present invention may be administered or coadministered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermaily, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally. The compounds and/or compositions according to the invention may also be administered or coadministered in slow release dosage forms.
[0224] The HDAC inhibitors and compositions comprising them may be administered or coadministered in any conventional dosage form. Coadministration in the context of tiiis invention is intended to mean the administration of more than one therapeutic agents, one of which includes a HDAC inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome. Such coadministration may also be coextensive, that is, occurring during overlapping periods of time.
[0225] Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application may optionally include one or more of the following components: a sterile diluent, such as 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; agents for the adjustment of tonicity such as sodium chloride or dextrose, and agents for adjusting the acidity or alkalinity of the composition, such as alkaline or acidifying agents or buffers like carbonates, bicarbonates, phosphates, hydrochloric acid, and organic acids like acetic and citric acid. Parenteral preparations may optionally be enclosed in ampules, disposable syringes or single or multiple dose vials made of glass, plastic or other suitable material.
[0226] When HDAC inhibitors according to the present invention exhibit insufficient solubility, methods for solubilizing the compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEN, or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.
[0227] Upon mixing or adding HDAC inhibitors according to the present invention to a composition, a solution, suspension, emulsion or the like may be formed. The form of the resulting composition will depend upon a number of factors, including the intended mode of administration, and the solubility of the compound in the selected carrier or vehicle. The effective concentration needed to ameliorate the disease being treated may be empirically determined.
[0228] Compositions according to the present invention are optionally provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, dry powders for inhalers, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds, particularly the pharmaceutically acceptable salts, preferably the sodium salts, thereof. The pharmaceutically tiierapeuticaliy active compounds and derivatives ti ereof are typically formulated and administered in unit-dosage forms or multiple-dosage forms. Unit- dose forms, as used herein, refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes individually packaged tablet or capsule. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pint or gallons. Hence, multiple dose form is a multiple of unit-doses that are not segregated in packaging. [0229] In addition to one or more HDAC inhibitors according to the present invention, the composition may comprise: 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 acaciagelatin, glucose, molasses, polvinylpyrrolidine, 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 compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents. Actual methods of preparing such dosage forms are known in the art, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, A. Gennaro, ed., 20th edition, Lippincott, Williams & Wilkins, Philadelphia, PA, 2000. The composition or formulation to be administered will, in any event, contain a sufficient quantity of a HDAC inhibitor of the present invention to reduce HDAC activity in vivo, thereby treating the disease state of the subject.
[0230] Dosage forms or compositions may optionally comprise one or more HDAC inhibitors according to the present invention in the range of 0.005% to 100% (weight/weight) with the balance comprising additional substances such as those described herein. For oral administration, a pharmaceutically acceptable composition may optionally comprise any one or more commonly employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate, sodium saccharin, talcum. Such compositions include solutions, suspensions, tablets, capsules, powders, dry powders for inhalers and sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, and biodegradable, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others. Methods for preparing these formulations are known to those skilled in the art. The compositions may optionally contain 0.01%-100% (weight/weight) of one or more HDAC inhibitors, optionally 0.1-95%, and optionally 1-95%.
[0231] Salts, preferably sodium salts, of the HDAC inhibitors may be prepared with carriers that protect the compound against rapid elimination from the body, such as time release formulations or coatings. The formulations may further include other active compounds to obtain desired combinations of properties.
A. Formulations For Oral Administration
[0232] Oral pharmaceutical dosage forms may be as a solid, gel or liquid. Examples of solid dosage forms include, but are not limited to tablets, capsules, granules, and bulk powders. More specific examples of oral tablets include compressed, chewable lozenges and tablets that may be enteric-coated, sugar-coated or film-coated. Examples of capsules include hard or soft gelatin capsules. Granules and powders may be provided in non-effervescent or effervescent forms. Each may be combined with other ingredients known to those skilled in the art.
[0233] In certain embodiments, HDAC inhibitors according to the present invention are provided as solid dosage forms, preferably capsules or tablets. The tablets, pills, capsules, troches and the like may optionally contain one or more of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.
[0234] Examples of binders that may be used include, but are not limited to, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste.
[0235] Examples of lubricants that may be used include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
[0236] Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
[0237] Examples of glidants that may be used include, but are not limited to, colloidal silicon dioxide. [0238] Examples of disintegrating agents that may be used include, but are not limited to, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
[0239] Examples of coloring agents that may be used include, but are not limited to, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate.
[0240] Examples of sweetening agents that may be used include, but are not limited to, sucrose, lactose, mannitol and artificial sweetening agents such as sodium cyclamate and saccharin, and any number of spray-dried flavors.
[0241] Examples of flavoring agents that may be used include, but are not limited to, natural flavors extracted from plants such as fruits and synthetic blends of compounds that produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.
[0242] Examples of wetting agents that may be used include, but are not limited to, propylene glycol monostβarate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylenβ lauryl ether.
[0243] Examples of anti-emetic coatings that may be used include, but are not limited to, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
[0244] Examples of film coatings that may be used include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyetiiylene glycol 4000 and cellulose acetate phthalate.
[0245] If oral administration is desired, the salt of d e compound may optionally 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 compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.
[0246] When the dosage unit form is a capsule, it may optionally additionally comprise a liquid carrier such as a fatty oil. In addition, dosage unit forms may optionally additionally comprise various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
[0247] Compounds according to the present invention may also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may optionally comprise, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
[0248] The HDAC inhibitors of the present invention may also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics. For example, if a compound is used for treating asthma or hypertension, it may be used with other bronchodilators and antihypertensive agents, respectively.
[0249] Examples of pharmaceutically acceptable carriers that may be included in tablets comprising HDAC inhibitors of the present invention include, but are not limited to 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 may be compressed tablets to which different layers of pharmaceutically acceptable substances are applied. Film-coated tablets may be compressed tablets that have been coated with polymers or other suitable coating. Multiple compressed tablets may be compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned. Coloring agents may also be used in tablets. Flavoring and sweetening agents may be used in tablets, and are especially useful in the formation of chewable tablets and lozenges.
[0250] Examples of liquid oral dosage forms tiiat may be used include, but are not limited to, aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
[0251] Examples of aqueous solutions that may be used include, but are not limited to, elixirs and syrups. As used herein, elixirs refer to clear, sweetened, hydroalcoholic preparations. Examples of pharmaceutically acceptable carriers that may be used in elixirs include, but are not limited to solvents. Particular examples of solvents that may be used include glycerin, sorbitol, ethyl alcohol and syrup. As used herein, syrups refer to concentrated aqueous solutions of a sugar, for example, sucrose. Syrups may optionally further comprise a preservative. [0252] Emulsions refer to two-phase systems in which one liquid is dispersed in the form of small globules throughout another liquid. Emulsions may optionally be oil-in-water or water-in-oil emulsions. Examples of pharmaceutically acceptable carriers that may be used in emulsions include, but are not limited to non-aqueous liquids, emulsifying agents and preservatives.
[0253] Examples of pharmaceutically acceptable substances that may be used in non- effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents.
[0254] Examples of pharmaceutically acceptable substances that may be used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic adds and a source of carbon dioxide.
[0255] Coloring and flavoring agents may optionally be used in all of the above dosage forms.
[0256] Particular examples of preservatives that may be used include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
[0257] Particular examples of non-aqueous liquids that may be used in emulsions include mineral oil and cottonseed oil.
[0258] Particular examples of emulsifying agents that may be used include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate. [0259] Particular examples of suspending agents that may be used include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia. Diluents include lactose and sucrose. Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as sodium cyclamate and saccharin.
[0260] Particular examples of wetting agents that may be used include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
[0261] Particular examples of organic acids that may be used include citric and tartaric acid.
[0262] Sources of carbon dioxide that may be used in effervescent compositions include sodium bicarbonate and sodium carbonate. Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof. [0263] Particular examples of flavoring agents that may be used include natural flavors extracted from plants such fruits, and synthetic blends of compounds that produce a pleasant taste sensation.
[0264] For a solid dosage form, the solution or suspension, in for example propylene carbonate, vegetable oils or triglycerides, is preferably encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. For a liquid dosage form, the solution, egg., for example, 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.
[0265] Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound 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 U.S. Pat. Nos. Re 28,819 and 4,358,603.
B. InjectableSj Solutions and Emulsions [0266] The present invention is also directed to compositions designed to administer the HDAC inhibitors of the present invention by parenteral administration, generally characterized by injection, either subcutaneously, intramuscularly or intravenously. Injectables may be prepared in any conventional form, for example as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. [0267] Examples of excipients that may be used in conjunction with inj ectables according to the present invention include, but are not limited to water, saline, dextrose, glycerol or ethanol. The injectable compositions may also optionally comprise 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. Implantation of a slow- release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Pat. No. 3,710,795) is also contemplated herein. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. [0268] Parenteral administration of the formulations includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as the lyophilized powders described herein, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions. The solutions may be either aqueous or nonaqueous.
[0269] When administered intravenously, examples of suitable carriers include, but are not limited to physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
[0270] Example of pharmaceutically acceptable carriers that may optionally be used in parenteral preparations include, but are not limited to aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.
[0271] Examples of aqueous vehicles tiiat may optionally be used include Sodium Chloride
Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and
Lactated Ringers Injection.
[0272] Examples of nonaqueous parenteral vehicles that may optionally be used include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
[0273] Antimicrobial agents in bacteriostatic or fungistatic concentrations may be added to parenteral preparations, particularly when the preparations are packaged in multiple-dose containers and thus designed to be stored and multiple aliquots to be removed. Examples of antimicrobial agents that may used include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
[0274] Examples of isotonic agents that may be used include sodium chloride and dextrose. Examples of buffers that may be used include phosphate and citrate. Examples of antioxidants that may be used include sodium bisulfate. Examples of local anesthetics that may be used include procaine hydrochloride. Examples of suspending and dispersing agents that may be used include sodium carboxymethylcellulose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Examples of emulsifying agents that may be used include Polysorbate 80 (Tween 80). A sequestering or chelating agent of metal ions include EDTA. [0275] Pharmaceutical carriers may also optionally include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
[0276] The concentration of a HDAC inhibitor in the parenteral formulation may be adjusted so that an injection administers a pharmaceutically effective amount sufficient to produce the desired pharmacological effect. The exact concentration of a HDAC inhibitor and/or dosage to be used will ultimately depend on the age, weight and condition of the patient or animal as is known in the art.
[0277] Unit-dose parenteral preparations may be packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration should be sterile, as is know and practiced in the art.
[0278] Injectables may be designed for local and systemic administration. Typically a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1 % w/w up to about 90% w/w or more, preferably more than 1% w/w of the HDAC inhibitor to the treated tissue(s). The HDAC inhibitor may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment will be a function of d e location of where the composition is parenterally administered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens may need to be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. Hence, the concentration ranges set forth herein are intended to be exemplary and are not intended to limit the scope or practice of the claimed formulations. [0279] The HDAC inhibitor may optionally be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease state and may be empirically determined.
C. Lyophilized Powders
[0280] The HDAC inhibitors of the present invention may also be prepared as lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. The lyophilized powders may also be formulated as solids or gels. [0281] Sterile, lyophilized powder may be prepared by dissolving the sodium salt in a sodium phosphate buffer solution containing dextrose or other suitable excipient. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. Briefly, the lyophilized powder may optionally be prepared by dissolving dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, about 1-20%, preferably about 5 to 15%, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in die art at, typically, about neutral pH. Then, a HDAC inhibitor is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35 °C, and stirred until it dissolves. The resulting mixture is diluted by adding more buffer to a desired concentration. The resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization. Each vial may contain a single dosage or multiple dosages of the HDAC inhibitor.
D. Topical Administration
[0282] The HDAC inhibitors of the present invention may also be administered as topical mixtures. Topical mixtures may be used for local and systemic administration. The resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
[0283] The HDAC inhibitors may be formulated as aerosols for topical application, such as by inhalation (see, U.S. Pat. Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract 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 typically diameters of less than 50 microns, preferably less than 10 microns. [0284] The HDAC inhibitors may also be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the HDAC inhibitor alone or in combination with other pharmaceutically acceptable excipients can also be administered.
E. Formulations For Other Routes of Administration
[0285] Depending upon the disease state being treated, otiier routes of administration, such as topical application, transdermal patches, a rectal administration, may also be used. For example, pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum that melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point. Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax, (polyoxyetiiylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm. Tablets and capsules for rectal administration may be manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.
F. Examples of Formulations
[0286] The following are particular examples of oral, intravenous and tablet formulations that may optionally be used with compounds of the present invention. It is noted that these formulations may be varied depending on the particular compound being used and the indication for which the formulation is going to be used. ORALFORMULATION
Compound of the Present Invention 10-100 mg
Citric Acid Monohydrate 105 mg
Sodium Hydroxide 18 mg
Flavoring
Water q.s. to 100 mL
INTRAVENOUS FORMULATION
Compound of the Present Invention 0.1-10 mg
Dextrose Monohydrate q.s. to make isotonic
Citric Acid Monohydrate 1.05 mg
Sodium Hydroxide 0.18 mg
Water for Injection q.s. to 1.0 mL
TABLETFORMULATION
Compound of the Present Invention 1 %
Microcrystalline Cellulose 73%
Stearic Acid 25%
Colloidal Silica 1%.
6. KITS COMPRISING HDAC INHIBITORS
[0287] The invention is also directed to kits and other articles of manufacture for treating diseases associated with HDAC. It is noted that diseases are intended to cover all conditions for which the HDAC possesses activity that contributes to the pathology and/or symptomology of the condition.
[0288] In one embodiment, a kit is provided that comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with instructions. The instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also comprise packaging materials. The packaging material may comprise a container for housing the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.
[0289] In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one HDAC inhibitor of the present invention in combination with packaging materials. The packaging material may comprise a container for housing the composition. The container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms. [0290] It is noted that the packaging material used in kits and articles of manufacture according to the present invention may form a plurality of divided containers such as a divided bottle or a divided foil packet. The container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule. The container that is employed will depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle that is in turn contained within a box. Typically the kit includes directions for the administration of the separate components. The Idt form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, topical, transdermal and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
[0291] One particular example of a kit according to the present invention is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
[0292] Another specific embodiment of a kit is a dispenser designed to dispense the daily doses one at a time in the order of their intended use. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter that indicates the number of daily doses that has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
7. COMBINATION THERAPY
[0293] A wide variety therapeutic agents may have a therapeutic additive or synergistic effect with HDAC inhibitors according to the present invention. Such therapeutic agents may additively or synergistically combine with the HDAC inhibitors to inhibit undesirable cell growth, such as inappropriate cell growti resulting in undesirable benign conditions or tumor growth.
[0294] In one embodiment, a method is provided for treating a cell proliferative disease state comprising treating cells with a compound according to the present invention in combination with an anti-proliferative agent, wherein the cells are treated witi the compound according to the present invention before, at the same time, and/or after the cells are treated with the anti-proliferative agent, referred to herein as combination therapy. It is noted that treatment of one agent before another is referred to herein as sequential therapy, even if the agents are also administered together. It is noted that combination therapy is intended to cover when agents are administered before or after each other (sequential therapy) as well as when the agents are administered at the same time.
[0295] Examples of therapeutic agents that may be used in combination with HDAC inhibitors include, but are not limited to, antieancer agents, alkylating agents, antibiotic agents, antimetabolic agents, hormonal agents, plant-derived agents, and biologic agents. [0296] Alkylating agents are polyfunctional compounds that have the ability to substitute alkyl groups for hydrogen ions. Examples of alkylating agents include, but are not limited to, bischloroethylamines (nitrogen mustards, e.g. chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, uracil mustard), aziridines (e.g. thiotepa), alkyl alkone sulfonates (e.g. busulfan), nitrosoureas (e.g. carmustine, lomustine, streptozocin), nonclassic alkylating agents (altretamine, dacarbazine, and procarbazine), platinum compounds (carboplastin and cisplatin). These compounds react with phosphate, amino, hydroxyl, sulfihydryl, carboxyl, and imidazole groups. Under physiological conditions, these drugs ionize and produce positively charged ion that attach to susceptible nucleic acids and proteins, leading to cell cycle arrest and/or cell death. Combination therapy including a HDAC inhibitor and an alkylating agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
[0297] Antibiotic agents are a group of drugs that produced in a manner similar to antibiotics as a modification of natural products. Examples of antibiotic agents include, but are not limited to, anthracyclines (e.g. doxorubicin, daunorubicin, epirabicin, idarubicin and anthraeenedione), mitomycin C, bleomycin, dactinomycin, plicatomycin. These antibiotic agents interfere with cell growth by targeting different cellular components. For example, anthracyclines are generally believed to interfere with the action of DNA topoisomerase II in the regions of transcriptionally active DNA, which leads to DNA strand scissions. Bleomycin is generally believed to chelate iron and forms an activated complex, which then binds to bases of DNA, causing strand scissions and cell death. Combination therapy including a HDAC inhibitor and an antibiotic agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotiierapeutic agents. [0298] Antimetabolic agents are a group of drugs that interfere with metabolic processes vital to the physiology and proliferation of cancer cells. Actively proliferating cancer cells require continuous synthesis of large quantities of nucleic acids, proteins, lipids, and other vital cellular constituents. Many of the antimetabolites inhibit the synthesis of purine or pyrimidine nucleosides or inhibit the enzymes of DNA replication. Some antimetabolites also interfere with the synthesis of ribonucleosides and RNA and/or amino acid metabolism and protein synthesis as well. By interfering with the synthesis of vital cellular constituents, antimetabolites can delay or arrest the growth of cancer cells. Examples of antimetabolic agents include, but are not limited to, fluorouracil (5-FU), floxuridine (5-FUdR), methotrexate, leucovorin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6-MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, and gemcitabine. Combination therapy including a HDAC inhibitor and a antimetabolic agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
[0299] Hormonal agents are a group of drug that regulate the growth and development of their target organs. Most of the hormonal agents are sex steroids and their derivatives and analogs thereof, such as estrogens, androgens, and progestins. These hormonal agents may serve as antagonists of receptors for the sex steroids to down regulate receptor expression and transcription of vital genes. Examples of such hormonal agents are synthetic estrogens (e.g. diethylstibestrol), antiestrogens (e.g. tamoxifen, toremifene, fluoxymesterol and raloxifene), antiandrogens (bicalutamide, nilutamide, flutamide), aromatase inhibitors (e.g., aminoglutethimide, anastrozole and tetrazole), ketoconazole, goserelin acetate, leuprolide, megestrol acetate and mifepristone. Combination tiierapy including a HDAC inhibitor and a hormonal agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with tiiese chemotherapeutic agents.
[0300] Plant-derived agents are a group of drugs that are derived from plants or modified based on the molecular structure of the agents. Examples of plant-derived agents include, but are not limited to, vinca alkaloids (e.g., vincristine, vinblastine, vindesine, vinzolidine and vinorelbine), podophyllotoxins (e.g., etoposide (VP-16) and teniposide (VM-26)), taxanes (e.g., paclitaxel and docetaxel). These plant-derived agents generally act as antimitotic agents that bind to tubulin and inhibit mitosis. Podophyllotoxins such as etoposide are believed to interfere with DNA synthesis by interacting with topoisomerase π, leading to DNA strand scission. Combination therapy including a HDAC inhibitor and a plant-derived agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these chemotherapeutic agents.
[0301] Biologic agents are a group of biomolecules that elicit cancer/tumor regression when used alone or in combination with chemotherapy and/or radiotherapy. Examples of biologic agents include, but are not limited to, immuno-modulating proteins such as cytokines, monoclonal antibodies against tumor antigens, tumor suppressor genes, and cancer vaccines. Combination therapy including a HDAC inhibitor and a biologic agent may have therapeutic synergistic effects on cancer, enhance the patient's immune responses to tumorigenic signals, and reduce potential sides affects associated with this chemotherapeutic agent. [0302] Cytokines possess profound immunomodulatory activity. Some cytokines such as interleukin-2 (IL-2, aldesleukin) and interferon have demonstrated antitumor activity and have been approved for the treatment of patients with metastatic renal cell carcinoma and metastatic malignant melanoma. IL-2 is a T-cell growth factor that is central to T-cell-mediated immune responses. The selective antitumor effects of IL-2 on some patients are believed to be the result of a cell-mediated immune response that discriminate between self and nonself. Examples of interleukins that may be used in conjunction with HDAC inhibitor include, but are not limited to, interleukin 2 (IL-2), and interleukin 4 (TL-4), interleukin 12 (IL-12). [0303] Interferon include more than 23 related subtypes with overlapping activities, all of the IFN subtypes within the scope of the present invention. IFN has demonstrated activity against many solid and hematologic malignancies, the later appearing to be particularly sensitive.
[0304] Other cytokines that may be used in conjunction with a HDAC inhibitor include those cytokines that exert profound effects on hematopoiesis and immune functions. Examples of such cytokines include, but are not limited to erythropoietin, granulocyte-CSF (filgrastin), and granulocyte, macrophage-CSF (sargramostim). These cytokines may be used in conjunction with a HDAC inhibitor to reduce chemotherapy-induced myelopoietic toxicity. [0305] Other immuno-modulating agents other than cytokines may also be used in conjunction with a HDAC inhibitor to inhibit abnormal cell growth. Examples of such immuno-modulating agents include, but are not limited to bacillus Calmette-Guerin, levamisole, and octreotide, a long-acting octapeptide that mimics the effects of the naturally occurring hormone somatostatin.
[0306] Monoclonal antibodies against tumor antigens are antibodies elicited against antigens expressed by tumors, preferably tumor-specific antigens. For example, monoclonal antibody HERCEPTIN® (Trastruzumab) is raised against human epidermal growth factor receptor2 (HER2) that is overexpressed in some breast tumors including metastatic breast cancer. Overexpression of HER2 protein is associated with more aggressive disease and poorer prognosis in the clinic. HERCEPTIN® is used as a single agent for the treatment of patients with metastatic breast cancer whose tumors over express the HER2 protein. Combination therapy including HDAC inhibitor and HERCEPTIN® may have therapeutic synergistic effects on tumors, especially on metastatic cancers.
[0307] Another example of monoclonal antibodies against tumor antigens is RITUXAN® (Rituximab) that is raised against CD20 on lymphoma cells and selectively deplete normal and malignant CD20+ pre-B and mature B cells. RITUXAN® is used as single agent for the treatment of patients with relapsed or refractory low-grade or follicular, CD20+, B cell non- Hodgkin' s lymphoma. Combination therapy including HDAC inhibitor and RITUXAN® may have therapeutic synergistic effects not only on lymphoma, but also on other forms or types of malignant tumors.
[0308] Tumor suppressor genes are genes that function to inhibit the cell growth and division cycles, thus preventing the development of neoplasia. Mutations in tumor suppressor genes cause the cell to ignore one or more of the components of the network of inhibitory signals, overcoming the cell cycle check points and resulting in a higher rate of controlled cell growth — cancer. Examples of the tumor suppressor genes include, but are not limited to, DPC-4, NF-1, NF-2, RB, p53, WT1, BRCA1 and BRCA2.
[0309] DPC-4 is involved in pancreatic cancer and participates in a cytoplasmic pathway that inhibits cell division. NF-1 codes for a protein that inhibits Ras, a cytoplasmic inhibitory protein. NF-1 is involved in neurofibroma and pheochromocytomas of the nervous system and myeloid leukemia. NF-2 encodes a nuclear protein that is involved in meningioma, schwanoma, and ependymoma of the nervous system. RB codes for the pRB protein, a nuclear protein that is a major inhibitor of cell cycle. RB is involved in retinoblastoma as well as bone, bladder, small cell lung and breast cancer. P53 codes for p53 protein that regulates cell division and can induce apoptosis. Mutation and/or inaction of p53 is found in a wide ranges of cancers. WT1 is involved in Wilm's tumor of the kidneys. BRCA1 is involved in breast and ovarian cancer, and BRCA2 is involved in breast cancer. The tumor suppressor gene can be transferred into the tumor cells where it exerts its tumor suppressing functions. Combination therapy including a HDAC inhibitor and a tumor suppressor may have therapeutic synergistic effects on patients suffering from various forms of cancers. [0310] Cancer vaccines are a group of agents that induce the body's specific immune response to tumors. Most of cancer vaccines under research and development and clinical trials are tumor-associated antigens (TAAs). TAA are structures (i.e. proteins, enzymes or carbohydrates) which are present on tumor cells and relatively absent or diminished on normal cells. By virtue of being fairly unique to the tumor cell, TAAs provide targets for the immune system to recognize and cause their destruction. Example of TAAs include, but are not limited to gangliosides (GM2), prostate specific antigen (PSA), alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA) (produced by colon cancers and other adenocarcinomas, e.g. breast, lung, gastric, and pancreas cancer s), melanoma associated antigens (MART-1, gplOO, MAGE 1,3 tyrosinase), papillomavirus E6 and E7 fragments, whole cells or portions/lysates of antologous tumor cells and allogeneic tumor cells.
[0311] An adjuvant may be used to augment the immune response to TAAs. Examples of adjuvants include, but are not limited to, bacillus Calmette-Guerin (BCG), endotoxin lipopolysaccharides, keyhole limpet hemocyanin (GKLH), interleukin-2 (IL-2), granulocyte- macrophage colony-stimulating factor (GM-CSF) and cytoxan, a chemotherapeutic agent which is believe to reduce tumor-induced suppression when given in low doses. , HDAC ACTIVITY ASSAY
[0312] Compound s according to the present invention may be screened for activity against one or more HDACs. Provided in tiiis example are assays for activity against HDACl, HDAC2, HDAC6 and HDAC8.
[0313] Purified HDACl, HDAC2, HDAC6, and HDACS may be obtained as follows. [0314] For HDACl, DNA encoding residues 1-482 of the full-length sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Xbal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the N-terminus. SEQ. ID. No. 1 corresponds to residues 1-482 with the N-terminal 6-histidine tag and SEQ. ID. No. 2 is the DNA sequence that was used to encode SEQ. ID. No. 1.
[0315] For HDAC2, DNA encoding residues 1-488 of the full-length sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Smal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the C-terminus. SEQ. ID. No. 3 corresponds to residues 1-488 with the C-terminal 6-histidine tag and SEQ. ID. No. 4 is the DNA sequence that was used to encode SEQ. ID. No. 3.
[0316] For HDAC6, DNA encoding residues 73-845 of the human enzyme may be amplified by PCR and cloned into the Smal site of pFastbac (Invitrogen), which incorporates a 6xHistidine tag at the C-terminus. SEQ. I.D. No.5 corresponds to residues 73-845 with the C- terminal 6-histidine tag and SEQ. ID. No. 6 is the DNA sequence that was used to encode SEQ. ID. No. 5.
[0317] For HD AC8 , DNA encoding residues 1 -377 corresponding to the entire sequence of the human enzyme may be amplified by PCR and cloned into the BamHI Smal sites of pFastbac (Invitrogen), which incorporates a 6-histidine tag at the N-terminus. SEQ. ID. No.7 corresponds to residues 1-377 with the N-terminal 6-histidine tag and SEQ. ID. No. 8 is the DNA sequence that was used to encode SEQ. ID. No. 7.
[0318] Recombinant baculo virus incorporating the HDAC constructs may be generated by transposition using the Bac-to-Bac system (Invitrogen). High-titer viral stocks may be generated by infection of Spodoptera frugiperda Sf9 cells; the expression of recombinant protein may be carried out by infection of Spodoptera frugiperda Sf9 or Trichoplusia ni Hi5 cells (Invitrogen) in 10L Wave Bioreactors (Wave Biotech).
[0319] Recombinant protein may be isolated from cellular extracts by passage over ProBond resin (Invitrogen). HDAC 1 and HDAC6 may then be treated with TEV protease for the removal of the N-terminal 6XHistidine affinity tag (residual uncleaved protein may be removed through a second passage over Probond Resin). Partially purified extracts of all HDACs may then be further purified by high pressure liquid chromatography over a BioSep S3000 gel filtration resin. The purity of HDAC proteins may be determined on denaturing SDS-PAGE gel. Purified HDACs may then be concentrated to a final concentration of 4.0 mg/ml for HDACl , 10 mg/ml for HDAC2, 4.0 mg/ml for HDAC6, and 3 mg/ml for HDAC8. The proteins may be either stored at -78 °C in a buffer containing 25mM TRIS-HCl pH 7.6, 150mM NaCI, O.lmM EDTA and 0.25 mM TCEP or at -20 °C in the presence of glycerol (final concentration of glycerol at 50%)
[0320] The inhibitory properties of compounds relative to HDACl , HDAC2, HDAC6 and HDAC8 may be determined using a white or black 384- well-plate format under the following reaction conditions: 25 mM Tris pH 8.0, 100 mM NaCI, 50 mM KC1, 0.1 mM EDTA, 0.01% Brij35, 0.1 mM TCEP. 50 uM tBoc-Lys(Ac)-AMC, 2% DMSO. Reaction product may be determined quantitatively by fluorescence intensity using a Fluorescence plate reader (Molecular Devices Gemini) with an excitation wavelength at 370 nm and emission at 480 nm (for white plates) or 465 nm (for black plates). [0321] The assay reaction may be initiated as follows: 5 ul of 150 uM tBoc-Lys(Ac)AMC was added to each well of the plate, followed by the addition of 5 ul of inhibitor (2 fold serial dilutions for 11 data points for each inhibitor) containing 6% DMSO. 5 ul of either HDAC 1 , HDAC2, HDAC6 or HDAC8 solution may be added to initiate the reaction (final enzyme concentrations were 2.5 nM for HDACl, 1 nM for HDAC2, 2.5 nM for HDAC6 and 10 nM for HDAC8). The reaction mixture may then be incubated at room temperature for 60 min, and quenched and developed by addition of 5 ul of 10 mM phenanthroline and 4 mg/ml trypsin (final concentration of phenanthroline is 2.5 mM, and trypsin is 1 mg/ml). Fluorescence intensities of the resulting reaction mixtures may be measured after a 30 minute incubation at room temperature.
[0322] IC50 values may be calculated by non-linear curve fitting of the compound concentrations and fluorescence intensities to the standard IC50 equation. As a reference point for this assay, suberanilohydroxamic acid (SAHA) showed an IC50 of 63 nM for HDACl, 69 nM for HDAC2, 108 nM for HDAC6 and 242 nM for HDAC8. [0323] The Section below provides examples of HDAC inhibitors that were assayed according to the above assays and found to have better than 1000 nM activity against HDAC 1 , HDAC2, HDAC6, and HDACS.
EXAMPLES
1. Synthetic Schemes For HDAC Inhibitors [0324] HDAC inhibitors according to the present invention may be synthesized according to a variety of reaction schemes. Some illustrative schemes are provided herein in the examples. Other reaction schemes could be readily devised by those skilled in the art.
Scheme 1:
Figure imgf000101_0001
heme 2:
Figure imgf000101_0002
General procedure for the synthesis of i¥1-substituted-phenyl-l,2-diamines (2).
[0325] To a solution of the appropriate l-fluoro-2-nitrobenzene (1, 1.77 mmol) and the appropriate amine (1.77 mmol) in DMF (5.0 mL) was added DIEA (1.94 mmol). The reaction was heated at 50-100 °C for 24-48 hrs and then cooled to ambient temperature. The resulting mixture was poured into H2O, extracted with EtOAc, washed with brine, and dried over
MgSO4. The organic layer was evaporated to dryness and the resulting material was purified if needed via flash chromatography to yield the desired 2-nitrophenylamines. The appropriate
N-substituted-2-nitrophenylamine (1.10 mmol) was dissolved in MeOH/AcOH (4: 1, 5.0 mL) and heated to 100 °C. Zinc dust (5.50 mmol) was added to the reaction portion wise until frothing ceased. The reaction was cooled to ambient temperature, filtered, and evaporated to dryness. The resulting N^substituted-phenyl-l^-diamine (2) was used without further purification. General procedure for the synthesis of 3-[3-(l-substituted-l/7-benzoimidazoI-2-yl)- phenylj-acrylic acids (4).
[0326] A solution of the appropriate 3-formyl-cinnamic acid (3, 0.57 mmol) and the appropriate N^substituted-phenyl-l^-diamine (2, 0.57 mmol) in EtOH (5.0 mL) was refluxed for 24-48 hrs. The reaction was cooled, evaporated to dryness and purified via flash chromatography to yield the desired 3-[3-(l-substituted-lH-benzoimidazol-2-yl)-phenyl]- acrylic acid (4).
General procedure for the synthesis of N«hydroxy-3-[3-(l -substituted- IH- benzoimidazoI-2-yl)-phenyl]-acrylamides (5).
[0327] To a solution of the appropriate 3-[3-(l-substituted-lH-benzoimidazol-2-yl)- phenyl] -acrylic acid (4, 0.25 mmol), and HOBt (0.38 mmol) in DMF (5.0 mL) was added EDCI (0.38mmol), 6>-(tetrahydro-ρyran-2-yl)-hydroxylamine (0.38 mmol), and DIEA (0.75 mmol). The reaction was stirred at ambient temperature for 18 hrs. The resulting mixture was poured into H2O, extracted with EtOAc, washed with brine, and dried over MgSO . The organic layer was evaporated to dryness and the resulting material was reconstituted in MeOH (2 mL). CSA (0.28 mmol) was added to the solution. The reaction was stirred for 2 hr at ambient temperature and, without further work-up, purified by preparative LCMS to yield the desired N-hydroxy-3-[3-(l-substitιιted-lH-benzoiιnidazol-2-yl)-phenyl]-acrylarnide (5).
General procedure for the synthesis of i¥-(2-anιino-phenyl)-3-[3-(l-substituted-lH- benzoimidagol-2-yl)-phenyl]-acrylanιide (6).
[0328] To a solution of the appropriate 3 -[3 -(1 -substituted- lH-benzoimidazol-2-yl)- phenyl] -acrylic acid (5, 0.25 mmol), and HOBt (0.38 mmol) in DMF (5.0 mL) was added EDCI (0.38mmol), 1 ,2-phenylenediamine (0.38 mmol), and DIEA (0.75 mmol). The reaction was stirred at ambient temperature for 18 hrs. The resulting mixture was poured into H2O, extracted with EtOAc, washed with brine, and dried over MgSO . The organic layer was evaporated to dryness, and the resulting material was reconstituted in MeOH (2 mL) and purified by preparative LCMS to yield the desired N-(2-amino-phenyl)-3-[3-(l-substituted- 1 H-benzoimidazol-2-yl)-phenyl] -acrylamide (6) . General procedure for the synthesis of N-hydroxy-3-[3-(l-substituted-lH- benzoimidazol-2-yl)-phenyl]-propionamides (7).
[0329] To a solution of the appropriate N-hydroxy-3-[3-(l-substituted-lH-benzoimidazol- 2-yl)-phenyl]- acrylamide (6; 0.65 mmol) in MeOH (1.0 mL) was added Pd/C (10%; 2.5 mg). H2(g) was bubbled through the stirring reaction for 1 hr. The reaction was filtered through Celite and purified via preparative LCMS to provide the desired N-hydroxy-3-[3-(l- substituted-lH-benzoimidazol-2-yl)-phenyl]-propionamide (7).
General procedure for the synthesis of 3-[3-(l-substituted-l/7-benzoimidazol-2-yl)- phenyl] -acrylic acids (9).
[0330] A solution of the appropriate 3-formyl-cinnamic acid methyl ester (8, 0.75 mmol) and the appropriate Ν^substituted-phenyl-l^-diamine (2, 0.75 mmol) in ethanol (2.0 mL) was heated at 80 °C for 24-48 hrs. The reaction was cooled, evaporated to dryness and purified via flash chromatography to yield the desired acrylic acid esters. To a solution of the appropriate acrylic acid methyl ester (0.50 mmol) in MeOH (1.0 mL) was added LiOH (1.0 mmol). The reaction was stirred at ambient temperature for 2 hrs and, poured into H2O, and acidified to pH = 2 witi HC1 (6Ν). The resulting heterogeneous mixture was tiien extracted with EtOAc. The organic layers were combined, washed with brine, dried over MgSO4, filtered, and evaporated to dryness to provide the appropriate 3-[3-(l-substituted-lH- benzoimidazol-2-yl)-phenyl]-acrylic acid (9) which were used in subsequent reactions without further purification.
General procedure for the synthesis of 3-[3-(l-substituted-lH-benzoimidazol-2-yl)- phenyl] -N-hydroxyacrylamides (10).
[0331] The procedure for the synthesis of N-hydroxy-3-[3-(l-substituted-lH- benzoimidazol-2-yl)-phenyl]-acrylamides (5) was used. Scheme 3:
Figure imgf000104_0001
General procedure for the synthesis of 3-[3-(lH-benzoimidazol-2-yl)-phenyI]-N- (tetrahydro-pyran-2-yloxy)-acrylamides (11).
[0332] A solution of the appropriate 3-formyl-cinnamic acid (3, 0.57 mmol) and the appropriate substituted-phenyl-l,2-diamine (0.57 mmol) in EtOH (5.0 mL) was refluxed for 24 hrs. The reaction was cooled, evaporated to dryness, and the resulting acrylic acid was used without further purification. To a solution of the acrylic acid (0.25 mmol) and HOBt (0.38 mmol) in DMF (5.0 mL) was added EDCI (0.38mmol), 0-(tetrahydro-pyran-2-yl)- hydroxylamine (0.38 mmol), and DIEA (0.75 mmol). The reaction was stirred at ambient temperature for 18 hrs. The resulting mixture was poured into H2O, extracted with EtOAc, washed with brine, dried over MgSO4 and concentrated to dryness. The resulting material was purified via flash chromatography to yield the desired 3-[3-(lH-benzoimidazol-2-yl)-phenyl]- N-(tetrahydro-pyran-2-yloxy)-acrylamide (11).
General procedure for the synthesis of 3-[3-(lH-benzoimidazol-2-yl)-phenyl]-N- hydroxy-acrylamide (12)
[0333] To a solution of the appropriate 3-[3-(lH-benzoimidazol-2-yl)-phenyl]-N- (tetrahydro-pyran-2-yloxy)-acrylamide (11, 0.25mmol) in MeOH (2 mL) was added CSA (0.28 mmol). The reaction was stirred for 2 hr at ambient temperature and, without further work-up, purified by preparative LCMS to yield the desired 3-[3-(lH-benzoimidazol-2-yl)- phenyl]-N-hydroxy-acrylamide (12). General procedure for the synthesis of N-hydroxy-3-[3-(l-substituted-liϊ- benzoimidazol-2-yl)-phenyl]- acrylamides (13).
[0334] To a solution of NaH (20 mg, 0.83 mmol) in anhydrous DMF (1 mL) was added a solution of 3-[3-(lH-benzoimidazol-2-yl)-phenyl]-N-(tetiahydro-pyran-2-yloxy)-acιylamide (11, 0.28 mmol) in anhydrous DMF drop wise at ambient temperature. The reaction was stirred for 30 min and then the appropriate alkyl bromide (0.31 mmol) was added. The reaction was stirred for 30 min, quenched with MeOH (200 μL) and partitioned between H2O and EtOAc. The organic layer was washed with brine, dried with MgSO4, and evaporated to dryness. The resulting material was reconstituted in MeOH (2 mL). CSA (0.31 mmol) was added to the solution. The reaction was stirred for 2 hr at ambient temperature and, without further work-up, purified by preparative LCMS to yield the desired N-hydroxy-3-[3-(l- substituted-lH-benzoimidazol-2-yl)-phenyl]- acrylamide (13).
[0335] As can be seen from the above reaction schemes, a wide variety of different HDAC inhibitors can be synthesized by these reaction schemes. It is noted that the invention is not intended to be limited to the particular compounds provided in this example. Rather, a wide variety of other compounds according to the present invention having HDAC inhibitory activity may be synthesized by the reaction schemes provided as well as otiier reaction schemes that may be devised by one of ordinary skill in the art in view of the present teachings.
2. Examples of Inhibitors According to the Present Invention [0336] Provided in this example are particular compounds that have been found to have HDAC8 activity based on the assay provided in Example 2. It is noted that these compounds may also have activity relative to other HDACs. It is also noted that these compounds are intended to illustrate various HDAC inhibitors according to the present invention and the present invention is not intended to be limited to these compounds:
COMPOUND 1.
Figure imgf000105_0001
3-r3-(lH-Benzoimidazol-2-yl)-phenyl1-N-hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 6.69 (d, IH), 7.45 (m, 3H), 7.82 (m, 5H), 8.14 (d, IH), 9.30 (s, IH), 10.90 (s, IH). ESI-MS: m/z 280.2 (M + H)+. COMPOUND 2.
Figure imgf000106_0001
N-Hvdroxy-3-r3-(l-memyl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 3.93 (s, 3H), 6.60 (d, IH), 7.33 (band, 2H), 7.60- 7.81 (band, 7H), 7.85 (m, IH), 8.05 (m, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: /z 294.3 (M + H)+.
COMPOUND 3.
Figure imgf000106_0002
N-Hydroxy-3-r3-fl-isopropyl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide. 1HNMR (400 MHz, OMSO-d6): δ 1.58 (d, 6H), 4.82 (m, IH), 6.57 (d, 6H), 7.15-7.68 (m, 8H), 8.12 (s, IH), 9.06 (s, IH), 10.78 (s, IH). ESI-MS: m/z 322.3 (M +H). COMPOUND 4.
Figure imgf000106_0003
(±)-3- { 3- f 1 -( 1 -Ethyl-piperidin-3-yD- lH-benzoinιidazol-2-yl] -phenyl } -N-hydroxy- acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 1.21 (t, 3H), 1.76 (m, IH), 2.23 (m, IH), 3.23 (m,
4H), 3.52 (m, IH), 3.77 (m, 2H), 4.79 (m, IH), 6.60 (d, IH), 7.41 (m, 2H), 7.54-7.91 (band,
6H), 8.14 (m, IH), 9.68 (s, IH). ESI-MS: m/z 391.2 (M + H)+. COMPOUND 5.
Figure imgf000107_0001
N-Hydroxy-3 -I3-I"l-(1 -methyl-piperidin-4-vD- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide.
1H ΝMR (400MHz, OMSO-d6): δ 2.27 (m, 2H), 2.80 (m, 5H), 3.20 (m, 2H), 3.57 (m, 2H), 4.70 (m, IH), 6.60 (d, IH), 7.45 (m, 2H), 7.60-7.95 (band, 6H), 8.14 (m, IH), 10.07 (s, IH). ESI-MS: m/z 377.1 (M + H)+.
COMPOUND 6.
Figure imgf000107_0002
N-Hydroxy-3- β-( 1 -piperidin-4-ylmethyl- 1 H-benzoimidazol-2-yl )-phenyl] -acrylamide.
The product is obtained in salt form with two 10-camphorsulfonic acid (CSA).
1H NMR (400MHz, DMSO-^6): δ 0.71 (s, 6H), 1.02 (s, 6H), 1.23 (m, 2H), 1.24 (q, 4H), 1.65 (m, 2H), 1.77 (m, 4H), 1.92 (t, 2H), 2.14 (m, IH), 2.22 (m, 2H), 2.35 (d, 2H), 2.62 (m, 2H), 2.65 (t, 2H), 2.85 (d, 2H), 3.12 (m, 2H), 4.47 (d, 2H), 6.64 (d, IH), 7.40-8.06 (band, 11H), 10.8 (s, IH). ESI-MS: m/z 377.1 (M + H)+.
COMPOUND 7.
Figure imgf000107_0003
N-Hydroxy-3- { 3-fl -(2-morpholin-4-yl-ethyl)- 1 H-benzoimidazol-2-vn -phenyl I - acrylamide.
The product is obtained in salt form with two 10-camphorsulfonic acid (CSA).
1H ΝMR (400 MHz, DMSO- ό): δ 0.71 (s, 6H), 1.02 (s, 6H), 1.24 (q, 4H), 1.78 (m,
4H), 1.92 (t, 2H), 2.22 (m, 2H), 2.35 (d, 2H), 2.65 (t, 2H), 2.81 (d, 2H), 3.14-3.95 (band, 8H), 6.62 (d, IH), 7.51-8.12 (band, 9H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 393.3 (M +H)+. COMPOUND 8.
Figure imgf000108_0001
N-Hvdroxy-3-r3-(l-phenyl-lH-benζoimidaζol-2-yl)-phenyl1-acrylamide. 1H ΝMR (400 MHz, DMSO- 6): δ 6.43 (d, IH), 6.38-7.83 (band, 14H), 9.16 (s, IH), 10.7 (s, IH). ESI-MS: m/z 356.4 (M +H) +. COMPOUND 9.
Figure imgf000108_0002
3 - { 3 - r 1 -(4-Chloro-phenyD- 1 H-benzoimidazol-2-yll -phenyl } -N-hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 6.45 (d, IH), 7.22-7.44 (band, 7H), 7.52 (d, 2H), 7.62 (d, IH), 7.66 (d, 2H), 7.82 (d, IH), 7.89 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 390.1 (M + H)+.
COMPOUND 10.
Figure imgf000108_0003
N-Hvdroxy-3-(3-ri-(4-memoxy-phenyl)-lH-benzoimidazol-2-yl1-phenyli-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 3.81 (s, 3H), 6.46 (d, IH), 7.12 (d, 2H), 7.17 (d, IH), 7.31 (m, 2H), 7.35-7.44 (band, 5H), 7.59 (d, IH), 7.79 (d, IH), 7.89 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 386.2 (M + H)+. COMPOUND 11.
Figure imgf000109_0001
N-Hydroxy-3- { 3- [ 1 -(4-phenoxy-phenvD- 1 H-benzoimidazol-2-yl1 -phenyl } -acrylamide.
1H ΝMR (400 MHz, DMSO-dό): δ 6.46 (d, IH), 7.10 (m, 3H), 7.26 (d, IH), 7.34 (m, 3H), 7.40-7.50 (band, 8H), 7.63 (d, IH), 7.79 (d, IH), 7.82 (s, IH), 9.12 (s, IH), 10.80 (s, IH). ESI-MS: m/z 448.2 (M + H)+.
COMPOUND 12.
Figure imgf000109_0002
3- { 3 - r 1 -(4-Benzyloxy-phenvD- 1 H-benzoimidazol-2-yll -phenyl } -N-hydroxy- acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 5.15 (s, 2H), 6.47 (d, IH), 7.17 (d, IH), 7.19 (d, 2H), 7.25-7.68 (band, 13H), 7.80 (d, IH), 7.90 (s, IH), 9.10 (s, IH), 10.81 (s, IH). ESI-MS: m/z 462.2 (M + H)+.
COMPOUND 13.
Figure imgf000109_0003
3-r3-(l-Benzyl-lH-benzoimidazol-2-yl)-phenyl1-N-hydroxy-acrylamιde.
1H ΝMR (400 MHz, DMSO- 6): δ 5.70 (s, 2H), 6.54 (d, IH), 7.08 (d, 2H), 7.29 (m, 3H), 7.49 (m, 2H), 7.65 (t, IH), 7.72 (d, IH), 7.76 (d, IH), 7.84 (d, 2H), 8.01 (s, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 370.2 (M + H)+. COMPOUND 14.
Figure imgf000110_0001
3 - { 3 - r 1 -(2-Chloro-benzyl)- 1 H-benzoimidazol-2-yll -phenyl } -N-hydroxy-acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 5.60 (s, 2H), 6.51 (d, IH), 6.65 (d, IH), 7.20-7.40 (band, 4H), 7.5-7.65 (band, 4H), 7.71 (d, IH), 7.81 (d, IH), 7.91 (s, IH), 9.10 (br s, IH), 10.88 (br s, IH). ESI-MS: m/z 404.2 (M + H)+.
COMPOUND 15.
Figure imgf000110_0002
3-{3-ri-(3-CMoro-benzyl)-lH-benzoimidazol-2-yl1-phenyl}-N-hydroxy-acrylaιτιide.
1H ΝMR (400 MHz, OMSO-d6): δ 5.60 (s, 2H), 6.54 (d, IH), 6.87 (m, IH), 7.09 (s, IH), 7.28 (m, 3H), 7.56 (m, 3H), 7.67 (d, IH), 7.76 (m, 2H), 7.94 (d, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 404.2 (M + H)+.
COMPOUND 16.
Figure imgf000110_0003
3 - { 3- f 1 -(4-Chloro-benzyl)- 1 H-benzoimidazol-2-vH -phenyl } -N-hydroxy-acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 5.60 (s, 2H), 6.54 (d, IH), 7.10 (d, 2H), 7.27 (m, 2H), 7.34 (d, 2H), 7.53 (m, 2H), 7.57 (d, IH), 7.66 (d, IH), 7.74 (d, 2H), 7.95 (s, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 404.2 (M + H)+. COMPOUND 17.
Figure imgf000111_0001
N-Hvdroxy-3-(3-[l-(2-trifluoromethoxy-benzyl -lH-benzoimidazol-2-yll-phenyl}- acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 5.65 (s, 2H), 6.52 (d, IH), 6.72 (d, IH), 7.26 (m, 3H), 7.38-7.55 (band, 4H), 7.58 (d, IH), 7.71 (d, IH), 7.78 (d, 2H), 7.92 (s, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 454.2 (M + H)+.
COMPOUND 18.
Figure imgf000111_0002
N-Hydroxy-3- { 3 - \ 1 -(3 -trifluoromethoxy-benzyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 5.71 (s, 2H), 6.55 (d, IH), 6.99 (m, 2H), 7.24 (d, 2H), 7.30 (d, IH), 7.53 (t, IH), 7.65 (m, 3H), 7.68 (d, IH), 7.76 (m, 2H), 7.92 (s, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 454.2 (M + H)+.
COMPOUND 19.
Figure imgf000111_0003
N-Hydroxy-3-{3-ri-(4-pyrazol-l-yl-benzyl)-lH-benzoimidazol-2-yl1-phenyll- acrylamide.
1H ΝMR (400 MHz, DMSO- ό): δ 5.66 (s, 2H), 6.56 (m, 2H), 7.13 (d, 2H), 7.28 (m, 2H), 7.55 (m, 3H), 7.68-7.76 (band, 6H), 8.00 (s, IH), 8.42 (d, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 436.2 (M + H)+. COMPOUND 20.
Figure imgf000112_0001
(R)-N-Hv(iroχy-3-l3-ri-(l-phenyl-emyl)-lH-beιιzoimidazol-2-yl1-phenv -acιylarmde.
1H ΝMR (400MHz, DMSO- ό): δ 1.97 (d, 3H), 5.85 (q, IH), 6.56 (d, IH), 7.07-7.32 (band, 8H), 7.52-7.72 (band, 5H), 7.92 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 384.1 (M + H)+.
COMPOUND 21.
Figure imgf000112_0002
(S)-N-Hydroxy-3-{3-[l-(l-phenyl-edιyl)-lH-benzoimidazol-2-yl1-phenyl}-acιylamide.
1HNMR (400MHz, DMSO-dβ): δ 2.00 (d, 3H), 5.85 (q, IH), 6.56 (d, IH), 7.07-7.32 (band, 8H), 7.52-7.72 (band, 5H), 7.92 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 384.1 (M + H)+.
COMPOUND 22.
Figure imgf000112_0003
(R)-N-Hydroxy-3- { 3- [ 1 -( 1 -p-tolyl-ethyl)- 1 H-benzoimidazol-2-yll-phenyl } -acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 1.95 (d, 3H), 2.22 (s, 3H), 5.80 (q, IH), 6.59 (d, IH), 7.05-7.22 (band, 7H), 7.52-7.73 (band, 4H), 7.78 (d, IH), 7.90 (s, IH), 9.15 (s, IH), 10.80 (s, IH). ESI-MS: m/z 398.2 (M + H)+. COMPOUND 23.
Figure imgf000113_0001
(R)-3-(3-{l-ri-(4-Huoro-phenyl)-ethyl1-lH-benzoimidazol-2-yl}-phenyl)-N-hvdroxy- acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 2.05 (d, 3H), 5.85 (q, IH), 6.59 (d, IH), 6.90-7.29 (band, 8H), 7.50-7.8 (band, 4H), 7.90 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 402.2 (M + H)+.
COMPOUND 24.
Figure imgf000113_0002
N-Hydroxy-3-r3-fl-phenethyl-lH-benzoimidazol-2-yl)-phenyl1-a rylamide.
1H ΝMR (400 MHz, DMSO-rfό): δ 2.99 (t, 2H), 4.53 (t, 2H), 6.53 (d, IH), 6.83 (m, 2H), 7.11 (t, 3H), 7.29 (m, 2H), 7.49-7.74 (band, 7H), 9.11 (s, IH), 10.8 (s, IH). ESI-MS: m/z 384.4 (M + H)+.
COMPOUND 25.
Figure imgf000113_0003
3-(3-π-r2-(3-Fluoro-phenyl -ethyl1-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide
1H ΝMR (400MHz, DMSO-d6): δ 3.00 (m, 2H), 4.57 (m, 2H), 6.55 (m, 2H), 6.67 (d, IH), 6.91 (m, IH), 7.13 (q, IH), 7.29 (m, 2H), 7.55 (m, 3H), 7.62 (s, IH), 7.67 (m, 3H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 402.1 (M + H)+.
-Ill- COMPOUND 26.
Figure imgf000114_0001
3-(3-{ l- [2-(4-Fluoro-phenyl)-ethyl1 - 1 H-benzoimidazol-2-yl } -phenyl)-N-hvdroxy- acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 2.96 (t, 2H), 4.55 (t, 2H), 6.53 (d, IH), 6.83-7.73 (band, 13H), 9.11 (s, IH), 10.78 (s, IH). ESI-MS: m/z 402.2 (M + H) +.
COMPOUND 27.
Figure imgf000114_0002
N-Hydroχy-3-(3-{ l-r2-f2-methoxy-phenyl)-ethyl1-lH-benzoimidazol-2-yl}-phenyl)- acrylamide.
1H NMR (400MHz, DMSO- 6): δ 2.95 (m, 2H), 3.48 (s, 3H), 4.53 (m, 2H), 6.52 (d, IH), 6.69 (m, 3H), 7.12 (m, IH), 7.30 (m, 2H), 7.55 (m, 4H), 7.70 (m, 3H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 414.17 (M + H)+.
COMPOUND 28.
Figure imgf000114_0003
N-Hydroxy-3-(3-( l-r2-(3-methoxy-phenyl)-ethyl1-lH-benzoimidazol-2-yll-phenyl)- acrylamide.
1H ΝMR (400MHz, DMSO-dό): δ 2.95 (m, 2H), 3.55 (s, 3H), 4.54 (m, 2H), 6.35 (m, 2H), 6.52 (d, IH), 6.65 (m, IH), 7.0 (m, IH), 7.27 (m, 2H), 7.45-7.80 (band, IB), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 414.1 (M + H)+. COMPOUND 29.
Figure imgf000115_0001
N-Hydroxy-3 -(3 - 11 - r2-(4-methoxy-phenyl)-ethyl] - lH-benζoimidaζol-2-yl ) -phenyl)- acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 2.95 (m, 2H), 3.65 (s, 3H), 4.50 (m, 2H), 6.55 (d, 4H), 6.65-6.80 (band, 4H), 7.37 (m, 2H), 7.45-7.80 (band, 7H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 414.1 (M + H)+.
COMPOUND 30.
Figure imgf000115_0002
3-(3-{ l-[2-(2-Chloro-phenyl)-ethyn-lH-benzoimidazol-2-yl}-phenyl)-N-hvdroxy- acrylamide.
1H ΝMR (400MHz, DMSQ- 6): δ 3.08 (m, 2H), 4.59 (m, 2H), 6.52 (d, IH), 6.79 (d, 1H), 7.11 (m, 3H), 7.31 (m, 2H), 7.54 (m, 4H), 7.71 (m, 3H), 9.10 (s, IH), 10.80 (s, IH). ESI- MS: m/z 418.1 (M + H)+.
COMPOUND 31.
Figure imgf000115_0003
3-(3-{ l-r2-(3-Chloro-phenyl)-ethyl1-lH-benzoimidazol-2-yll-phenyl)-N-hvdroxy- acrylamide. 1H NMR (400 MHz, DMSO- 6): δ 2.95 (t, 2H), 4.56 (t, 2H), 6.52 (d, IH), 6.88 (s, IH), 7.07-7.74 (band, 11H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 418.3 (M +H). COMPOUND 32.
Figure imgf000116_0001
3-(3-{ l-r2-(4-Chloro-phenyl)-ethyl1-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 2.96 (m, 2H), 4.56 (m, 2H), 6.54 (d, IH), 6.79 (d, 2H), 7.11 (m, 2H), 7.29 (m, 2H), 7.51 (m, 3H), 7.64-7.74 (band, 4H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 418.1 (M + H)+.
COMPOUND 33.
Figure imgf000116_0002
(±)-3-f3-( l-r2-(4-Huoro-phenyl)-l-me yl-emyll-lH-benzoimidazol-2-yll-phenyl)-N- hydroxy-acrylamide. H ΝMR (400 MHz, DMSO-dό): δ 1.68 (d, 3H), 3.11 (m, 2H), 4.68 (m, IH), 6.49 (d, IH), 6.58 (t, 2H), 6.84 (t, 2H), 7.20 (d, IH), 7.32 (s, IH), 7.45-7.75 (band, 5H), 9.10 (s, IH), 10.81 (s, IH). ESI-MS: m/z 416.2 (M + H) +.
COMPOUND 34.
Figure imgf000116_0003
(R)-N-Hvdroxy-3 - { 3 - [ 1 -(2-phenyl-propyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide.
1H NMR (400MHz, DMSO- 6): δ 1.10 (m, 3H), 3.10 (m, IH), 4.47 (m, 2H), 6.56 (d, IH), 6.75 (d, 2H), 7.10 (m, 3H), 7.30 (m, 2H), 7.50-7.80 (band, 7H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 398.2 (M + H)+.
COMPOUND 35.
Figure imgf000117_0001
(S)-N-Hydroxy-3-(3-ri-(2-phenyl-propyl)-lH-benzoimidazol-2-yll-phenyll- acrylamide.
1H ΝMR (400MHz, DMSO-dό): δ 1.10 (m, 3H), 3.10 (m, IH), 4.47 (m, 2H), 6.56 (d, IH), 6.75 (d, 2H), 7.10 (m, 3H), 7.3 (m, 2H), 7.50-7.80 (band, 7H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 398.2 (M + H)+.
COMPOUND 36.
Figure imgf000117_0002
(S)-N-Hydroxy-3 - ( 3 - \ 1 -( 1 -hvdroxymethyl-2-phenylethyl)- 1 H-benzoimidazol-2-yl1 - phenyl } -acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 3.08 (m, IH), 4.05 (m, IH), 4.40 (m, 2H), 5.24 (t, IH), 6.43 (d, IH), 6.55 (d, 2H), 7.00 (t, 2H), 7.10 (m, IH), 7.20-8.00 (band, 7H), 9.11 (s, IH), 10.81 (s, IH). ESI-MS: m/z 414.4 (M + H) +. COMPOUND 37.
Figure imgf000118_0001
(R)-N-Hydroxy-3 - { 3 - r 1 -( 1 -hydroxymethyl-2-phenylethyl)- lH-benzoimidazol-2-yl1 - phenyl I -acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 3.09 (m, IH), 4.06 (m, IH), 4.40 (m, 2H), 5.24 (t, IH), 6.43 (d, IH), 6.55 (d, 2H), 7.00 (t, 2H), 7.10 (m, IH), 7.20-8.00 (band, 7H), 9.11 (s, IH), 10.81 (s, IH). ESI-MS: m/z 414.4 (M + H) +.
COMPOUND 38.
Figure imgf000118_0002
(R)-N-Hydroxy-3- { 3- [ 1 -(2-hydroxy-2-phenyl-ethvD- lH-benzoimidazol-2-yl1 -phenyl } - acrylamide.
1H ΝMR (400 MHz, DMSO-Λ5): δ 4.40 (d, 2H), 5.91 (t, IH), 5.78 (br., IH), 6.55 (d, IH), 7.03-7.29 (band, 5H), 7.39 (t, 2H), 7.57 (m, 2H), 7.70-7.89 (band, 5H), 9.10 (s IH), 10.80 (s, IH). ESI-MS: m/z 400.2 (M +H) +.
COMPOUND 39.
Figure imgf000118_0003
N-Hydroxy-3 - ( 3 - [ 1 -(2-pyridin-2-yl-ethvD- 1 H-benzoimidazol-2-yn -phenyl I - acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 3.14 (t, 2H), 4.69 (t, 2H), 6.54 (d, IH), 6.80 (d, IH), 7.17 (t, IH), 7.26 (m, 2H), 7.52-7.94 (band, 7H), 8.38 (d, IH), 9.09 (s, IH), 10.78 (s, IH). ESI-MS: m/z 385.1 (M +H) +. COMPOUND 40.
Figure imgf000119_0001
N-Hydroxy-3 - ( 3- \ 1 -(2-pyridin-3 -yl-ethyl)- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide.
1H ΝMR (400MHz, OMSO-d6): δ 3.20 (m, 2H), 4.80 (m, 2H), 6.55 (d, IH), 7.46-7.90 (band, 11H), 7.95 (m, IH), 8.30 (m, IH), 8.54 (d, IH). ESI-MS: m/z 385.1 (M + H)+.
COMPOUND 41.
Figure imgf000119_0002
N-Hydroxy-3-{3-[l-(2-pyridin-4-yl-ethyl)-lH-benzoimidazol-2-yll-phenyl}- acrylamide.
1H ΝMR (400MHz, DMSO-Λ5): δ 3.25 (m, 2H), 4.82 (m, 2H), 6.52 (d, IH), 7.35-7.67 (band, 7H), 7.76-7.82 (m, 3H), 7.95 (m, IH), 8.54 (d, 2H). ESI-MS: m/z 385.1 (M + H)+.
COMPOUND 42.
Figure imgf000119_0003
N-Hvdroxy-3-(3- { 1 -f2-( 1 H-indol-3-yl)-ethyll- lH-benzoimidazol-2-yl } -phenyl)- acrylamide. lB ΝMR (400 MHz, DMSO-dtf): δ 3.09 (t, 2H), 4.53 (t, 2H), 6.52 (d, IH), 6.80 (m, 2H), 6.91 (s, IH), 7.25-8.13 (m, 14H), 9.11 (s, IH), 10.77 (s, IH). 12.7 (s, IH). ESI-MS: m/z
-111- 423.1 (M +H) +.
COMPOUND 43.
Figure imgf000120_0001
N-Hydroxy-3-[3-(l-indan-2-yl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide
1H ΝMR (400MHz, DMSO- 6): δ 3.56 (m, 4H), 5.51 (m, IH), 6.62(d, IH), 7.07-7.33 (band, 7H), 7.54-7.77 (band, 5H), 7.92 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 396.2 (M + H)+.
COMPOUND 44.
Figure imgf000120_0002
N-Hydroxy-3-{3-ri-(3-phenyl-piOpyl)-lH-benzoimidazol-2-yl]-phenyl}-acrylamide.
1H ΝMR (400MHz, OMSO-dδ): δ 2.07 (m, 2H), 2.53 (m, 2H), 4.29 (m, 2H), 6.56 (d,
IH), 7.07-7.32 (band, 8H), 7.52-7.72 (band, 5H), 7.92 (s, IH), 9.10 (s, IH), 10.80 (s, IH).
ESI-MS: m/z 398.2 (M + H)+. COMPOUND 45.
Figure imgf000120_0003
3 - [3 -(5 -Fluoro- 1 H-benzoimidazol-2-yl)-phenyll -N-hydroxy-acrylamide. 1H ΝMR (400 MHz, DMSO-dό): δ 6.61 (d, IH), 7.19 (m, IH), 7.52 (m, 2H), 7.70 (m, 3H), 8.13 (d, IH), 8.38 (s, IH), 9.40 (s, IH), 10.90 (s, IH). ESI-MS: m/z 298.2 (M + H)+. COMPOUND 46.
Figure imgf000121_0001
N-Hvdroxy-3-r3-(6-propylsulfanyl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.92 (t, 3H), 1.53 (m, 2H), 2.95 (t, 2H), 6.58 (d, IH), 7.31 (dd, IH), 7.50 (d, IH), 7.62 (m, 3H), 7.73 (d, IH), 8.08 (d, IH), 8.38 (s, IH), 9.40 (s, IH), 10.90 (s, IH). ESI-MS: m/z 354.2 (M + H)+.
COMPOUND 47.
Figure imgf000121_0002
N-Hvdroxy-3-r3-(7-methyl-lH-benzoin idazol-2-yl)-phenyll-acrylamide.
1H ΝMR (400 MHz, DMSO-dδ): δ 2.63 (s, 3H), 6.58 (d, IH), 7.27 (t, IH), 7.50 (d,
IH), 7.53 (s, 2H), 7.65 (I, IH), 7.75 (d, IH), 8.14 (d, IH), 8.38 (s, IH), 9.40 (s, IH), 10.90 (s,
IH). ESI-MS: m/z 294.2 (M + H)+. COMPOUND 48.
Figure imgf000121_0003
2-r3-(2-Hydroxycarbamoyl-vinyl)-phenyll-3H-benzoimidazole-5-carboxylic acid methyl ester.
1H NMR (400 MHz, DMSO- 6): δ 3.91 (s, 3H), 6.62 (d, IH), 7.54 (d, IH), 7.62 (t, IH), 7.74 (m, 2H), 7.89 (d, IH), 8.14 (d, IH), 8.23 (s, IH), 8.40 (s, IH), 9.40 (s, IH), 10.90 (s, IH). ESI-MS: m/z 338.2 (M + H)+.
COMPOUND 49.
Figure imgf000121_0004
3-[3-(6,7-Difluoro-lH-benzoimidazol-2-yl)-phenyl1-N-hydroxy-acrylamide. 1H ΝMR (400 MHz, DMSO-cfø): δ 6.58 (d, IH), 7.21 (m, IH), 7.35 (m, IH), 7.48 (d, IH), 7.54 (t, IH), 7.65 (d, IH), 8.09 (d, IH), 8.38 (s, IH), 9.40 (s, IH), 10.90 (s, IH). ESI- MS: m/z 316.2 (M + H)+. COMPOUND 50.
Figure imgf000122_0001
3-13-11 -(4-Methoxy-phenyl)- 1 H-benζoimidaζol-2-yl] -phenyl } -acrylic acid.
1H NMR (400 MHz, OMSO-d6): δ 3.72 (s, 3H), 6.23 (d, IH), 7.06 (d, 2H), 7.10 (d, IH), 7.23 (m, 2H), 7.29-7.39 (band, 3H), 7.42 (s, IH), 7.49 (d, IH), 7.62 (d, IH), 7.69 (s, IH), 7.73 (d, IH). ESI-MS: m z 371.2 (M + H)+.
COMPOUND 51.
Figure imgf000122_0002
3-13-11 -(4-Benzyloxy-phenyD- 1 H-benzofmidazol-2-yll -phenyl } -acrylic acid.
1H NMR (400 MHz, ΩMSO-d6): δ 5.10 (s, 2H), 6.23 (d, IH), 7.10 (d, IH), 7.14 (m, 2H), 7.18-7.38 (band, 8H), 7.43 (m, 3H), 7.52 (d, IH), 7.64 (d, IH), 7.78 (s, IH), 7.73 (d, IH). ESI-MS: m/z 447.2 (M + H)+.
COMPOUND 52.
Figure imgf000122_0003
3- { 3 - f 1 -(4-Chloro-phenyD- 1 H-benzoimidazol-2-yll -phenyl 1 -acrylic acid. 1H NMR (400 MHz, DMSO- 6): δ 6.35 (d, IH), 7.25 (d, IH), 7.33 (m, 2H), 7.41-7.53 (band, 5H), 7.66 (d, 2H), 7.72 (d, IH), 7.77 (s, IH), 7.82 (d, IH). ESI-MS: m/z 375.2 (M + H)+.
COMPOUND 53.
Figure imgf000123_0001
(R)-3-(3- { 1 - [ 1 -(4-Fluoro-phenyl)-ethyl1 - lH-benzo imidazol-2-yl I -phenyD-acrylic acid.
1H NMR (400 MHz, DMSO- 6): δ 2.05 (d, 3H), 5.89 (q, IH), 6.65 (d, IH), 7.15-7.33 (band, 7H), 7.68 (m, 2H), 7.76 (d, 2H), 7.96 (d, IH), 7.98 (s, IH). ESI-MS: m/z 387.2 (M + H)+.
COMPOUND 54.
Figure imgf000123_0002
(R)-3-{3-ri-(l-p-Tolyl-ethyl)-lH-benzoimidazol-2-yl1-phenyll-acrylic acid.
1H NMR (400 MHz, DMSO-^6): δ 1.95 (d, 3H), 2.23 (s, 3H), 5.80 (q, IH), 6.55 (d, IH), 7.04-7.27 (band, 7H), 7.63 (m, 2H), 7.71 (m, 2H), 7.90 (m, 2H). ESI-MS: m/z 383.2 (M + H)+.
COMPOUND 55.
Figure imgf000123_0003
3-{3-ri-(4-Phenoxy-phenyl)-lH-benzoimidazol-2-yl1-phenyl)-acrylic acid.
1H NMR (400 MHz, DMSO-dό): δ 6.25 (d, IH), 7.05 (d, 2H), 7.12 (m, 2H), 7.18 (m,
2H), 7.25 (m, 2H), 7.35-7.42 (band, 5H), 7.46 (d, IH), 7.64 (m, 3H), 7.76 (d, IH). ESI-MS: m/z 433.2 (M + H)+. COMPOUND 56.
Figure imgf000124_0001
N-(2- Amino-phenyl)-3 - ( 3 - [ 1 -(4-methoxy-phenyl)- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide.
XH ΝMR (400 MHz, DMSO- 6): δ 3.81 (s, 3H), 5.01 (br s, 2H), 6.58 (t, IH), 6.76 (d, IH), 6.93 (t, IH), 6.97 (d, IH), 7.13 (d, 2H), 7.16 (d, IH), 7.26-7.43 (band, 6H), 7.50 (d, IH), 7.63 (d, IH), 7.80 (d, IH), 8.05 (s, IH), 8.21 (s, IH) 9.42 (s, IH). ESI-MS: m/z 461.2 (M + H)+.
COMPOUND 57.
Figure imgf000124_0002
N-(2-Amino-phenyl)-3- { 3-[ l-(4-benzyloxy-phenyl)- lH-benzoimidazol-2-yll-phenyl } - acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 5.00 (br s, 2H), 5.15 (s, 2H), 6.58 (t, IH), 6.76 (d, IH), 6.93 (t, IH), 6.98 (d, IH), 7.17 (d, IH), 7.21 (d, 2H), 7.26-7.45 (band, 9H), 7.50 (m, 3H), 7.65 (d, IH), 7.81 (d, IH), 8.05 (s, IH), 9.50 (s, IH). ESI-MS: m/z 537.2 (M + H)+. COMPOUND 58.
Figure imgf000125_0001
N-(2-Amino-phenyl)-3- 13-[ 1 -(4-chloro-phenvD- lH-benzoimidazol-2-yll -phenyl 1 - acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 5.01 (br s, 2H), 6.52 (t, IH), 6.69 (d, IH), 6.85 (t, IH), 6.94 (d, IH), 7.19 (d, IH), 7.22-7.39 (band, 6H), 7.46 (m, 3H), 7.60 (m, 3H), 7.76 (d, IH), 7.96 (s, IH), 9.38 (s, IH). ESI-MS: m/z 465.2 (M + H)+.
COMPOUND 59.
Figure imgf000125_0002
ro-phenyl)-edιyl1-lH-benzoimidazol-2-yl}- phenyD-acrylamide.
1H ΝMR (400 MHz, DMSO-dό): δ 2.00 (d, 3H), 5.00 (br s, 2H), 5.89 (q, IH), 6.59 (t, IH), 6.77 (d, IH), 6.93 (t, IH), 7.04 (d, IH), 7.10-7.28 (band, 7H), 7.38 (d, IH), 7.62-7.74 (band, 4H), 7.84 (d, IH), 8.01 (s, IH), 9.45 (s, IH). ESI-MS: m/z All.2 (M + H)+.
COMPOUND 60.
Figure imgf000125_0003
(R)-N-(2-Amino-phenyl)-3-l3-ri-(l-ρ-tolyl-ethyl)-lH-benzoimidazol-2-yll-ρhenyll- acrylamide.
1H ΝMR (400 MHz, DMSO-J6): δ 1.94 (d, 3H), 2.23 (s, 3H), 4.98 (br s, 2H), 5.84 (q, IH), 6.57 (t, IH), 6.76 (d, IH), 6.92 (t, IH), 7.00-7.22 (band, 8H), 7.36 (d, IH), 7.60-7.71 (band, 4H), 7.82 (d, IH), 7.97 (s, IH), 9.40 (s, IH). ESI-MS: m/z 473.2 (M + H)+. COMPOUND 61.
Figure imgf000126_0001
N-(2- Amino-phenyl)-3 - { 3 - f 1 -(4-phenoxy-phenvD- 1 H-benzoimidazol-2-yl1 -phenyl I - acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 5.00 (br s, 2H), 6.59 (t, IH), 6.76 (d, IH), 6.93 (t, IH), 6.96 (d, IH), 7.11 (d, 2H), 7.19 (m, 3H), 7.23-7.39 (band, 4H), 7.41-7.55 (band, 7H), 7.67 (t, IH), 7.93 (s, IH), 8.05 (s, IH), 8.21 (s, IH) 9.45 (s, IH). ESI-MS: m/z 523.2 (M + H)+.
COMPOUND 62.
Figure imgf000126_0002
N-Hvdroxy-3-r4-(l-phenethyl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1H ΝMR (400 MHz, DMSO-dό): δ 3.00 (t, 2H), 4.62 (t, 2H), 6.60 (d, IH), 6.92 (m, 2H), 7.15 (m, 3H), 7.44 (m, 2H), 7.55 (d, IH), 7.62 (d, 2H), 7.75 (m, 3H), 7.88 (d, IH), 9.10 (s, IH), 10.88 (s, IH). ESI-MS: m/z 384.2 (M + H)+.
COMPOUND 63.
Figure imgf000126_0003
N-(2-Amino-phenyl)-3-r4-(l-phenethyl-lH-benzoimidazol-2-yl)-phenyll-acrylamide.
1H ΝMR (400 MHz, DMSO- c5): δ 3.00 (t, 2H), 4.65 (t, 2H), 6.71 (t, IH), 6.83 (d,
IH), 6.92 (m, 2H), 6.99 (t, IH), 7.11 (d, IH), 7.16 (m, 3H), 7.43 (m, 3H), 7.65 (m, 3H), 7.78 (m, 3H), 7.88 (d, IH), 9.78 (s, IH). ESI-MS: m/z 459.2 (M + H)+. COMPOUND 64.
Figure imgf000127_0001
3- [4-( 1 -Phenethyl- lH-benzoimidazol-2-yl)-phenyl1 -acrylic acid.
1H NMR (400 MHz, DMSO- 6): δ 2.90 (t, 2H), 4.45 (t, 2H), 6.56 (d, IH), 6.84 (m, 2H), 7.06 (m, 3H), 7.20 (m, 2H), 7.52 (m, 3H), 7.61 (m, 2H), 7.71 (d, 2H). ESI-MS: m/z 369.2 (M + H)+.
COMPOUND 65.
Figure imgf000127_0002
N-Hydroxy-3- { 3-fl -(2-piperidin- 1 -yl-ethyl)- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide.
1H ΝMR (400 MHz, ΩMSO-dά): δ 1.19-1.29 (m, 6H), 2.10-2.20 (m, 4H), 2.83 (t, 2H), 4.49 (t, 2H), 6.56 (d, IH), 7.22-7.30 (band, 2H), 7.56-7.82 (band, 6H), 7.96 (s, IH), 9.09 (br.,s, IH), 10.86 (br.,s, IH). ESI-MS: m/z 391.4 (M + H) +.
COMPOUND 66.
Figure imgf000127_0003
N-Hydroxy-3-{3-ri-(trøn^-4-hydroxy-cyclohexyl)-lH-berιzoimidazol-2-yll-phenyl}- acrylamide.
1H ΝMR (400 MHz, DMSO- 5): δ 1.20 (m, 2H), 2.95 (m, 4H), 2.48 (m, 2H), 3.64 (m, IH), 4.42 (m, IH), 4.62 (d, IH), 6.54 (d, IH), 7.23 (m, 2H), 7.56-7.86 (band, 6H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 378.2 (M +H) +. COMPOUND 67.
Figure imgf000128_0001
3 - \3 -( 1 -tert-Butyl- 1 H-benζoimidaζol-2-yl)-phenyn -N-hvdroxy-acrylamide. 1H ΝMR (400 MHz, DMSO- 6): δ 1.58 (d, 9H), 6.53 (d, IH), 7.25-7.96 (band, 8H), 9.06 (s, IH), 10.78 (s, IH). ESI-MS: m/z 336.3 (M + H)+. COMPOUND 68.
Figure imgf000128_0002
(±)-3 - Ϊ3 -( 1 -Cyclohexyl- 1 H-benzoimidazol-2-yl)-phenyl1 -N-hydroxy-acrylamide 1H ΝMR (400 MHz, DMSO-d6): δ 0.88 (m, 2H), 1.28 (m, 4H), 1.57 (m, 4H), 4.14 (m,
IH), 6.53 (d, IH), 7.25-7.91 (band, 9H), 9.06 (s, IH), 10.71 (s, IH). ESI-MS: m z 362.2 (M
+H) +.
COMPOUND 69.
Figure imgf000128_0003
3-{2-[3-(2-Hydroxycarbamoyl-vinyl)-phenyl1-benzoimidazol-l-yl}-piperidine-l- carboxylic acid tert-butyl ester
1H NMR (400MHz, DMSO- 6): δ 1.24-1.42 (band, 1 IH), 1.78 (d, IH), 2.09 (m, IH), 2.56 (m, IH), 2.97 (m, IH), 3.65 (m, IH), 3.89 (m, IH), 4.27 (m, IH), 6.59 (d, IH), 7.29 (m, 2H), 7.56-7.99 (band, 7H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 463.2 (M + H)+. COMPOUND 70.
Figure imgf000129_0001
(±)-N-Hydroxy-3-[3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1HΝMR (400MHz, DMSO- 6): δ 0.82 (m, IH), 1.24 (m, IH), 1.44 (m, IH), 1.76 (m, IH), 2.06 (m, IH), 2.45 (m, IH), 2.67 (m, IH), 2.89 (m, IH), 3.14 (m, IH), 3.14 (m, IH), 4.37 (m, IH), 6.57-6.61 (d, IH), 7.26 (m, 2H), 7.56-7.99 (band, 7H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 363.2 (M + H)+.
COMPOUND 71.
Figure imgf000129_0002
3-(3-ri-(2-Diethylamino-ethyl)-lH-benzoimidazol-2-yn-phenyl}-N-hydiOxy- acrylamide. lB ΝMR (400MHz, DMSO-dό): δ 0.63 (t, 6H), 2.25 (q, 4H), 2.62 (t, 2H), 4.36 (t, IH), 6.59 (d, IH), 7.28 (m, 2H), 7.55-8.10 (band, 7H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 379.2 (M + H)+.
COMPOUND 72.
Figure imgf000129_0003
(±)-N-Hvdroxy-3- { 3 - F 1 -( 1 -methyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 1.50 (m, IH), 1.78 (d, IH), 2.02 (m, IH), 2.25 (m, 5H), 2.74 (m, 2H), 2.94 (m, IH), 4.42 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.56-7.88 (band, 7H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 376.19 (M + H)+. COMPOUND 73.
Figure imgf000130_0001
(±)-N-Hy( oxy-3-(3-ri-(l-isopropyl-piperiα^n-3-yl)-lH-benzoimidazol-2-yll-phenyl|- acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 0.95 (m, 6H), 1.45 (m, IH), 1.78 (d, IH), 2.02 (m, IH), 2.26-2.31 (m, 2H), 2.68-2.76 (m, 2H), 2.94-2.96 (d, 2H), 4.35-4.37 (m, IH), 6.57-6.61 (d, IH), 7.23-7.30 (m, 2H), 7.55-7.89 (band, 7H), 9.09 (s, IH), 10.86 (s, IH). ESI-MS: m/z 405.2 (M + H)+.
COMPOUND 74.
Figure imgf000130_0002
(±)-N-Hydroxy-3-(3- { l-fl -(2-hvdroxy-ethyl)-piperidin-3-yll- lH-benzoimidazol-2-yl I- phenyl)-acrylamide.
1H ΝMR (400 MHz, DMSO- d6): δ 1.49-1.55 (m, IH), 1.78 (d, IH), 2.02-2.05 (m, IH), 2.12-2.18 (t, IH), 2.28-2.33 (m, IH), 2.45 (t, 2H), 2.83-2.89 (t, 2H), 3.02 (m, IH), 3.45-3.49 (m, 2H), 4.40 (m, IH), 6.57-6.61 (d, IH), 7.23-7.29 (m, 2H), 7.55-7.71 (band, 4H), 7.78 (m, IH), 7.86-7.89 (m, 2H). ESI-MS: m/z 407.2 (M + H)+.
COMPOUND 75.
Figure imgf000131_0001
(±)-3-{3-ri-(l -Ethyl-pyrrolidin-2-ylmethyl) - 1 H-benzoimidazol-2-yll -phenyl I -N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.69 (t, 3H), 1.29 (m, IH), 1.44 (m, 3H), 2.00 (m, 2H), 2.22 (m, IH), 2.85 (m, 2H), 4.31 (m, 2H), 6.59 (d, IH), 7.27 (m, 2H), 7.55-7.75 (band, 5H), 7.90 (d, IH), 8.12 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 391.1 (M + H)+.
COMPOUND 76.
Figure imgf000131_0002
(±)-3-{3-fl-(l -Ethyl-pyrrolidin-3 -yl)- 1 H-benzoimidazol-2-yll -phenyl } -N-hydroxy- acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 1.10 (t, 3H), 2.33 (m, 4H), 2.63 (m, 2H), 3.19 (m, 2H), 5.10 (m, IH), 6.59 (d, IH), 7.27 (m, 2H), 7.55-7.71 (band, 4H), 7.77 (d, IH), 7.85 (s, IH), 8.23-8.25 (d, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 377.1 (M + H)+.
COMPOUND 77.
Figure imgf000132_0001
(±)-3 - { 3- f 1 -(2-Dimethylamino-ethyl)- 1 H-benzoimidazol-2-vH -phenyl ) -N-hydroxy- acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 2.00 (s, 6H), 2.55 (t, 2H), 4.38 (t, 2H), 6.59 (d, IH), 7.27 (m, 2H), 7.55-7.80 (band, 6H), 7.99 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 351.1 (M + H)+.
COMPOUND 78.
Figure imgf000132_0002
(±)-3- { 3- f 1 -(2-Dimethylamino- 1 -methyl-ethyl)- 1 H-benzoimidazol-2-yll -phenyl } -N- hydiOxy-acrylamide.
1HΝMR (400 MHz, DMSO- 6): δ 1.62 (d, 3H), 1.87 (s, 6H), 2.50 (m, IH), 3.05 (m, IH), 4.60 (m, IH), 6.57 (d, IH), 7.24 (m, 2H), 7.54-7.82 (band, 6H), 7.86 (s, IH), 9.11 (s, IH), 10.79 (s, IH). ESI-MS: m/z 365.1 (M + H)+.
COMPOUND 79.
Figure imgf000133_0001
(±)-3-{3-ri-(2-Diisopropylaιmno-e yl)-lH-benzoimidazol-2-yl1-phenyl)-N-hydroxy- acrylamide.
1H ΝMR (400 MHz, DMSO- ): δ 0.62 (d, 12H), 2.57 (t, 2H), 2.75 (m, 2H), 4.28 (t, 2H), 6.59 (d, IH), 7.28 (m, 2H), 7.55-7.85 (band, 6H), 8.00 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 407.1 (M + H)+.
COMPOUND 80.
Figure imgf000133_0002
(±)-3-{3-[l-(l-Aza-bicyclor2.2.21oct-2-yl)-lH-benzoimidazol-2-yl1-phenyl}-N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 1.53 (m, IH), 1.64 (m, 2H), 2.06 (m, IH), 2.30 (m, IH), 2.73 (m, 2H), 2.90 (m, IH), 3.20 (m, IH), 3.35 (m, IH), 3.60 (m, IH), 4.60 (m, IH), 6.57 (d, IH), 7.30 (m, 2H), 7.53-7.90 (band, 7H), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 389.1 (M + H)+. COMPOUND 81.
Figure imgf000134_0001
(R)-3-(3-ri-(l-Ethyl-piperidin-3-yl)-6.7-difluoro-lH-benzoimidazol-2-vn-phenyll-N- hydroxy-acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 0.96 (t, 3H), 1.47 (m, IH), 1.79 (m, IH), 1.96 (m, 4H), 2.37 (m, 2H), 2.83 (m, IH), 3.07 (m, IH), 4.39 (m, IH), 6.59 (d, IH), 7.39 (m, IH), 7.58 (m, 2H), 7.70 (d, 2H), 7.81 (m, IH), 7.92 (s, IH), 9.10 (s, IH), 10.80 (s, IH). ESI-MS: m/z 427.1 (M + H)+.
COMPOUND 82.
Figure imgf000134_0002
(R)-3-{3-ri-(l-Ed yl-piperidin-3-yl)-5,6-difluoiO-lH-benzoiιnidazol-2-yll-phenyl}-N- hydroxy-acrylamide-trifluoroacetic acid.
1H ΝMR (400 MHz, OMSO-d6): δ 1.18 (t, 3H), 1.60 (m, IH), 2.14 (m, 2H), 3.22 (m, 3H), 3.55 (m, IH), 3.82 (m, 3H), 4.72 (m, IH), 6.59 (d, IH), 7.61 (m, 3H), 7.85 (m, 3H), 8.27 (m, IH), 9.40 (s, IH), 10.80 (s, IH). ESI-MS: m/z 427.1 (M + H)+.
COMPOUND 83.
Figure imgf000135_0001
(R)-3-{3-ri-(l-Ethyl-piperidin-3-yl)-4.6-difluoro-lH-benzoimidazol-2-yll-phenyl)-N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.98 (t, 3H), 1.42 (m, IH), 1.78 (m, IH), 2.10 (m, 2H), 2.35 (m, 3H), 2.71 (m, 2H), 3.00 (m, IH), 4.36 (m, IH), 6.59 (d, IH), 7.18 (m, IH), 7.57 (d, IH), 7.69 (m, 2H), 7.81 (m, IH), 7.85 (s, IH), 7.92 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 427.1 (M + H)+.
COMPOUND 84.
Figure imgf000135_0002
(R)-3-{3-ri-(l-Etfayl-piperidin-3-yl)-5,6 -trifluoro-lH-benzoimidazol-2-yll-phenyl}- N-hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.95 (t, 3H), 1.47 (m, IH), 1.78 (m, IH), 2.03 (m, 3H), 2.38 (m, 3H), 2.85 (m, IH), 3.10 (m, IH), 4.42 (m, IH), 6.59 (d, IH), 7.62 (d, IH), 7.66 (m, 2H), 7.74 (m, IH), 7.84 (m, 2H), 9.11 (s, IH), 10.81 (s, IH). ESI-MS: m/z 445.1 (M + H)+.
Figure imgf000136_0001
(R)-3- { 3-1" l-( 1 -Ethyl-piperidin-3-yl)-4,6 -trifluoro- lH-benzoimidazol-2-yll-phenyl } - N-hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.95 (t, 3H), 1.46(m, IH), 1.78 (m, IH), 2.03 (m, 3H), 2.37 (m, 3H), 2.85 (m, IH), 3.10 (m, IH), 4.42 (m, IH), 6.64 (d, IH), 7.54 (m, 2H), 7.67 (m, 2H), 7.83 (m, IH), 7.88 (s, IH), 9.12 (s, IH), 10.81 (s, IH). ESI-MS: m/z 445.1 (M + H)+.
COMPOUND 86.
Figure imgf000136_0002
(R)-3- { 3- f 1 -( 1 -Ethyl-piperidin-3-yl)-4,5,6,7-tetrafluoro- 1 H-benζoimidaζol-2-yl1- phenyl } -N-hydroxy-acrylamide.
*H ΝMR (400 MHz, OMSO-d6): δ 0.95 (t, 3H), 1.46 (m, IH), 1.79 (m, IH), 2.04 (m, 3H), 2.37 (m, 3H), 2.85 (m, IH), 3.17 (m, IH), 4.45 (m, IH), 6.60 (d, IH), 7.58 (d, IH), 7.67 (m, 2H), 7.85 (m, IH), 7.88 (s, IH), 9.12 (s, IH), 10.81 (s, IH). ESI-MS: m/z 463.1 (M + H)+.
COMPOUND 87.
Figure imgf000136_0003
(R)-3-(3-ri-ri-Ethyl-ρiρeridin-3-yl)-4-fluoro-lH-benzoimidazol-2-yll-phenyl}-N- hvdroxy-acrylamide. 1HNMR (400 MHz, OMSO-d6): δ 0.96 (t, 3H), 1.48 (m, IH), 1.79 (m, IH), 2.05 (m, 2H), 2.32 (m, 3H), 2.71 (m, IH), 2.84 (m, IH), 3.04(m, IH), 4.38 (m, IH), 6.60 (d, IH), 7.08 (m, IH), 7.27 (m, IH), 7.57 (d, IH), 7.65 (m, 2H), 7.73 (d, IH), 7.80 (m, IH), 7.87 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 409.1 (M + H)+.
COMPOUND 88.
Figure imgf000137_0001
(R)-3-{3-[l-(l -Ethyl-piperidin-3-yl)-5-fluoro- 1 H-benzoimidazol-2-yn -phenyl } -N- hydroxy-acrylamide.
1H ΝMR (400 MHz, OMSO-d6): δ 0.96 (t, 3H), 1.48 (m, IH), 1.79 (m, IH), 2.05 (m, 2H), 2.32 (m, 3H), 2.71 (m, IH), 2.84 (m, IH), 3.03 (m, IH), 4.38 (m, IH), 6.59 (d, IH), 7.14 (m, IH), 7.49 (m, IH), 7.57 (d, IH), 7.65 (m, 2H), 7.79 (m, IH), 7.89 (s, IH), 7.90-7.93 (m, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 409.1 (M + H)+.
COMPOUND 89.
Figure imgf000137_0002
(R)-3-{3-ri-(l-Ethyl-piperidin-3-yl)-6-fluoro-lH-benzoimidazol-2-yll-phenyl}-N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.96 (t, 3H), 1.47 (m, IH), 1.78 (m, IH), 2.11 (m, 2H), 2.32 (m, 3H), 2.71 (m, IH), 2.83 (m, IH), 3.00 (m, IH), 4.36 (m, IH), 6.59 (d, IH), 7.12 (m, IH), 7.56 (d, IH), 7.63 (m, 2H), 7.70 (m, IH), 7.77 (m, 2H), 7.84 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 409.1 (M + H)+. COMPOUND 90.
Figure imgf000138_0001
(R)-N-Hydroxy-3 - { 3 - [" 1 -( 1 -isopropyl-piperidin-3-yl)- 1 H-benzoimidazole-2-yn - phenyl I -acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 0.95 (d, 6H), 1.45 (m, IH), 1.79 (m, IH), 2.01 (m, IH), 2.29 (m, 2H), 2.74 (m, 2H), 3.04 (m, 2H), 4.35 (m, IH), 6.59 (d, IH), 7.25 (m, 2H), 7.56 (d, IH), 7.63 (m, 2H), 7.69 (m, IH), 7.79 (m, IH), 7.84 (s, IH), 7.88 (d, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 405.2 (M + H)+.
COMPOUND 91.
Figure imgf000138_0002
(R)-3- { 3-r4-Fluoro- 1 -( 1 -isopropyl-piperidm-3-yl)- lH-benzoimidazol-2-yll-phenyl } -N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- ): δ 0.95 (d, 6H), 1.45 (m, IH), 1.79 (m, IH), 2.02 (m, IH), 2.27 (m, 2H), 2.73 (m, 2H), 2.98 (m, 2H), 4.35 (m, IH), 6.60 (d, IH), 7.08 (m, IH), 7.26 (m, IH), 7.57 (d, IH), 7.65 (m, 2H), 7.74 (d, IH), 7.80 (m, IH), 7.86 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 423.1 (M + H)+. COMPOUND 92.
Figure imgf000139_0001
(R)-3-{3-r5-Ruoro-l-(l-isopropyl-piperiάin-3-yl)-lH-benzoimidazol-2-yll-phenyl)-N- hvdroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- ): δ 0.95 (d, 6H), 1.44 (m, IH), 1.79 (m, IH), 2.04 (m, IH), 2.27 (m, 2H), 2.73 (m, 2H), 2.98 (m, 2H), 4.35 (m, IH), 6.59 (d, IH), 7.14 (m, IH), 7.50 (m, IH), 7.56 (d, IH), 7.64 (m, 2H), 7.79 (m, IH), 7.84 (s, IH), 7.92 (m, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 423.1 (M + H)+.
COMPOUND 93.
Figure imgf000139_0002
(R)-3- { 3- [4-Fluoro- 1 -( 1 -methyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yl1 -phenyl } -N- hvdroxy-acrylamide.
1H NMR (400 MHz, DMSO- ): δ 1.49 (m, IH), 1.78 (m, IH), 2.06 (m, 2H), 2.24 (m, 4H), 2.72 (m, 2H), 2.95 (m, IH), 4.42 (m, IH), 6.60 (d, IH), 7.08 (m, IH), 7.26 (m, IH), 7.58 (d, IH), 7.65 (m, 2H), 7.73 (d, IH), 7.80 (m, IH), 7.88 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 395.1 (M + H)+. COMPOUND 94.
Figure imgf000140_0001
(R)-3 - { 3- r5-Fluoro- 1 -( 1 -methyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yl] -phenyl } -N- hydroxy-acrylamide.
1HNMR (400 MHz, OMSO-d6): δ 1.49 (m, IH), 1.77 (m, IH), 2.03 (m, 2H), 2.24 (m, 4H), 2.72 (m, 2H), 2.99 (m, IH), 4.43 (m, IH), 6.59 (d, IH), 7.14 (m, IH), 7.50 (m, IH), 7.57 (d, IH), 7.64 (m, 2H), 7.78 (m, IH), 7.85 (s, IH), 7.91 (m, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 395.1 (M + H)+.
COMPOUND 95.
Figure imgf000140_0002
(R)-3 - { 5- r 1 -( 1 -Ethyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yll -2-fluoro-phenyl } -N- hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- ): δ 0.96 (t, 3H), 1.51 (m, IH), 1.79 (m, IH), 2.04 (m, 2H), 2.34 (m, 3H), 2.75 (m, IH), 2.86 (m, IH), 3.03 (m, IH), 4.37 (m, IH), 6.70 (d, IH), 7.26 (m, 2H), 7.48 (m, IH), 7.58 (d, IH), 7.71 (m, 2H), 7.87 (m, IH), 7.95 (d, IH), 9.15 (s, IH), 10.90 (s, IH). ESI-MS: m/z 409.1 (M + H)+. COMPOUND 96.
Figure imgf000141_0001
(R)-3- ( 3-[ 1 -( 1 -Ethyl-piperidin-3-vD- lH-benzoimidazol-2-yll-4-fluoro-phenyl 1 -N- hvdroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 1.01 (t, 3H), 1.48 (m, IH), 1.79 (m, IH), 1.99 (m, 2H), 2.32 (m, 3H), 2.71 (m, IH), 2.85 (m, IH), 2.98 (m, IH), 4.12 (m, IH), 6.53 (d, IH), 7.30 (m, 2H), 7.53 (m, 2H), 7.72 (d, IH), 7.90 (m, 3H), 9.10 (s, IH), 10.77 (s, IH). ESI-MS: m/z 409.1 (M + H)+.
COMPOUND 97.
Figure imgf000141_0002
(R)-3-{4-FluoiO-3-r5-fluoro-l-(l-methyl-piperidin-3-yl)-lH-benzoimidazol-2-yl1- phenyl } -N-hydroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 1.47 (m, IH), 1.75 (m, IH), 1.90 (m, IH), 2.05 (m, IH), 2.19 (m, 4H), 2.69 (m, 2H), 2.87 (m, IH), 4.11 (m, IH), 6.53 (d, IH), 7.19 (m, IH), 7.54 (m, 3H), 7.87 (m, 2H), 7.96 (m, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 413.1 (M + H)+. COMPOUND 98.
Figure imgf000142_0001
(R)-3-(3- ( 1-ri -(2.2-Difluoro-efhyl)-ρiperidin-3-yl1-4-fluoro- lH-benzoimidazol-2-yl} - phenyl)-N-hvdroxy-acrylamide.
1H ΝMR (400 MHz, DMSO- 6): δ 1.49 (m, IH), 1.78 (m, IH), 2.05 (m, IH), 2.32 (m, 2H), 2.83 (m, 3H), 3.08 (m, 2H), 4.40 (m, IH), 6.13 (m, IH), 6.60 (d, IH), 7.08 (m, IH), 7.26 (m, IH), 7.58 (d, IH), 7.66 (m, 2H), 7.75 (d, IH), 7.81 (m, IH), 7.87 (s, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 445.1 (M + H)+.
COMPOUND 99.
Figure imgf000142_0002
(±)-3-{3-[l-(2-Dimethylamino-2-ρhenyl-ethyl)-lH-benzoimidazol-2-yl1-phenyl}-N- hydroxy-acrylamide.
1H ΝMR (400MHz, DMSO-^6): δ 1.92 (s, 6H), 3.46 (t, IH), 4.52 (q, 1), 5.02 (q, IH), 6.55 (d, IH), 6.83 (d, 2H), 7.05-7.28 (band, 5H), 7.52-7.72 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 427.2 (M + H)+. COMPOUND 100.
Figure imgf000143_0001
(±)-3-{3-ri-(l-Benzyl-piperidin-3-yl)-lH-benzoimidazol-2-yl1-phenyll-N-hydroxy- acrylamide.
1HΝMR (400MHz, DMSO- 6): δ 1.50 (m, IH), 1.78 (d, IH), 2.03 (br s, 2H), 2.14 (m, IH), 2.33 (m, IH), 2.82 (m, 2H), 2.95 (m, IH), 3.55 (m, IH), 4.40 (m, IH), 6.60 (d, IH), 7.23- 7.34 (band, 7H), 7.53-7.69 (band, 4H), 7.75-7.87 (band, 3H), 9.11 (s, IH), 10.80 (s, IH). ESI- MS: m/z 453.2 (M + H)+.
COMPOUND 101.
Figure imgf000143_0002
(±)-N-Hvdroxy-3- { 3 - \ 1 -( 1 -propyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yll -phenyl ) - acrylamide.
1H ΝMR (400MHz, OM$0-d6): δ 0.79 (t, 3H), 1.45 (m, 3H), 1.78 (d, IH), 2.04 (m, 2H), 2.29 (m, 3H), 2.78 (m, 2H), 3.02 (m, IH), 4.40 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.55- 7.71 (band, 4H), 7.78 (m, IH), 7.86 (m, 2H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 405.2 (M + H)+. COMPOUND 102.
Figure imgf000144_0001
(±)-N-Hydroxy-3- 13- [ 1 -( 1 -isobutyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yl1 -phenyl I - acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 0.82 (d d, 6H), 1.46 (m, IH), 1.77 (m, 2H), 1.97 (m, IH), 2.09 (m, 3H), 2.30 (m, IH), 2.73 (m, 2H), 3.04 (d, IH), 4.38 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.55-7.89 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 419.2 (M + H)+.
COMPOUND 103.
Figure imgf000144_0002
(±)-3-{3-ri-(l-Allyl-piperidin-3-yl)-lH-benζoirnidaζol-2-yl1-phenyl}-N-hydroxy- aciylarnide.
1HΝMR (400MHz, OMSO-d6): δ 1.49 (in, IH), 1.79 (d, IH), 2.09 (rn, 2H), 2.30 (rn, IH), 2.80 (rn, 2H), 2.97 (rn, 3H), 4.40 (rn, IH), 5.15 (M, 2H), 5.79 (rn, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.56-7.71 (band, 4H), 7.82 (m, 3H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 403.2 (M + H)+. COMPOUND 104.
Figure imgf000145_0001
±)-3-{3-[l- acrylamide.
1H ΝMR (400MHz, CDC13- ): δ 1.60 (m, IH), 1.94 (m, 2H), 2.29 (s, 3H), 2.73 (m, IH), 3.28 (t, IH), 3.60 (t, IH), 3.99 (d, IH), 4.57 (m, IH), 5.39 (d, IH), 6.88 (d, IH), 7.34 (m, 2H), 7.51-7.69 (band, 5H), 7.89 (m, 2H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: /z 405.1 (M + H)+.
COMPOUND 105.
Figure imgf000145_0002
(S)-N-Hvdroxy-3-{3-ri-(l-methyl-piperidin-3-yl)-lH-benzoimidazol-2-vn-phenyl}- acrylamide.
1HΝMR (400MHz, DMSO- 6): δ 1.50 (m, IH), 1.78 (d, IH), 2.02 (m, IH), 2.24 (m, 5H), 2.74 (br s, 2H), 2.94 (br s, IH), 4.42 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.56-7.88 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 377.1 (M + H)+.
COMPOUND 106.
Figure imgf000146_0001
(S)-3-{3-ri-(l-Ethyl-piperidin-3-yl)-lH-benzoimidazol-2-yn-phenyl}-N-hydroxy- acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 0.96 (t, 3H), 1.50 (m, IH), 1.78 (d, IH), 2.05 (m, 2H), 2.33 (m, 3H), 2.75 (t, IH), 2.84 (d, IH), 3.02 (m, IH), 4.40 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.51-7.88 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 391.2 (M + H)+.
COMPOUND 107.
Figure imgf000146_0002
(R)-N-Hydroxy-3- { 3 - \ 1 -( 1 -methyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yll -phenyl } - acrylamide. lB ΝMR (400MHz, DMSO- 6): δ 1.50 (m, IH), 1.78 (d, IH), 2.02 (m, 2H), 2.21-2.33 (m, 4H), 2.75 (m, 2H), 2.94 (m, IH), 4.42 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.56-7.88 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 377.1 (M + H)+.
COMPOUND 108.
Figure imgf000146_0003
(R)-3-{3-ri-(l-Ethyl-piperidin-3-yl)-lH-benzoimidazol-2-yn-phenyl}-N-hydroxy- acrylamide. 1H NMR (400MHz, DMSO-J6): δ 0.96 (t, 3H), 1.50 (m, IH), 1.78 (d, IH), 2.05 (m, 2H), 2.33 (m, 3H), 2.75 (t, IH), 2.84 (d, IH), 3.02 (d, IH), 4.40 (m, IH), 6.59 (d, IH), 7.26 (m, 2H), 7.55-7.88 (band, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 391.2 (M + H)+.
COMPOUND 109.
Figure imgf000147_0001
(R)-N-Hydroxy-3-(3-r3-(l-isopropyl-piperidin-3-yl)-3H-irmdaζor4,5-blpyridin-2-yl1- phenyl I -acrylamide.
1H ΝMR (400MHz, DMSO- 6): δ 0.95 (br s, 6H), 1.43 (m, IH), 1.78 (m, IH), 1.99 (m, IH), 2.19 (m, IH), 2.75 (m, 3H), 2.92 (m, IH), 3.25 (m, IH), 4.35 (m, IH), 6.58 (d, IH), 7.31 (m, IH), 7.57 (d, IH), 7.65 (m, 2H), 7.81 (d, IH), 7.89 (s, IH), 8.11 (d, IH), 8.41 (d, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 406.2 (M + H)+.
COMPOUND 110.
Figure imgf000147_0002
(S)-N-Hydroxy-3-r3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1H ΝMR (400MHz, OMSO-d6): δ 1.70 (m, IH), 1.90 (m, IH), 2.20 (m, IH), 2.49 (m, 3H), 3.30 (m, 2H), 4.70 (m, IH), 6.60 (d, IH), 7.31 (m, 2H), 7.51-7.90 (m, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 363.2 (M + H)+. COMPOUND 111.
Figure imgf000148_0001
(R)-N-Hy(koxy-3-r3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl1-acrylamide.
1HΝMR (400MHz, DMSO- 6): δ 1.39 (m, IH), 1.70 (m, IH), 2.00 (m, IH), 2.45 (m, 3H), 2.80 (m, IH), 3.05 (m, IH), 4.30 (m, IH), 6.60 (d, IH), 7.31 (m, 2H), 7.51-7.90 (m, 7H), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 363.2 (M + H)+.
COMPOUND 112.
Figure imgf000148_0002
(±)-N-Hydroxy-3- { 3-[ 1 -( 1 -propyl-piperidin-3-yl)- 1 H-benζoimidaζol-2-yl1 -phenyl } - propionamide.
1H ΝMR (400MHz, DMSO-^6): δ 0.98 (t, 3H), 1.71 (m, 3H), 2.01 (m, 2H), 2.31 (m, IH), 2.97 (m, 2H), 3.20 (m, IH), 3.49 (m, IH), 3.98 (m, 2H), 4.68 (m, IH), 7.35 (m, 2H), 7.48 (d, IH), 7.54 (m, 3H), 7.74 (m, IH), 8.08 (m, IH), 9.11 (s, IH), 10.80 (s, IH). ESI-MS: m/z 407.2 (M + H)+.
COMPOUND 113.
Figure imgf000148_0003
(±)-3-{3-[l-(l -Ethyl-piperidin-3-vD- 1 H-benzoimidazol-2-yn -phenyl } -N-hydroxy- propionamide. 1H NMR (400MHz, DMSO- 6): δ 1.29 (t, 3H), 1.75 (m, IH), 1.98 (m, 2H), 2.31 (m, 2H), 2.48 (m, 2H), 2.95 (m, 2H), 3.20 (m, 2H), 3.42 (m, IH), 3.82 (m, IH), 3.95 (m, IH), 4.68 (m, IH), 7.35 (m, 2H), 7.48 (d, IH), 7.54 (m, 3H), 7.74 (m, IH), 8.08 (m, IH), 9.68 (s, IH), 10.80 (s, IH). ESI-MS: m/z 393.2 (M + H)+.
COMPOUND 114.
Figure imgf000149_0001
N-Hydroxy-3-[3-(l-phenethyl-lH-benzoimidazol-2-yl)-phenvn-propionamide.
1HΝMR (400MHz, OMSO-d6): δ 2.28 (t, 2H), 2.97 (t, 2H), 3.05 (t, 2H), 4.62 (t, 2H), 6.85 (m, 2H), 7.15 (m, 3H), 7.31 (s, IH), 7.38 (m, IH), 7.48 (m, 4H), 7.77 (m, IH), 7.96 (d, IH), 8.70 (s, IH), 10.45 (s, IH). ESI-MS: m/z 386.2 (M + H)+.
COMPOUND 115.
Figure imgf000149_0002
N-Hvdroxy-3-(3-[3-(l-methyl-piperidin-3-yl)-3H-imidazor4.5-b1pyridin-2-yn- phenyl } -acrylamide.
1H NMR (400MHz, OMSO-d6): δ 1.49 (m, IH), 1.76 (m, IH), 1.95 (m, 2H), 2.20
(s, 3H), 2.70 (m, 2H), 2.93 (m, IH), 3.09 (m, IH), 4.41 (m, IH), 6.58 (d, IH), 7.31 (m,
IH), 7.56-7.60 (d, IH), 7.66 (m, 2H), 7.81 (d, IH), 7.90 (s, IH), 8.11 (d, IH), 8.41 (d, IH).
ESI-MS: m/z 377.19 (M + H)+. COMPOUND 116.
Figure imgf000150_0001
(R)-3-(3-[3-(l-Ethyl-piperidin-3-yl)-3H-imidazor4.5-blρyridin-2-yll-phenyll-N- hydroxy-acrylamide.
ESI-MS: m/z 392.2 (M + H)+. COMPOUND 117.
Figure imgf000150_0002
N-Hydroxy-3-r5-(l-phenethyl-lH-benzoimidazol-2-yl)-pyridin-3-yl1-acrylajrιide.
1H NMR (400MHz, OMSO-d6): δ 2.97 (t, 2H), 4.61 (t, 2H), 6.65 (m, 3H), 7.02 (m, 3H), 7.32 (d t, 2H), 7.52 (d, IH), 7.80 (m, 3H), 8.61 (s, IH), 8.83 (s, IH). ESI-MS: m/z 385.2 (M + H)+.
COMPOUND 118.
Figure imgf000150_0003
(R)-N-HydiOxy-3-{5-[l-(l-methyl-piperidin-3-yl)-lH-benzoimidazol-2-yl1- pyridin-3-yl } -acrylamide.
1H NMR (400MHz, OMSO-d6): δ 1.56 (m, IH), 1.77 (m, IH), 2.05 (m, 2H), 2.27 (m, 4H), 2.75 (m, 2H), 3.02 (m, IH), 4.35 (m, IH), 6.70 (d, IH), 7.31 (m, 2H), 7.61 (d, IH), 7.72 (m, IH), 7.90 (d, IH), 8.29 (s, IH), 8.83 (s, IH), 8.98 (s, IH). ESI-MS: m/z 378.2 (M + H)+. COMPOUND 119.
Figure imgf000151_0001
(R)-N-Hvdroxy-3-(5-r3-(l-methyl-piperidin-3-yl)-3H-imidazor4.5-blpyridin-2-yll- pyridin-3-yl ) -acrylamide.
1H NMR (400MHz, OMSO-d6): δ 1.51 (m, IH), 1.92 (m, 3H), 2.15 (s, 3H), 2.71 (m, 2H), 3.02 (m, 2H), 4.33 (m, IH), 6.78 (br d, IH), 7.33 (m, 2H), 8.13 (d, IH), 8.27 (s, IH), 8.42 (d, IH), 8.79 (s, IH), 8.95 (s, IH). ESI-MS: m/z 379.2 (M + H)+.
[0337] As used herein the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Standard single-letter or thee-letter abbreyiations are generally used to designate amino acid residues, which are assumed to be in the L-configuration unless oti erwise noted. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. Specifically, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams);
L (liters); mL (milliliters); μL (microliters); psi (pounds per square inch);
M (molar); mM (millimolar); i.v. (intravenous); Hz (Hertz);
MHz (megahertz); mol (moles); mmol (millimoles); RT (ambient temperature); min (minutes) ;h (hours); mp (melting point); TLC (thin layer chromatography);
Tr (retention time); RP (reverse phase);
MeOH (methanol); i-PrOH (isopropanol);
TEA (triethylamine); TFA (trifluoroacetic acid); TFAA (trifluoroacetic anhydride); THF (tetrahydrofuran);
DMSO (dimethylsulf oxide); EtOAc (ethyl acetate);
DME ( 1 ,2-dimethoxyethane) ; DCM (dichloromethane) ;
DCE (dichloroethane); DMF (N,N-dimethylformamide);
DMPU (N,N'-dimethylpropyleneurea); CDI (1,1-carbonyldiimidazole);
IBCF (isobutyl chloroformate); HO Ac (acetic acid);
HOSu (N-hydroxysuccinimide); HOBT (1-hydroxybenzotriazole);
Et2O (diethyl ether); EDCI (ethylcarbodiimide hydrochloride);
BOC (tert-butyloxycarbonyl); FMOC (9-fluorenylmethoxycarbonyl);
DCC (dicyclohexylcarbodiimide); CBZ (benzyloxycarbonyl);
Ac (acetyl); atm (atmosphere);
TMSE (2-(trimethylsilyl)ethyl); TMS (trimethylsilyl);
TIPS (triisopropylsilyl); TBS (t-butyldimethylsilyl);
DMAP (4-dimethylaminopyridine); Me (methyl);
OMe (methoxy); Et (ethyl);
Et (ethyl); tBu (tert-butyl);
HPLC (high pressure liquid chromatography);
BOP (bis(2-oxo-3-oxazolidinyl)phosphinic chloride);
TBAF (tetra-n-butylammonium fluoride); mCPBA (meta-chloroperbenzoic acid. [0338] All references to ether or Et2O are to diethyl ether; brine refers to a saturated aqueous solution of NaCI. Unless otherwise indicated, all temperatures are expressed in °C (degrees Centigrade). All reactions conducted under an inert atmosphere at RT unless otherwise noted.
[0339] !H NMR spectra were recorded on a Bruker Avance 400. Chemical shifts are expressed in parts per million (ppm). Coupling constants are in units of hertz (Hz). Splitting patterns describe apparent multiplicities and are designated as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad).
[0340] Low-resolution mass spectra (MS) and compound purity data were acquired on a Waters ZQ LC/MS single quadrupole system equipped with electrospray ionization (ESI) source, UV detector (220 and 254 nm), and evaporative light scattering detector (ELSD). Thin-layer chromatography was performed on 0.25 mm E. Merck silica gel plates (60F-254), visualized with UV light, 5% ethanolic phosphomolybdic acid, Ninhydrin or p-anisaldehyde solution. Flash column chromatography was performed on silica gel (230-400 mesh, Merck). [0341] It will be apparent to those skilled in the art that various modifications and variations can be made to the compounds, compositions, kits, and methods of the present invention without departing from the spirit or scope of the invention. Thus, it is intended that the present invention cover the modifications and variations of this invention provided they come within the scope of the appended claims and their equivalents.

Claims

What is claimed is:
1. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000154_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen, and each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which Rι2 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
2. A compound according to claim 1 wherein the compound comprises the formula
Figure imgf000155_0001
3. A compound of claim 1, wherein Q is a substituted or unsubstituted phenyl ring.
4. A compound of claim 1 , wherein Q is a substituted or unsubstituted heteroaryl.
5. A compound of claim 1 , wherein Q is a substituted or unsubstituted heteroaryl selected from die group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phfhalazine, benthiazole, and triazine.
6. A compound according to claim 1, wherein at least one X in the six membered ring is a substituted carbon atom.
7. A compound according to claim 1, wherein at least one of the X substituents in the six membered ring is -CF.
8. A compound according to claim 1 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
9. A compound according to claim 1 , wherein M is selected from the group consisting of:
Figure imgf000156_0001
10. A compound according to claim 1, wherein M comprises a hydroxamic acid moiety.
11. A compound according to claim 1, wherein -Q-L-M is
Figure imgf000156_0002
12. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000157_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR1 ;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R2, R3, R4 and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each Rι2 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
13. A compound according to claim 12, wherein the compound comprises the formula
Figure imgf000158_0001
14. A compound according to claim 12, wherein at least one of R2, R3, R4, or R5 is fluorine.
15. A compound according to claim 12, wherein M comprises a member selected from d e group consisting of trifluoroacetyl (-C(O)-CF3)s -NH-P(O)OH-CH3, sulfonamides (-SO NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
16. A compound according to claim 12, wherein M is selected from the group consisting of:
Figure imgf000158_0002
17. A compound according to claim 12, wherein M comprises a hydroxamic acid moiety.
18. A compound according to claim 12, wherein -Q-L-M is
Figure imgf000159_0001
19. A compound comprising a formula selected from the group consisting of:
Figure imgf000159_0002
wherein each X is independently selected from the group consisting of CR1 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen; R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
20. A compound according to claim 19, wherein the compound comprises the formula
Figure imgf000160_0001
21. A compound according to claim 19, wherein at least one of R6, R7, R8, and R is fluorine.
22. A compound according to claim 19, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
23. A compound according to claim 19, wherein M is selected from the group consisting of: o o o ^X\ o r^i N"0H AB.o„ -i-- H ΛΛJ ΛN J ΛNH!
NH2 H d)H
Figure imgf000161_0001
24. A compound according to claim 19, wherein M comprises a hydroxamic acid moiety.
25. A compound according to claim 19, wherein -L-M is
Figure imgf000161_0002
26. A compound comprising a formula selected from the group consisting of:
Figure imgf000162_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from d e group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R6, R7, R8) and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
27. A compound according to claim 26, wherein the compound comprises the formula
Figure imgf000163_0001
28. A compound according to claim 26 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(0)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH ), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
29. A compound according to claim 26, wherein M is selected from the group consisting of:
Figure imgf000163_0002
30. A compound according to claim 26, wherein M comprises a hydroxamic acid moiety.
31. A compound according to claim 26, wherein -L-M is
Figure imgf000164_0001
32. A compound according to claim 26, wherein at least one of R6, R , R8, and R9 is fluorine.
33. A compound according to claim 26, wherein at least one X in the six membered ring is a substituted carbon atom.
34. A compound according to claim 26, wherein at least one of the X substituents in the six membered ring is -CF.
35. A compound comprising a formula selected from the group consisting of:
Figure imgf000164_0002
Figure imgf000165_0001
wherein each X is independently selected from the group consisting of CRι2 and N; each Y is independently selected from the group consisting of O, S and NR12;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
R2, R3, R4, R5, R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
36. A compound according to claim 35, wherein the compound comprises the formula
Figure imgf000166_0001
37. A compound according to claim 35, wherein at least one of R2, R3, R^., or R5 is fluorine.
38. A compound according to claim 35, wherein at least one of R6, R7, R8, and R9 is fluorine.
39. A compound according to claim 35, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R1 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
40. A compound according to claim 35, wherein M is selected from the group consisting of:
Figure imgf000166_0002
41. A compound according to claim 35, wherein M comprises a hydroxamic acid moiety.
42. A compound according to claim 35, wherein -L-M is
Figure imgf000167_0001
43. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000167_0002
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and thio in the case where the ring atom to which Ri is bound is nitrogen;
Rio and Rn are taken together to form a substituted or unsubstituted aromatic ring; each Rπ is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen; Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
44. A compound according to claim 43 , wherein the substituted or unsubstituted aromatic ring formed when R10 and Rn are taken together is selected from the group consisting of substituted or unsubstituted aryl and heteroaryl.
45. A compound according to claim 43 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
46. A compound according to claim 43, wherein M is selected from the group consisting of:
Figure imgf000168_0001
47. A compound according to claim 43, wherein M comprises a hydroxamic acid moiety.
48. A compound according to claim 43, wherein -L-M is
Figure imgf000169_0001
49. A compound comprising the formula
Figure imgf000169_0002
wherein
Ri is selected from the group consisting of hydrogen, halo, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Ri is not halo, cyano, nitro and ti io in the case where die ring atom to which Ri is bound is nitrogen;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
50. A compound according to claim 49, wherein at least one of R2, R3, R4, or R5 is fluorine.
51. A compound according to claim 49, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein Ri3 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
52. A compound according to claim 49, wherein M is selected from the group consisting of:
Figure imgf000170_0001
53. A compound according to claim 49, wherein M comprises a hydiOxamic acid moiety.
54. A compound according to claim 49, wherein -L-M is
^AA.^OH
H
55. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000171_0001
Ri is selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
56. A compound according to claim 55, wherein at least one of R2, R3, j, or R5 is fluorine.
57. A compound according to claim 55, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-Rι3 wherein Rι3 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
58. A compound according to claim 55, wherein M is selected from the group consisting of:
Figure imgf000172_0001
59. A compound according to claim 55, wherein M comprises a hydroxamic acid moiety.
60. A compound according to claim 55, wherein -L-M is
Figure imgf000172_0002
61. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000172_0003
Ri is selected from the group consisting of (Ci )alkyl, phenyl, l-piperidin-4-ylmethyl, 2-morpholi-4-yl-ethyl, 2-halo-phenyl, 2-halo-phen(C1-4)alkyl, 3-halo-phen(C1-4)alkyl, 2-CF3O- phen(C1-4)alkyl, 3-CF3O-phen(Ci )alkyl, 3-halo-phenyl, 4-halo-phenyl, 2-methoxy-phenyl, 3- methoxy-phenyl, 4-methoxy-phenyl, 4-phenoxy-phenyl, 4-benzyloxyphenyl, 4-pyrazol-l-yl- benzyl, l-p-tolyl-ethyl, pyrrolidin-3-yl, l-(C14)alkyl-pyrrolidin-2-yl, l-(Ci4)alkyl-pyrrolidin- 2-yl; 2-di(Ci-4)alkylamino-ethyl, 2-di(Ci4)alkylamino-l -methyl-ethyl, 2-di(C14)alkylamino- ethyl, 2-hydroxy-2-phenyl-ethyl, 2-pyridin-2-yl-ethyl, 2-pyridin-3-yl-ethyl, 2-pyridin-4-yl- ethyl, 2-(lH-indol-3-yl)-ethyl, 3-indolyl(Ci4)alkyl, l-indan-2-yl, R-α-(HOCH2)-phen(Ci. 4)alkyl, S-α-(HOCH2)-phen(C1-4)alkyl, S-β-(HOCH2)-phen(C14)alkyl, R-β-(CH3)-phen(Cι- 4)alkyl, 6-propylsulfanyl, trαns-4-hydroxy-cyclohexyl, l-aza-bicyclo[2.2.2]oct-2-yl, l-(Ci. 4)alkyl-piperidin-3-yl, l-(2,2-difluoro-ethyl)-piperidin-3-yl, (2-di(C1-4)alkylamino-2-phenyl- ethyl), l-benzyl-piperidin-3-yl, l-allyl-piperidin-3-yl, l-acetyl-piperidin-3-yl, piperidin-3-yl, and ρhen(Cι-4)alkyl;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro;
M is selected from the group consisting of:
Figure imgf000173_0001
and L is E, Z or mixtures of E/Z -CH2=CH -.
62. A compound according to claim 61 wherein at least one of R2, R3, R4, or R5 is fluorine.
63. A compound according to claim 61 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein Rι3 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
64. A compound according to claim 61, wherein M is selected from the group consisting of:
Figure imgf000174_0001
65. A compound according to claim 61, wherein M comprises a hydroxamic acid moiety.
66. A compound according to claim 61, wherein -L-M is
Figure imgf000174_0002
67. A compound selected from the group consisting of:
3- [3-( lH-Benzoimidazol-2-yl)-phenyl] -N-hydroxy-acrylamide;
N-Hydroxy-3-[3-(l-methyl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
N-Hydroxy-3-[3-(l-isopropyl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
(±)-3 - { 3- [ 1 -( 1 -Ethyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
N-Hydroxy-3- { 3-[ 1 -( 1 -methyl-piperidin-4-yl)- lH-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3-[3-(l-piperidin-4-ylmed yl-lH-benzomιidazol-2-yl)-phenyl]-acιylarnide;
N-Hydroxy-3 - { 3 - [ 1 - (2-morpholin-4-yl-ethyl) - 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3-[3-(l-phenyl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
3-{3-[l-(4-Chloro-phenyl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy-acrylamide;
N-Hydroxy-3-{3-[l-(4-me oxy-phenyl)-lH-benzoiniidazol-2-yl]-phenyl}-aciylamide;
N-Hydroxy-3-{3-[l-(4-phenoxy-phenyl)-lH-benzoimidazol-2-yl]-phenyl}-acrylamide; 3 - { 3 - [ 1 -(4-B enzyloxy-phenyl) - 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
3-[3-(l-Benzyl-lH-benzoimidazol-2-yl)-phenyl]-N-hydroxy-acrylamide;
3 - { 3 - [ 1 -(2-Chloro-benzyl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy-acrylamide;
3-{3-[l-(3 -Chloro-benzyl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy-acrylamide;
3 - { 3 - [ 1 -(4-Chloro-benzyl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy-acrylamide;
N-Hydroxy-3- { 3-[ 1 -(2-trifluoromethoxy-benzyl)- lH-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3-{3-[l-(3-trifluoromethoxy-benzyl)-lH-benzoimidazol-2-yl]-phenyl}- acrylamide;
N-Hydroxy-3- { 3- [ 1 -(4-pyrazol- 1 -yl-benzyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
(R)-N-Hydroxy-3-{3-[l-(l-phenyl-emyl)-lH-benzoiιmdazol-2-yl]-phenyl}-ac]ylamide;
(S)-N-Hydroxy-3-{3-[l-(l-phenyl-emyl)-lH-benzoimidazol-2-yl]-phenyl}-acιylamide;
(R)-N-Hydroxy-3- { 3-[ 1 -( 1-p-tolyl-eti yl)- lH-benzoimidazol-2-yl]-phenyl }-acrylamide;
(R)-3-(3- { 1 - [ 1 -(4-Fluoro-phenyl)-ethyl] - lH-benzoimidazol-2-yl } -phenyl)-N-hydroxy- acrylamide;
N-Hydroxy-3 - [3 -( 1 -phenethyl- 1 H-benzoimidazol-2-yl)-phenyl] -acrylamide;
3-(3-{ l-[2-(3-Fluoro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide;
3-(3-{ l-[2-(4-Fluoro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide;
Ν-Hydroxy-3-(3-{ l-[2-(2-methoxy-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)- acrylamide;
N-Hydroxy-3-(3-{ l-[2-(3-methoxy-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)- acrylamide;
N-Hydroxy-3-(3-{ l-[2-(4-methoxy-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)- acrylamide;
3-(3-{ l-[2-(2-Chloro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide;
3-(3-{ l-[2-(3-Chloro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-ρhenyl)-N-hydroxy- acrylamide;
3-(3-{ l-[2-(4-Chloro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-N-hydroxy- acrylamide;
(±)-3-(3-{ l-[2-(4-Fluoro-phenyl)-l-metiιyl-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-N- hydroxy-acrylamide;
(R)-N-Hydroxy-3 - { 3 - [ 1 -(2-phenyl-propyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
(S)-N-Hydroxy-3-{3-[l-(2-phenyl-propyl)-lH-benzoimidazol-2-yl]-phenyl}- acrylamide;
(S)-N-Hyαroxy-3-{3-[l-(l-hydroxymethyl-2-phenylethyl)-lH-benzoimidazol-2-yl]- phenyl } -acrylamide;
(R)-N-Hydroxy-3- {3-[l-(l -hydroxymethyl-2-phenylethyl)- lH-benzoimidazol-2-yl]- phenyl } -acrylamide;
(R)-N-Hydroxy-3- { 3-[ l-(2-hydroxy-2-phenyl-emyl)- lH-benzoimidazol-2-yl]-phenyl } - acrylamide;
N-Hydroxy-3- { 3 - [ 1 -(2-pyridin-2-yl-ethyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3- { 3-[ l-(2-pyridin-3-yl-ethyl)-lH-benzoimidazol-2-yl]-phenyl } - acrylamide;
N-Hydroxy-3 - { 3 - [ 1 -(2-pyridin-4-yl-ethyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3-(3- { 1 -[2-( lH-indol-3-yl)-ethyl]- lH-benzoimidazol-2-yl }-phenyl)- acrylamide;
N-Hydroxy-3-[3-( 1 -indan-2-yl- lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
N-Hydroxy-3- { 3-[ 1 -(3-phenyl-propyl)- lH-benzoimidazol-2-yl]-phenyl } -acrylamide;
3-[3-(5-Fluoro-lH-benzoimidazol-2-yl)-phenyl]-N-hydroxy-acrylamide;
N-Hydroxy-3-[3-(6-propylsulfanyl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
N-Hydroxy-3-[3-(7-methyl-lH-benzoinιidazol-2-yl)-phenyl]-aci7lamide;
2-[3-(2-Hydroxycarbamoyl-vinyl)-phenyl]-3H-benzoimidazole-5-carboxylic acid methyl ester;
3-[3-(6,7-Difluoro-lH-benzoimidazol-2-yl)-phenyl]-N-hydroxy-acrylamide; 3- { 3 - [ 1 -(4-Methoxy-phenyl)- 1 H-benzoimidazol-2-yl] -phenyl } -acrylic acid;
3- { 3- [ 1 -(4-Benzyloxy-phenyl)- lH-benzoimidazol-2-yl] -phenyl } -acrylic acid;
3 - { 3- [ 1 -(4-Chloro-ρhenyl)- 1 H-benzoimidazol-2-yl] -phenyl } -acrylic acid;
(R)-3-(3-{ l-[l-(4-Fluoro-phenyl)-ethyl]-lH-benzoimidazol-2-yl}-phenyl)-acrylic acid;
(R)-3-{3-[l-(l-p-Tolyl-ethyl)-lH-benzoimidazol-2-yl]-phenyl}-acrylic acid;
3- { 3-[l-(4-Phenoxy-phenyl)- lH-benzoimidazol-2-yl] -phenyl } -acrylic acid;
N-(2-Amino-phenyl)-3- { 3-[ 1 -(4-methoxy-phenyl)- lH-benzoimidazol-2-yl]-phenyl } - acrylamide;
N-(2-Amino-phenyl)-3-{3-[l-(4-benzyloxy-phenyl)-lH-benzoimidazol-2-yl]-phenyl}- acrylamide;
N-(2- Amino-phenyl)-3 - { 3- [ 1 -(4-chloro-phenyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
(R)-N-(2-Amino-phenyl)-3-(3-{ l-[l-(4-fluoro-phenyl)-emyl]-lH-benzoimidazol-2-yl}- phenyl)-acrylamide;
(R)-N-(2- Amino-phenyl)-3 -{3-[l-(l -p-tolyl-etiiyi)- lH-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-(2-Amino-phenyl)-3-{3-[l-(4-phenoxy-phenyl)-lH-ben∑oinιidazol-2-yl]-phenyl}- acrylamide;
N-Hydroxy-3-[4-(l-phenethyl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
N-(2-Amino-phenyl)-3-[4-(l-phened yl-lH-benzoimidazol-2-yl)-phenyl]-acιylamide;
3 - [4-( 1 -Phenethyl- 1 H-benzoimidazol-2-yl)-phenyl] -acrylic acid;
N-Hydroxy-3 - { 3- [ 1 -(2-piperidin- 1 -yl-ethyl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
N-Hydroxy-3 - { 3- [ 1 -(trαn^-4-hydroxy-cyclohexyl)- lH-benzoimidazol-2-yl] -phenyl } - acrylamide;
3-[3-(l-tert-Butyl-lH-benzoiι dazol-2-yl)-phenyl]-N-hydroxy-acιylamide;
(±)-3-[3-(l-Cyclohexyl-lH-benzoimidazol-2-yl)-phenyl]-N-hydroxy-acrylamide;
3- { 2-[3-(2-Hydroxycarbamoyl-vinyl)-phenyl] -benzoimidazol- 1 -yl } -piperidine- 1 - carboxylic acid tert-butyl ester;
(±)-N-Hydroxy-3-[3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
3- { 3-[l-(2-Diethylamino-ethyl)- lH-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
(±)-N-Hydroxy-3 - { 3- [ 1 -( 1 -methyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide;
(±)-N-Hydroxy-3- { 3-[ 1 -(l-isopropyl-piperidin-3-yl)- lH-benzoimidazol-2-yl]-phenyl } - acrylamide;
(±)-N-Hydroxy-3-(3-{ l-[l-(2-hyα oxy-ethyl)-piperidin-3-yl]-lH-benzoimidazol-2-yl}- phenyl)-acrylamide;
(±)-3-{3-[l-(l -Ethyl-pyrrolidin-2-ylmethyl) - 1 H-benzoimidazol-2-yl] -phenyl } -N- hydroxy-acrylamide;
(±)-3-{3-[l-(l-Ethyl-pyrrolidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy- acrylamide;
(±)-3 - { 3 - [ 1 -(2-Dimethylamino-ethyl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
(±)-3 - { 3 - [ 1 -(2-Dimethylamino- 1 -methyl-ethyl)- 1 H-benzoimidazol-2-yl] -phenyl } -N- hydroxy-acrylamide;
(±)-3-{3-[l-(2-Diisopropylamino-emyl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy- acrylamide;
(±)-3-{3-[l-(l-Aza-bicyclo[2.2.2]oct-2-yl)-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-6,7-difluoiO-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[l-(l-Etuyl-piperidin-3-yl)-5,6-difluoro-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide-trifluoroacetic acid;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-4,6-difluoro-lH-benzoimidazol-2-yl]-ρhenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-5,6,7-trifluoro-lH-benzoimidazol-2-yl]-phenyl}- N-hydroxy-acrylamide;
(R)-3- { 3-[ 1 -( l-Ethyl-piperidin-3-yl)-4,6,7-trifluoro- lH-benzoimidazol-2-yl] -phenyl } - N-hydroxy-acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-4,5,6,7-tetrafluoro-lH-benzoimidazol-2-yl]- phenyl } -N-hydroxy-acrylamide; (R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-4-fluoro-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-5-fluoro-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-6-fluoro-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-N-Hydroxy-3 -{3-[l-(l -isopropyl-piperidin-3-yl)- lH-benzoimidazole-2-yl] - phenyl } -acrylamide;
(R)-3-{3-[4-Huoro-l-(l-isopropyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3-{3-[5-Huoro-l-(l-isopropyl-piperid -3-yl)-lH-berizoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(R)-3 - { 3- [4-Fluoro- 1 -( 1 -methyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yl] -phenyl } -N- hydroxy-acrylamide;
(R)-3- { 3-[5-Fluoro- 1 -( 1 -methyl-piperidin-3-yl)- lH-benzoimidazol-2-yl] -phenyl } -N- hydroxy-acrylamide;
(R)-3-{5-[l-(l-Ethyl-piperidin-3-yl)-lH-benzoirnidazol-2-yl]-2-fluoiO-phenyl}-N- hydroxy-acrylamide;
(R)-3 -{3-[l-(l -Ethyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yl] -4-fluoro-phenyl } -N- hydroxy-acrylamide;
(R)-3-{4-Fluoro-3-[5-fluoro-l-(l-metiιyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]- phenyl } -N-hydroxy-acrylamide;
(R)-3-(3-{ l-[l-(2,2-Difluoro-ethyl)-ρiρeridin-3-yl]-4-fluoro-lH-benzoimidazol-2-yl}- phenyl)-N-hydroxy-acrylamide;
(±)-3-{3-[l-(2-Dimethylamino-2-phenyl-ethyl)-lH-benzoimidazol-2-yl]-phenyl}-N- hydroxy-acrylamide;
(±)-3-{3-[l-(l -Benzyl-piperidin-3 -yl)- 1 H-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
(±)-N-Hydroxy-3- { 3 - [ 1 -( 1 -propyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yl] -phenyl } - acrylamide; (±)-N-Hydroxy-3- { 3- [ 1 -( 1 -isobutyl-piperidin-3-yl)- lH-benzoimidazol-2-yl]-phenyl } - acrylamide;
(±)-3-{3-[l-(l-Allyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy- acrylamide;
(±)-3-{3-[l-(l - Acetyl-piperidin-3-yl)- lH-benzoimidazol-2-yl] -phenyl } -N-hydroxy- acrylamide;
(S)-N-Hydroxy-3-{3-[l-(l-methyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}- acrylamide;
(S)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy- acrylamide;
(R)-N-Hydroxy-3- { 3-[ 1 -( 1 -methyl-piperidin-3-yl)- lH-benzoirnidazol-2-yl]-phenyl } - acrylamide;
(R)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-N-hydroxy- acrylamide;
(R)-N-Hydroxy-3-{3-[3-(l-isopropyl-piperidin-3-yl)-3H-imidazo[4,5-b]pyridin-2-yl]- phenyl } -acrylamide;
(S)-N-Hydroxy-3-[3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl]-acrylamide;
(R)-N-Hydroxy-3-[3-(l-piperidin-3-yl-lH-benzoimidazol-2-yl)-phenyl]-acιylamide;
(±)-N-Hydroxy-3 - { 3 - [ 1 -( 1 -propyl-piperidin-3-yl)- 1 H-benzoimidazol-2-yl] -phenyl } - propionamide;
(±)-3-{3-[l-(l-Ethyl-piperidin-3-yl)-lH-benzoimidazol-2-yl]-phenyl}-Ν-hydiOxy- propionamide;
N-Hydroxy-3- [3 -( 1 -phenethyl- 1 H-benzoimidazol-2-yl)-phenyl] -propionamide ;
Ν-Hydroxy-3-{3-[3-(l-methyl-piperidin-3-yl)-3H-imidazo[4,5-b]pyridin-2-yl]- phenyl } -acrylamide;
(R)-3-{3-[3-(l-Ethyl-piperidin-3-yl)-3H-imidazo[4,5-b]ρyridin-2-yl]-phenyl}-N- hydroxy-acrylamide;
N-Hydroxy-3-[5-(l-phenethyl-lH-benzoimidazol-2-yl)-pyridin-3-yl]-acrylamide;
(R)-N-Hydroxy-3- { 5-[ 1 -( 1 -methyl-piperidin-3-yl)- lH-benzoimidazol-2-yl] - pyridin-3-yl} -acrylamide; and
(R)-N-Hydroxy-3-{5-[3-(l-methyl-ρiρeridin-3-yl)-3H-imidazo[4,5-b]pyridin-2-yl]- ρyridin-3 -yl } -acrylamide.
68. A compound comprising a formula selected from the group consisting of:
Figure imgf000181_0001
wherein each V is independently selected from the group consisting of C(Rι2) and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso thatRι2is not halo, cyano, nitro, and thio in the case where the ring atom to which Rι is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring.
69. A compound according to claim 68, wherein the compound comprises the formula
Figure imgf000181_0002
70. A compound according to claim 68, wherein V are selected so that the ring is a unsubstituted or substituted piperdin-3-yl moiety.
71. A compound according to claim 68, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
72. A compound according to claim 68, wherein M is selected from the group consisting of:
Figure imgf000182_0001
73. A compound according to claim 68, wherein M comprises a hydroxamic acid moiety.
74. A compound according to claim 68, wherein -L-M is
Figure imgf000182_0002
75. A compound comprising the formula
Z L M
wherein
Z is selected from the group consisting of
Figure imgf000183_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso tiiat R1 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring.
76. A compound according to claim 75, wherein the compound comprises the formula
Figure imgf000183_0002
77. A compound according to claim 75, wherein V are selected so that the ring is an N- substituted piperdin-3-yl moiety.
78. A compound according to claim 75, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-Rι3 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
79. A compound according to claim 75, wherein M is selected from the group consisting of:
Figure imgf000184_0001
80. A compound according to claim 75, wherein M comprises a hydroxamic acid moiety.
81. A compound comprising a formula selected from the group consisting of:
Figure imgf000184_0002
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
82. A compound according to claim 81 , wherein the compound comprises the formula
Figure imgf000185_0001
13. A compound according to claim 81, wherein V are selected so that d e ring is an N- substituted piperdin-3-yl moiety.
84. A compound according to claim 81 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH , sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
85. A compound according to claim 81, wherein M is selected from the group consisting of:
Figure imgf000186_0001
86. A compound according to claim 81, wherein M comprises a hydroxamic acid moiety.
87. A compound according to claim 81 , wherein -L-M is
Figure imgf000186_0002
88. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000187_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2; each Rι is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
89. A compound according to claim 88 wherein the compound comprises the formula
Figure imgf000187_0002
90. A compound according to claim 88, wherein V are selected so that the ring is an N- substituted piperdin-3-yl moiety.
91. A compound of claim 88, wherein Q is a substituted or unsubstituted phenyl ring.
92. A compound of claim 88, wherein Q is a substituted or unsubstituted heteroaryl.
93. A compound of claim 88, wherein Q is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, benthiazole, and triazine.
94. A compound according to claim 88, wherein at least one X in the six membered ring is a substituted carbon atom.
95. A compound according to claim 88, wherein at least one of the X substituents in d e six membered ring is -CF.
96. A compound according to claim 88 , wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-Rι3 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
97. A compound according to claim 88, wherein M is selected from the group consisting of:
Figure imgf000189_0001
98. A compound according to claim 88, wherein M comprises a hydroxamic acid moiety.
99. A compound according to claim 88, wherein -Q-L-M is
Figure imgf000189_0002
100. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000190_0001
Figure imgf000190_0002
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR1 ; each X is independently selected from the group consisting of CR1 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R4 and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which Rι2 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring; M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
101. A compound according to claim 100, wherein the compound comprises the formula
Figure imgf000191_0001
102. A compound according to claim 100, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
103. A compound according to claim 100, wherein at least one of R2, R3, R , or R5 is fluorine.
104. A compound according to claim 100, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
105. A compound according to claim 100, wherein M is selected from the group consisting of:
Figure imgf000192_0001
106. A compound according to claim 100, wherein M comprises a hydroxamic acid moiety.
107. A compound according to claim 100, wherein -Q-L-M is
Figure imgf000192_0002
108. A compound comprising a formula selected from the group consisting of:
Figure imgf000192_0003
wherein each V is independently selected from the group consisting of C(R12)2 andNR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
109. A compound according to claim 108, wherein the compound comprises the formula
Figure imgf000193_0001
110. A compound according to claim 108, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
111. A compound according to claim 108, wherein at least one of R6, R , R8, and R9 is fluorine.
112. A compound according to claim 108, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
113. A compound according to claim 108, wherein M is selected from the group consisting of:
Figure imgf000194_0001
114. A compound according to claim 108, wherein M comprises a hydroxamic acid moiety.
115. A compound according to claim 108, wherein -L-M is
Figure imgf000194_0002
116. A compound comprising a formula selected from the group consisting of:
Figure imgf000195_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR1 ; each X is independently selected from the group consisting of CRι2 and N; each Y is independently selected from the group consisting of O, S and NR12;
R6, R , Rg, and R9 are each independently selected from d e group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
117. A compound according to claim 116, wherein the compound comprises the formula
Figure imgf000196_0001
118. A compound according to claim 116, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
119. A compound according to claim 116, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
120. A compound according to claim 116, wherein M is selected from the group consisting of:
Figure imgf000196_0002
121. A compound according to claim 116, wherein M comprises a hydroxamic acid moiety.
122. A compound according to claim 116, wherein -L-M is
Figure imgf000197_0001
123. A compound according to claim 116, wherein at least one of R6, R7, Rg, and R is fluorine.
124. A compound according to claim 116, wherein at least one X in the six membered ring is a substituted carbon atom.
125. A compound according to claim 116, wherein at least one of the X substituents in the six membered ring is -CF.
126. A compound comprising a formula selected from the group consisting of:
Figure imgf000197_0002
Figure imgf000198_0001
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R4, R5, R6, R , R8> and R are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
127. A compound according to claim 126, wherein the compound comprises the formula
Figure imgf000199_0001
128. A compound according to claim 126, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
129. A compound according to claim 126, wherein at least one of R2, R3, R4, or R5 is fluorine.
130. A compound according to claim 126, wherein at least one of R6, R7, R8, and R9 is fluorine.
131. A compound according to claim 126, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3)s -NH-P(O)OH-CH3s sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
132. A compound according to claim 126, wherein M is selected from the group consisting of:
Figure imgf000199_0002
133. A compound according to claim 126, wherein M comprises a hydroxamic acid moiety.
134. A compound according to claim 126, wherein -L-M is
Figure imgf000200_0001
135. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000200_0002
Figure imgf000200_0003
wherein each V is independently selected from the group consisting of C(Rι2)2 and NR12 where at least one V is NR12; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; Rio and Rπ are taken together to form a substituted or unsubstituted aromatic ring; each R1 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
136. A compound according to claim 135, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
137. A compound accordmg to claim 135, wherein the substituted or unsubstituted aromatic ring formed when R10 and Rπ are taken together is selected from the group consisting of substituted or unsubstituted aryl and heteroaryl.
138. A compound according to claim 135, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF ), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
139. A compound according to claim 135, wherein M is selected from the group consisting of:
Figure imgf000202_0001
140. A compound according to claim 135, wherein M comprises a hydroxamic acid moiety.
141. A compound according to claim 135, wherein -L-M is
Figure imgf000202_0002
142. A compound comprising the formula
Figure imgf000202_0003
wherein each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NRι2; each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, R , and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the remainder of the compound.
143. A compound according to claim 142, wherein V are selected so that the ring is an N-substituted piperdin-3-yl moiety.
144. A compound according to claim 142, wherein at least one of R2, R3, R , or R5 is fluorine.
145. A compound according to claim 142, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
146. A compound according to claim 142, wherein M is selected from the group consisting of:
Figure imgf000204_0001
147. A compound according to claim 142, wherein M comprises a hydroxamic acid moiety.
148. A compound according to claim 142, wherein -L-M is
..OH
H
149. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000204_0002
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 2-10 atoms separation between M and the Q substituent.
150. A compound according to claim 149, wherein V are selected so that the ring is an
N-substituted piperdin-3-yl moiety.
151. A compound according to claim 149, wherein at least one of R2, R3, R , or R5 is fluorine.
152. A compound according to claim 149, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
153. A compound according to claim 149, wherein M is selected from the group consisting of:
Figure imgf000206_0001
154. A compound according to claim 149, wherein M comprises a hydroxamic acid moiety.
155. A compound according to claim 149, wherein -L-M is
Figure imgf000206_0002
156. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000206_0003
each V is independently selected from the group consisting of C(R12)2 and NR12 where at least one V is NR12;
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12 is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
M is selected from the group consisting of
Figure imgf000207_0001
and L is E, Z or mixtures of E/Z -CH2=CH2-.
157. A compound according to claim 156, wherein V are selected so tiiat the ring is an N-substituted piperdin-3-yl moiety.
158. A compound according to claim 156 wherein at least one of R2, R3, R4, or R5 is fluorine.
159. A compound according to claim 156, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
160. A compound according to claim 156, wherein M is selected from the group consisting of:
Figure imgf000208_0001
161. A compound according to claim 156, wherein M comprises a hydroxamic acid moiety.
162. A compound according to claim 156, wherein -L-M is
Figure imgf000208_0002
163. A compound comprising a formula selected from the group consisting of:
Figure imgf000208_0003
wherein each X is independently selected from the group consisting of CRι2 and N; each Y is independently selected from the group consisting of O, S and NRι2; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
Ri4 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring÷
164. A compound according to claim 163, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000209_0001
165. A compound according to claim 163, wherein the compound comprises a formula selected from d e group consisting of
Figure imgf000209_0002
166. A compound according to claim 163, wherein the compound comprises the formula
Figure imgf000209_0003
167. A compound according to claim 163, wherein R1 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
168. A compound according to claim 163, wherein R14 is a substituted or unsubstituted Cw alkyl.
169. A compound according to claim 163, wherein R14 is a substituted or unsubstituted -C(O)Cι-6 alkyl.
170. A compound according to claim 163, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
171. A compound according to claim 163, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO NH2)3 hydroxysulfonamides (-SO NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkyla ino, or an alkoxy group.
172. A compound according to claim 163, wherein M is selected from the group consisting of:
J A l .OH ? s h'-N H-OH
Figure imgf000210_0001
Figure imgf000210_0002
173. A compound according to claim 163, wherein M comprises a hydroxamic acid moiety.
174. A compound according to claim 163, wherein -L-M is
Figure imgf000211_0001
175. A compound comprising the formula
z- -M
wherein
Z is selected from the group consisting of
Figure imgf000211_0002
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which Rι2 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and or a metal ion; and
L is a substituent providing between 0-10 atoms separation between M and the ring.
176. A compound according to claim 175, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000212_0001
177. A compound according to claim 175, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000212_0002
178. A compound according to claim 175 wherein the compound comprises the formula
Figure imgf000213_0001
179. A compound according to claim 175, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
180. A compound according to claim 175, wherein R14 is a substituted or unsubstituted C1-6 alkyl.
181. A compound according to claim 175, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
182. A compound according to claim 175, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
183. A compound according to claim 175, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
184. A compound according to claim 175, wherein M is selected from the group consisting of:
Figure imgf000214_0001
185. A compound according to claim 175, wherein M comprises a hydroxamic acid moiety.
186. A compound according to claim 175, wherein -L-M is
Figure imgf000214_0002
187. A compound comprising the formula
z- Q L M
wherein
Z is selected from the group consisting of
Figure imgf000214_0003
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
188. A compound according to claim 187, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000215_0001
189. A compound according to claim 187, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000216_0001
190. A compound according to claim 187, wherein the compound comprises the formula
Figure imgf000216_0002
191. A compound according to claim 190, wherein R1 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
192. A compound according to claim 190, wherein Rι is a substituted or unsubstituted Ci-6 alkyl.
193. A compound according to claim 190, wherein Rι is a substituted or unsubstituted -C(O)C1-6 alkyl.
194. A compound according to claim 190, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
195. A compound of claim 187, wherein Q is a substituted or unsubstituted phenyl ring.
196. A compound of claim 187, wherein Q is a substituted or unsubstituted heteroaryl.
197. A compound of claim 187, wherein Q is a substituted or unsubstituted heteroaryl selected from the group consisting of furan, thiophene, pyrrole, pyrazole, triazole, isoxazole, oxazole, thiazole, isothiazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, indole, isobenzazole, quinoline, isoquinoline, cinnoline, quinazoline, naphthyridine, pyridopyridine, quinoxaline, phthalazine, benthiazole, and triazine.
198. A compound according to claim 187, wherein at least one X in the six membered ring is a substituted carbon atom.
199. A compound according to claim 187, wherein at least one of the X substituents in the six membered ring is -CF.
200. A compound according to claim 187, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
201. A compound according to claim 187, wherein M is selected from the group consisting of:
Figure imgf000217_0001
202. A compound according to claim 187, wherein M comprises a hydroxamic acid moiety.
203. A compound according to claim 187, wherein -Q-L-M is
Figure imgf000218_0001
204. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000218_0002
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R2, R3, -t and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; and each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R1 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
205. A compound according to claim 204, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000220_0001
RR1M4
Figure imgf000220_0003
Figure imgf000220_0002
R
Figure imgf000220_0004
206. A compound according to claim 204, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000220_0005
Figure imgf000220_0006
207. A compound according to claim 204, wherein the compound comprises the formula
Figure imgf000221_0001
208. A compound according to claim 204, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
209. A compound according to claim 204, wherein R14 is a substituted or unsubstituted Cι-6 alkyl.
210. A compound accordmg to claim 204, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
211. A compound according to claim 204, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
212. A compound according to claim 204, wherein at least one of R2, R3, R4, or R5 is fluorine.
213. A compound according to claim 204, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
214. A compound according to claim 204, wherein M is selected from the group consisting of:
Figure imgf000222_0001
215. A compound according to claim 204, wherein M comprises a hydroxamic acid moiety.
216. A compound according to claim 204, wherein -Q-L-M is
Figure imgf000222_0002
217. A compound comprising a formula selected from the group consisting of:
Figure imgf000222_0003
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NRι2;
R6, R , R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between die M substituent and the ring.
218. A compound according to claim 217, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000223_0001
219. A compound according to claim 217, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000224_0001
220. A compound according to claim 217, wherein the compound comprises the formula
Figure imgf000224_0002
221. A compound according to claim 217, wherein R1 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
222. A compound according to claim 217, wherein Rι4 is a substituted or unsubstituted Cι-6 alkyl.
223. A compound according to claim 217, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
224. A compound according to claim 217, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
225. A compound according to claim 217, wherein at least one of R6, R7, R8, and R9 is fluorine.
226. A compound according to claim 217, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
227. A compound according to claim 217, wherein M is selected from the group consisting of:
Figure imgf000225_0001
228. A compound according to claim 217, wherein M comprises a hydroxamic acid moiety.
229. A compound according to claim 217, wherein -L-M is
Figure imgf000225_0002
230. A compound comprising a formula selected from the group consisting of:
Figure imgf000226_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which Rι2 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted; M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
231. A compound according to claim 230, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000227_0001
232. A compound according to claim 230, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000228_0001
233. A compound according to claim 230, wherein the compound comprises the formula
Figure imgf000228_0002
234. A compound according to claim 230, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
235. A compound according to claim 230, wherein R14 is a substituted or unsubstituted Cι-6 alkyl.
236. A compound according to claim 230, wherein R1 is a substituted or unsubstituted -C(O)C1-6 alkyl.
237. A compound according to claim 230, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
238. A compound according to claim 230, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
239. A compound according to claim 230, wherein M is selected from the group consisting of:
Figure imgf000229_0001
240. A compound according to claim 230, wherein M comprises a hydroxamic acid moiety.
241. A compound according to claim 230, wherein -L-M is
Figure imgf000229_0002
242. A compound according to claim 230, wherein at least one of R6, R , R8, and R9 is fluorine.
243. A compound according to claim 230, wherein at least one X in the six membered ring is a substituted carbon atom.
244. A compound according to claim 230, wherein at least one of the X substituents in the six membered ring is -CF.
245. A compound comprising a formula selected from the group consisting of:
Figure imgf000230_0001
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12; R2, R3, , R5, R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroaiylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that R12is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the ring.
246. A compound according to claim 245, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000232_0001
247. A compound according to claim 245, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000233_0001
248. A compound according to claim 245, wherein the compound comprises the formula
Figure imgf000233_0002
249. A compound according to claim 245, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
250. A compound according to claim 245, wherein R14 is a substituted or unsubstituted Cw alkyl.
251. A compound according to claim 245, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
252. A compound according to claim 245, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
253. A compound according to claim 245, wherein at least one of R2, R3, R4, or R5 is fluorine.
254. A compound according to claim 245, wherein at least one of R6, R , Rg, and R9 is fluorine.
255. A compound according to claim 245, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
256. A compound according to claim 245, wherein M is selected from the group consisting of:
Figure imgf000235_0001
257. A compound according to claim 245, wherein M comprises a hydroxamic acid moiety.
258. A compound according to claim 245, wherein -L-M is
Figure imgf000235_0002
259. A compound comprising the formula
Z Q L M
wherein
Z is selected from the group consisting of
Figure imgf000235_0003
wherein each X is independently selected from the group consisting of CR12 and N; each Y is independently selected from the group consisting of O, S and NR12;
R10 and Rn are taken together to form a substituted or unsubstituted aromatic ring; each R12 is independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted, with the proviso that Rι2is not halo, cyano, nitro, and thio in the case where the ring atom to which R12 is bound is nitrogen;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
Q is a substituted or unsubstituted aromatic ring;
M is a substituent capable of complexing witii a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the Q substituent.
260. A compound according to claim 259, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000237_0001
261. A compound according to claim 259, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000237_0002
262. A compound according to claim 259, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
263. A compound according to claim 259, wherein R14 is a substituted or unsubstituted C1-6 alkyl.
264. A compound according to claim 259, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
265. A compound according to claim 259, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
266. A compound according to claim 259, wherein the substituted or unsubstituted aromatic ring formed when R10 and Rn are taken together is selected from the group consisting of substituted or unsubstituted aryl and heteroaryl.
267. A compound according to claim 259, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein Rι3 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
268. A compound according to claim 259, wherein M is selected from the group consisting of:
Figure imgf000238_0001
269. A compound according to claim 259, wherein M comprises a hydroxamic acid moiety.
270. A compound according to claim 259, wherein -L-M is
Figure imgf000239_0001
271. A compound comprising the formula
Figure imgf000239_0002
wherein
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing with a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 0-10 atoms separation between the M substituent and the remainder of the compound.
272. A compound according to claim 271, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000240_0001
273. A compound according to claim 271 , wherein the compound comprises a formula selected from the group consisting of
Figure imgf000240_0002
274. A compound according to claim 271, wherein R1 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
275. A compound according to claim 271, wherein R14 is a substituted or unsubstituted Cw alkyl.
276. A compound according to claim 271, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
277. A compound according to claim 271, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
278. A compound according to claim 271, wherein at least one of R2, R3, R4, or R5 is fluorine.
279. A compound accordmg to claim 271, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
280. A compound according to claim 271, wherein M is selected from the group consisting of:
Figure imgf000241_0001
281. A compound according to claim 271 , wherein M comprises a hydroxamic acid moiety.
282. A compound according to claim 271, wherein -L-M is
Figure imgf000241_0002
283. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000242_0001
wherein
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, thio, cyano, nitro, and a carbonyl group, each substituted or unsubstituted;
R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is a substituent capable of complexing witii a deacetylase catalytic site and/or a metal ion; and
L is a substituent providing between 2-10 atoms separation between the M substituent and the Q substituent.
284. A compound according to claim 283, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000242_0002
285. A compound according to claim 283, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000243_0001
286. A compound according to claim 283, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
287. A compound according to claim 283, wherein R14 is a substituted or unsubstituted Cw alkyl.
288. A compound according to claim 283, wherein R1 is a substituted or unsubstituted
-C(O)C1-6 alkyl.
289. A compound according to claim 283, wherein R1 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
290. A compound according to claim 283, wherein at least one of R2, R3, R4, or R5 is fluorine.
291. A compound according to claim 283, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO2NH2), hydroxysulfonamides (-SO2NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein Rι3 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
292. A compound according to claim 283, wherein M is selected from the group consisting of:
Figure imgf000244_0001
293. A compound according to claim 283, wherein M comprises a hydroxamic acid moiety.
294. A compound according to claim 283, wherein -L-M is
Figure imgf000244_0002
295. A compound comprising the formula:
Z Q L M
wherein
Z-Q- is selected from the group consisting of
Figure imgf000244_0003
wherein
R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, cyano, and nitro; R14 is selected from the group consisting of hydrogen, halo, alkyl, alkoxy, aryl, heteroaryl, aminosulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aryloxy, heteroaryloxy, arylalkyl, heteroarylalkyl, amino, and a carbonyl group, each substituted or unsubstituted;
M is selected from the group consisting of
Figure imgf000245_0001
and L is E, Z or mixtures of E/Z -CH2=CH2-.
296. A compound according to claim 295, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000245_0002
297. A compound according to claim 295, wherein the compound comprises a formula selected from the group consisting of
Figure imgf000245_0003
298. A compound according to claim 295, wherein R14 comprises a member selected from the group consisting of hydrogen and a substituent that is convertible in vivo to hydrogen.
299. A compound according to claim 295, wherein R14 is a substituted or unsubstituted Cw alkyl.
300. A compound according to claim 295, wherein R14 is a substituted or unsubstituted -C(O)C1-6 alkyl.
301. A compound according to claim 295, wherein R14 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, butyl, acetyl, and BOC.
302. A compound according to claim 295, wherein at least one of R , R3, R4, or R5 is fluorine.
303. A compound according to claim 295, wherein M comprises a member selected from the group consisting of trifluoroacetyl (-C(O)-CF3), -NH-P(O)OH-CH3, sulfonamides (-SO NH2), hydroxysulfonamides (-SO NHOH), thiols(-SH), and carbonyl groups having the formula -C(O)-R13 wherein R13 is hydroxylamino, hydroxyl, amino, alkylamino, or an alkoxy group.
304. A compound according to claim 295, wherein M is selected from the group consisting of:
Figure imgf000246_0001
305. A compound according to claim 295, wherein M comprises a hydroxamic acid moiety.
306. A compound according to claim 295, wherein -L-M is
Figure imgf000247_0001
PCT/US2004/008342 2003-03-17 2004-03-17 Histone deacetylase inhibitors WO2004082638A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2006507336A JP2006520796A (en) 2003-03-17 2004-03-17 Histone deacetylase inhibitor
EP04757631A EP1608628A2 (en) 2003-03-17 2004-03-17 Histone deacetylase inhibitors
CA002518318A CA2518318A1 (en) 2003-03-17 2004-03-17 Histone deacetylase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US45543703P 2003-03-17 2003-03-17
US60/455,437 2003-03-17
US53120303P 2003-12-19 2003-12-19
US60/531,203 2003-12-19

Publications (2)

Publication Number Publication Date
WO2004082638A2 true WO2004082638A2 (en) 2004-09-30
WO2004082638A3 WO2004082638A3 (en) 2005-05-06

Family

ID=33032690

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/008342 WO2004082638A2 (en) 2003-03-17 2004-03-17 Histone deacetylase inhibitors

Country Status (6)

Country Link
US (3) US7169801B2 (en)
EP (1) EP1608628A2 (en)
JP (1) JP2006520796A (en)
KR (1) KR20050122210A (en)
CA (1) CA2518318A1 (en)
WO (1) WO2004082638A2 (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1863811A1 (en) * 2005-03-21 2007-12-12 S*Bio Pte Ltd Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
JP2009503117A (en) * 2005-08-04 2009-01-29 サートリス ファーマシューティカルズ, インコーポレイテッド Benzimidazole derivatives as sirtuin modulators
JP2009507829A (en) * 2005-09-08 2009-02-26 エス*バイオ プライベート リミティッド Heterocyclic compounds
US7507826B2 (en) 2004-03-30 2009-03-24 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
JP2009521424A (en) * 2005-12-19 2009-06-04 メチルジーン インコーポレイテッド Histone deacetylase inhibitors for enhancing antifungal activity
US7666880B2 (en) 2005-03-21 2010-02-23 S*Bio Pte Ltd. Imidazo[1,2-A]pyridine derivatives: preparation and pharmaceutical applications
US7709468B2 (en) 2005-09-02 2010-05-04 Abbott Laboratories Imidazo based heterocycles
US7723332B2 (en) 2007-03-06 2010-05-25 Wyeth Llc Aryl sulfonamides useful for modulation of the progesterone receptor
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
US7767816B2 (en) 2006-01-17 2010-08-03 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of janus kinases
US7772245B2 (en) 2005-02-14 2010-08-10 Miikana Therapeutics, Inc. Inhibitors of histone deacetylase
WO2011003365A1 (en) * 2009-07-10 2011-01-13 天津尚德药缘科技有限公司 Histone deacetylase inhibitors with branched structure synthesized through click chemistry
US7884105B2 (en) 2005-10-27 2011-02-08 Janssen Pharmaceutica, N.V. Squaric acid derivatives as inhibitors of histone deacetylase
US7947830B2 (en) 2004-07-28 2011-05-24 Janssen Pharmaceutica N.V. Substituted propenyl piperazine derivatives as novel inhibitors of histone deacetylase
US7973069B2 (en) 2004-07-14 2011-07-05 Ptc Therapeutics, Inc. Methods for treating hepatitis C
US8017781B2 (en) 2005-11-15 2011-09-13 Vertex Pharmaceuticals Incorporated Azaindazoles useful as inhibitors of kinases
US8071615B2 (en) 2002-03-13 2011-12-06 Janssen Pharmaceutica N.V. Carbonylamino-derivatives as novel inhibitors of histone deacetylase
US8101616B2 (en) 2006-01-19 2012-01-24 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8114876B2 (en) 2006-01-19 2012-02-14 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8114999B2 (en) 2002-03-13 2012-02-14 Janssen Pharmaceutica N.V. Aminocarbonyl-derivatives as novel inhibitors of histone deacetylase
US8119650B2 (en) 2006-01-19 2012-02-21 Janssen Pharmaceutica N.V. Aminophenyl derivatives as novel inhibitors of histone deacetylase
US8138168B1 (en) 2007-09-26 2012-03-20 Takeda Pharmaceutical Company Limited Renin inhibitors
US8138198B2 (en) 2005-05-18 2012-03-20 Angibaud Patrick Rene Substituted aminopropenyl piperidine or morpholine derivatives as novel inhibitors of histone deacetylase
US8163733B2 (en) 2002-03-13 2012-04-24 Janssen Pharmaceutica N.V. Sulfonylamino-derivatives as novel inhibitors of histone deacetylase
US8163765B2 (en) 2006-01-19 2012-04-24 Janssen Pharmaceutica N.V. Substituted indolyl-alkyl-amino-derivatives as inhibitors of histone deacetylase
US8247421B2 (en) 2006-12-21 2012-08-21 Vertex Pharmaceuticals Incorporated 5-cyano-4-(pyrrolo [2,3B] pyridine-3-yl)-pyrimidine derivatives useful as protein kinase inhibitors
WO2013043520A1 (en) 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Triazolopyridyl compounds as aldosterone synthase inhibitors
US8476289B2 (en) 2008-03-27 2013-07-02 Janssen Pharmaceutica Nv Aza-bicyclohexyl substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylace
US8501737B2 (en) 2002-03-13 2013-08-06 Janssen Pharmaceutica N.V. Piperazinyl-, piperidinyl- and morpholinyl-derivatives as novel inhibitors of histone deacetylase
US8518911B2 (en) 2008-08-05 2013-08-27 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyridines as MARK inhibitors
US8624040B2 (en) 2009-06-22 2014-01-07 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
WO2014055595A1 (en) 2012-10-05 2014-04-10 Merck Sharp & Dohme Corp. Indoline compounds as aldosterone synthase inhibitiors related applications
US8778931B2 (en) 2010-12-22 2014-07-15 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
US8828392B2 (en) 2005-11-10 2014-09-09 Topotarget Uk Limited Histone deacetylase (HDAC) inhibitors (PXD101) for the treatment of cancer alone or in combination with chemotherapeutic agent
US8835501B2 (en) 2005-05-13 2014-09-16 Topotarget Uk Limited Pharmaceutical formulations of HDAC inhibitors
US8871774B2 (en) 2010-12-16 2014-10-28 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US8987273B2 (en) 2010-07-28 2015-03-24 Bayer Intellectual Property Gmbh Substituted imidazo[1,2-B]pyridazines
US9051319B2 (en) 2011-08-01 2015-06-09 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US9096518B2 (en) 2009-06-22 2015-08-04 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
US9206185B2 (en) 2011-04-07 2015-12-08 Bayer Intellectual Property Gmbh Imidazopyridazines as Akt kinase inhibitors
US9351973B2 (en) 2011-09-22 2016-05-31 Merck Sharp & Dohme Corp. Pyrazolopyridyl compounds as aldosterone synthase inhibitors
US9636341B2 (en) 2004-07-28 2017-05-02 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US9771361B2 (en) 2013-11-13 2017-09-26 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US9808459B2 (en) 2009-06-17 2017-11-07 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10016401B2 (en) 2014-09-17 2018-07-10 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10023569B2 (en) 2013-11-13 2018-07-17 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US10273233B2 (en) 2015-05-13 2019-04-30 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10285959B2 (en) 2005-02-03 2019-05-14 Topotarget Uk Limited Combination therapies using HDAC inhibitors
US10533004B2 (en) 2015-05-13 2020-01-14 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US11168093B2 (en) 2018-12-21 2021-11-09 Celgene Corporation Thienopyridine inhibitors of RIPK2
US11891382B2 (en) 2017-04-26 2024-02-06 Basilea Pharmaceutica International AG Processes for the preparation of furazanobenzimidazoles and crystalline forms thereof
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds

Families Citing this family (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1608628A2 (en) 2003-03-17 2005-12-28 Takeda San Diego, Inc. Histone deacetylase inhibitors
EP1611088B1 (en) 2003-04-07 2009-06-17 Pharmacyclics, Inc. Hydroxamates as therapeutic agents
BRPI0414581C1 (en) 2003-09-22 2021-05-25 Mei Pharma Inc compound, pharmaceutical composition comprising said compound and use of said compound
US20050197336A1 (en) * 2004-03-08 2005-09-08 Miikana Therapeutics Corporation Inhibitors of histone deacetylase
US7345043B2 (en) * 2004-04-01 2008-03-18 Miikana Therapeutics Inhibitors of histone deacetylase
US7507552B1 (en) * 2004-04-16 2009-03-24 Takeda San Diego, Inc. Crystallization of histone deacetylase 2
BRPI0511834A (en) 2004-07-14 2008-01-08 Ptc Therapeutics Inc methods for treating hepatitis c
CA2620333A1 (en) 2005-08-26 2007-03-01 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP2258358A3 (en) 2005-08-26 2011-09-07 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP1940389A2 (en) 2005-10-21 2008-07-09 Braincells, Inc. Modulation of neurogenesis by pde inhibition
CA2625210A1 (en) 2005-10-31 2007-05-10 Braincells, Inc. Gaba receptor mediated modulation of neurogenesis
CN101374828B (en) 2006-01-19 2012-09-19 詹森药业有限公司 Heterocyclylalkyl derivatives as novel inhibitors of histone deacetylase
EP1979328B1 (en) 2006-01-19 2012-12-26 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8168658B2 (en) * 2006-02-28 2012-05-01 Merck Sharp & Dohme Corp. Inhibitors of histone deacetylase
US20100216734A1 (en) 2006-03-08 2010-08-26 Braincells, Inc. Modulation of neurogenesis by nootropic agents
MX2008014320A (en) 2006-05-09 2009-03-25 Braincells Inc 5 ht receptor mediated neurogenesis.
AU2007249399A1 (en) 2006-05-09 2007-11-22 Braincells, Inc. Neurogenesis by modulating angiotensin
EP2056808A4 (en) * 2006-08-28 2009-12-23 Univ California Small molecule potentiator of hormonal therapy for breast cancer
MX2009002496A (en) 2006-09-08 2009-07-10 Braincells Inc Combinations containing a 4-acylaminopyridine derivative.
US20100184806A1 (en) 2006-09-19 2010-07-22 Braincells, Inc. Modulation of neurogenesis by ppar agents
NZ579635A (en) * 2007-03-20 2011-07-29 Curis Inc Fused amino pyridine as hsp90 inhibitors
CA2692153A1 (en) * 2007-06-27 2009-01-08 Richard W. Heidebrecht, Jr. Pyridyl and pyrimidinyl derivatives as histone deacetylase inhibitors
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US8008344B2 (en) * 2007-09-14 2011-08-30 NatureWise Biotech and Medicals Corporation Compounds for the inhibition of histone deacetylase
MX2010003230A (en) * 2007-09-25 2010-04-07 Topotarget Uk Ltd Methods of synthesis of certain hydroxamic acid compounds.
CA2706750A1 (en) * 2007-11-27 2009-06-04 Ottawa Health Research Institute Amplification of cancer-specific oncolytic viral infection by histone deacetylase inhibitors
CN101970399A (en) * 2007-12-14 2011-02-09 乔治敦大学 Histone deacetylase inhibitors
US7802883B2 (en) 2007-12-20 2010-09-28 Johnson & Johnson Vision Care, Inc. Cosmetic contact lenses having a sparkle effect
CA2717207A1 (en) * 2008-03-07 2009-09-11 Topotarget A/S Methods of treatment employing prolonged continuous infusion of belinostat
US8623853B2 (en) 2008-07-23 2014-01-07 The Brigham And Women's Hospital, Inc. Treatment of cancers characterized by chromosomal rearrangement of the NUT gene
US9107942B2 (en) * 2008-10-31 2015-08-18 University Of Rochester Methods of diagnosing and treating fibrosis
GB0900555D0 (en) * 2009-01-14 2009-02-11 Topotarget As New methods
AU2010204608B2 (en) * 2009-01-16 2013-12-05 Curis, Inc. Fused amino pyridines for the treatment of brain tumors
US20100216805A1 (en) 2009-02-25 2010-08-26 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
US7994357B2 (en) * 2009-04-03 2011-08-09 Naturewise Biotech & Medicals Corporation Cinamic compounds and derivatives therefrom for the inhibition of histone deacetylase
EP2236503B1 (en) 2009-04-03 2014-02-26 NatureWise Biotech & Medicals Corporation Cinamic compounds and derivatives therefrom for the inhibition of histone deacetylase
US8603521B2 (en) * 2009-04-17 2013-12-10 Pharmacyclics, Inc. Formulations of histone deacetylase inhibitor and uses thereof
EP2277387B1 (en) 2009-07-22 2016-10-19 NatureWise Biotech & Medicals Corporation New use of histone deacetylase inhibitors in changing mrjp3 protein in royal jelly
TWI600638B (en) * 2010-01-22 2017-10-01 艾斯特隆製藥公司 Reverse amide compounds as protein deacetylase inhibitors and methods of use thereof
CN107277527B (en) 2010-07-15 2020-02-18 威勒斯媒体国际有限公司 Decoding device, decoding method, encoding device, and encoding method
ES2568260T3 (en) 2010-11-16 2016-04-28 Acetylon Pharmaceuticals, Inc. Pyrimidine hydroxy amide compounds as protein deacetylase inhibitors and methods of use thereof
RU2609833C2 (en) 2011-09-13 2017-02-06 Фармасайкликс Элэлси Dosage forms of histone deacetylase inhibitor in combination with bendamustine and their application
WO2013043518A1 (en) 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Imidazopyridyl compounds as aldosterone synthase inhibitors
CN104136410A (en) * 2011-12-29 2014-11-05 药品循环公司 Cinnamic acid hydroxyamides as inhibitors of histone deacetylase 8
EP2920181B1 (en) 2012-11-16 2019-01-09 University Health Network Pyrazolopyrimidine compounds
JP6800750B2 (en) 2013-08-02 2020-12-16 ファーマサイクリックス エルエルシー Treatment method for solid tumors
US9403779B2 (en) 2013-10-08 2016-08-02 Acetylon Pharmaceuticals, Inc. Combinations of histone deacetylase inhibitors and either Her2 inhibitors or PI3K inhibitors
ES2862126T3 (en) 2013-10-10 2021-10-07 Acetylon Pharmaceuticals Inc Pyrimidine-hydroxyamide compounds as histone deacetylase inhibitors
EP4137135B1 (en) 2013-10-24 2024-06-05 Mayo Foundation for Medical Education and Research Treatment of polycystic diseases with an hdac6 inhibitor
JP6535670B2 (en) 2013-12-03 2019-06-26 アセチロン ファーマシューティカルズ インコーポレイテッドAcetylon Pharmaceuticals,Inc. Combination of histone deacetylase inhibitors and immunomodulators
US9464073B2 (en) 2014-02-26 2016-10-11 Acetylon Pharmaceuticals, Inc. Pyrimidine hydroxy amide compounds as HDAC6 selective inhibitors
US9833466B2 (en) 2014-07-07 2017-12-05 Acetylon Pharmaceuticals, Inc. Treatment of leukemia with histone deacetylase inhibitors
JP2017537164A (en) 2014-12-05 2017-12-14 ユニヴァーシティ・オブ・モデナ・アンド・レッジョ・エミリア Combination of histone deacetylase inhibitor and bendamustine for the treatment of lymphoma
US9745253B2 (en) 2015-03-13 2017-08-29 Forma Therapeutics, Inc. Alpha-cinnamide compounds and compositions as HDAC8 inhibitors
US10272084B2 (en) 2015-06-01 2019-04-30 Regenacy Pharmaceuticals, Llc Histone deacetylase 6 selective inhibitors for the treatment of cisplatin-induced peripheral neuropathy
CN107922352B (en) 2015-06-08 2021-08-06 埃斯泰隆制药公司 Process for preparing protein deacetylase inhibitors
JP6816036B2 (en) 2015-06-08 2021-01-20 アセチロン ファーマシューティカルズ インコーポレイテッドAcetylon Pharmaceuticals,Inc. Crystalline morphology of histone deacetylase inhibitors
SI3436461T1 (en) 2016-03-28 2024-03-29 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
WO2017184774A1 (en) 2016-04-19 2017-10-26 Acetylon Pharmaceuticals, Inc. Hdac inhibitors, alone or in combination with btk inhibitors, for treating chronic lymphocytic leukemia
AR108257A1 (en) 2016-05-02 2018-08-01 Mei Pharma Inc POLYMORPHIC FORMS OF 3- [2-BUTIL-1- (2-DIETILAMINO-ETIL) -1H-BENCIMIDAZOL-5-IL] -N-HYDROXY-ACRYLAMIDE AND USES OF THE SAME
US11324744B2 (en) 2016-08-08 2022-05-10 Acetylon Pharmaceuticals Inc. Methods of use and pharmaceutical combinations of histone deacetylase inhibitors and CD20 inhibitory antibodies
KR20190129034A (en) 2016-12-29 2019-11-19 세레니티 테라퓨틱스 (버뮤다), 리미티드 Metal Enzyme Inhibitor Compounds
WO2018125799A2 (en) 2016-12-29 2018-07-05 Viamet Pharmaceuticals (Bermuda), Ltd. Metalloenzyme inhibitor compounds

Family Cites Families (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4482571A (en) 1982-06-21 1984-11-13 University Of Pittsburgh Sickle cell anemia treatment and compound
US4997815A (en) 1988-11-01 1991-03-05 Children's Hospital Medical Center Of Northern California Method for augmenting fetal hemoglobin by treatment with activin and/or inhibin
IE900394L (en) 1989-02-08 1990-08-08 Abbott Lab Thiazole derivatives
US5216004A (en) 1990-09-13 1993-06-01 Children's Hospital Medical Center Of North California Method for preventing malaria
US5656644A (en) 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
US5439939A (en) 1992-03-17 1995-08-08 Children's Hospital Medical Center Of Northern California Pharmaceutical compositions and methods using isobutyramide for treating betaglobin disorders
WO1995011699A1 (en) 1993-10-29 1995-05-04 The Trustees Of Boston University Physiologically stable compositions of butyric acid, and butyric acid salts and derivatives as anti-neoplastic agents
AU679818B2 (en) 1993-11-10 1997-07-10 Sloan-Kettering Institute For Cancer Research Butyric ester cyto-differentiating agents
US5569675A (en) 1994-03-07 1996-10-29 Bar Ilan University Methods of using carboxylic acid esters to increase fetal-hemoglobin levels
US6040342A (en) 1994-09-16 2000-03-21 Bar-Ilan University Retinoyloxy (alkyl-substituted) methyl butyrates useful for the treatment of cancer and other proliferative diseases
US6071923A (en) 1994-09-16 2000-06-06 Bar-Ilan University Retinoyloxy aryl-substituted alkylene butyrates useful for the treatment of cancer and other proliferative diseases
US6011000A (en) 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US5700826A (en) 1995-06-07 1997-12-23 Ontogen Corporation 1,2,4,5-tetra substituted imidazoles as modulators of multi-drug resistance
JP3964478B2 (en) 1995-06-30 2007-08-22 エーザイ・アール・アンド・ディー・マネジメント株式会社 Heterocycle-containing carboxylic acid derivative and pharmaceutical containing the same
US5922837A (en) 1995-09-20 1999-07-13 Merck & Co., Inc. Antiprotozoal cyclic tetrapeptides
US6110697A (en) 1995-09-20 2000-08-29 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents
US6777217B1 (en) 1996-03-26 2004-08-17 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US6197743B1 (en) 1996-07-26 2001-03-06 The Trustees Of Boston University Compositions and methods for the treatment of viral disorders
US5939456A (en) 1996-07-26 1999-08-17 Perrine; Susan P. Pulsed administration of compositions for the treatment of blood disorders
EP0827742A1 (en) 1996-09-04 1998-03-11 Vrije Universiteit Brussel Use of histone deacetylase inhibitors for treating fribosis or cirrhosis
US6068987A (en) * 1996-09-20 2000-05-30 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents
US6174905B1 (en) * 1996-09-30 2001-01-16 Mitsui Chemicals, Inc. Cell differentiation inducer
US6235474B1 (en) 1996-12-30 2001-05-22 The Johns Hopkins University Methods and kits for diagnosing and determination of the predisposition for diseases
US6130248A (en) 1996-12-30 2000-10-10 Bar-Ilan University Tricarboxylic acid-containing oxyalkyl esters and uses thereof
US5939455A (en) 1997-03-11 1999-08-17 Beacon Laboratories, Inc. Therapeutic augmentation of oxyalkylene diesters and butyric acid derivatives
US6110970A (en) 1997-03-11 2000-08-29 Beacon Laboratories, Inc. Nitrogen-containing oxyalkylene esters and uses thereof
US6124495A (en) 1997-03-11 2000-09-26 Beacon Laboratories, Inc. Unsaturated oxyalkylene esters and uses thereof
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
US6043389A (en) * 1997-03-11 2000-03-28 Mor Research Applications, Ltd. Hydroxy and ether-containing oxyalkylene esters and uses thereof
US6110955A (en) 1997-03-11 2000-08-29 Beacon Laboratories, Inc. Metabolically stabilized oxyalkylene esters and uses thereof
US6387673B1 (en) 1997-05-01 2002-05-14 The Salk Institute For Biological Studies Compounds useful for the modulation of processes mediated by nuclear hormone receptors, methods for the identification and use of such compounds
US6506783B1 (en) 1997-05-16 2003-01-14 The Procter & Gamble Company Cancer treatments and pharmaceutical compositions therefor
US6231880B1 (en) 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
AUPO864097A0 (en) 1997-08-19 1997-09-11 Peplin Pty Ltd Anti-cancer compounds
US20010049379A1 (en) * 1997-08-27 2001-12-06 Lowe John Adams 2-aminopyridines containing fused ring substituents
WO1999011659A1 (en) 1997-09-02 1999-03-11 Japan Energy Corporation Novel cyclic tetrapeptide derivatives and medicinal use thereof
US6884597B1 (en) * 1998-01-20 2005-04-26 Medical & Biological Laboratories, Co., Ltd. Method for detecting acetyltransferase and deacetylase activities and method for screening inhibitors or enhancers of these enzymes
US6262116B1 (en) 1998-01-23 2001-07-17 Sloan-Kettering Institute For Cancer Research Transcription therapy for cancers
AU774861B2 (en) 1998-02-11 2004-07-08 Douglas V Faller Compositions and methods for the treatment of cystic fibrosis
US6287843B1 (en) 1998-04-03 2001-09-11 Pioneer Hi-Bred International, Inc. Maize histone deacetylases and their use
US6919366B2 (en) 1998-05-22 2005-07-19 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
US6099851A (en) 1998-06-02 2000-08-08 Weisman; Kenneth M. Therapeutic uses of leuprolide acetate
US20030236204A1 (en) * 2002-06-14 2003-12-25 Isis Pharmaceuticals Inc. Antisense modulation of histone deacetylase 2 expression
JP2002523029A (en) 1998-08-21 2002-07-30 スミスクライン ビーチャム コーポレーション Human histone deacetylase gene HD4
EP1123309A2 (en) 1998-10-13 2001-08-16 Fujisawa Pharmaceutical Co., Ltd. Cyclic tetrapeptides and their use as histone deacetylase inhibitor
WO2000028020A2 (en) 1998-11-10 2000-05-18 Board Of Regents, The University Of Texas System Methods for preventing cardiac hypertrophy and heart failure by inhibition of mef2 transcription factor
US6287790B1 (en) 1998-11-30 2001-09-11 The Regents Of The University Of California Utilization of nuclear structural proteins for targeted therapy and detection of proliferative and differentiation disorders
WO2000052204A2 (en) 1999-02-22 2000-09-08 Orntoft Torben F Gene expression in bladder tumors
JP4269041B2 (en) 1999-03-02 2009-05-27 国立大学法人九州工業大学 Novel cyclic tetrapeptide derivatives and their pharmaceutical uses
US6518012B1 (en) 1999-04-02 2003-02-11 Health Research, Inc. Method for regulating the expression of MHC antigens and CD40 by inhibitors of histone deacetylation
US20030078216A1 (en) * 1999-05-03 2003-04-24 Macleod A. Robert Inhibition of histone deactylase
US6630451B1 (en) 1999-06-29 2003-10-07 Orthomcneil Pharmaceutical, Inc. Benzimidazolone peptidometics as thrombin receptor antagonist
US20030078369A1 (en) * 1999-07-23 2003-04-24 Meinke Peter T. Apicidin-derived cyclic tetrapeptides
AU6794700A (en) 1999-08-20 2001-03-19 Board Of Regents, The University Of Texas System Methods and compositions relating to hdac 4 and 5 regulation of cardiac gene expression
JP2001081031A (en) 1999-08-30 2001-03-27 Schering Ag Benzamide derivative-containing preparation having improved solubility and oral adsorption
EP1231919B1 (en) * 1999-09-08 2015-09-30 Sloan-Kettering Institute For Cancer Research Derivatives of 1-amino-1-(hetero)arylaminocarbonyl-6-hydroxyaminocarbonylhexane useful in the treatment of tumors
EP1212357B1 (en) * 1999-09-08 2007-05-02 Sloan-Kettering Institute For Cancer Research Crystal structure of a deacetylase and inhibitors thereof
US20040024373A1 (en) * 1999-10-01 2004-02-05 Anna Chernyakhovsky Method and device for correcting or reducing the formation of facial lines
KR20020070285A (en) * 1999-11-23 2002-09-05 메틸진, 인크. Inhibitors of histone deacetylase
JP2001149081A (en) * 1999-11-29 2001-06-05 Cyclex Co Ltd Method for assaying activities of deacetylases, and method for screening inhibitor or activator of these enzymes
ES2225282T3 (en) 1999-12-08 2005-03-16 Xcyte Therapies, Inc. DEPSIPEPTIDE AND CONGENERES OF THE SAME TO USE AS IMMUNOSUPPRESSORS.
US6875598B1 (en) * 1999-12-08 2005-04-05 Applera Corporation Histone deacetylase-8 proteins, nuclei acids, and methods for use
US6828302B1 (en) 1999-12-08 2004-12-07 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
PL357109A1 (en) * 1999-12-28 2004-07-12 Pfizer Products Inc. Non-peptidyl inhibitors of vla-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US6544957B2 (en) 2000-01-04 2003-04-08 The Johns Hopkins University Methods and reagents for facilitating transcription
EE05515B1 (en) * 2000-01-14 2012-02-15 Schering@Aktiengesellschaft 1 2 diar 1-benzimidazoles for the treatment of diseases associated with microglial activation
CA2398590C (en) 2000-02-08 2012-08-28 Sangamo Biosciences, Inc. Cells for drug discovery
EP1280764B1 (en) 2000-03-24 2010-11-24 Methylgene, Inc. Inhibitors of histone deacetylase
WO2003006652A2 (en) 2000-03-24 2003-01-23 Methylgene, Inc. Inhibition of specific histone deacetylase isoforms
US20020137162A1 (en) 2000-03-24 2002-09-26 Zuomei Li Antisense oligonucleotide inhibition of specific histone deacetylase isoforms
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
EP1170008A1 (en) * 2000-07-07 2002-01-09 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
DE60137932D1 (en) * 2000-07-17 2009-04-23 Astellas Pharma Inc REDUCED FK228 AND ITS USE
US6673587B1 (en) 2000-08-11 2004-01-06 The Salk Institute For Biological Studies Histone deacetylase, and uses therefor
PE20020354A1 (en) 2000-09-01 2002-06-12 Novartis Ag HYDROXAMATE COMPOUNDS AS HISTONE-DESACETILASE (HDA) INHIBITORS
US6538030B2 (en) * 2000-09-20 2003-03-25 Yih-Lin Chung Treating radiation fibrosis
EP1328510B1 (en) * 2000-09-29 2013-11-20 TopoTarget UK Limited (e)-n-hydroxy-3-(3-sulfamoyl-phenyl)-acrylamide compounds and their therapeutic use
AU2002215018A1 (en) 2000-10-13 2002-04-22 Bayer Aktiengesellschaft Human histone deacetylase gene
DE10053474A1 (en) 2000-10-24 2002-05-02 Schering Ag Indirubin derivatives containing sulfur, their production and use
US20020103192A1 (en) 2000-10-26 2002-08-01 Curtin Michael L. Inhibitors of histone deacetylase
US20040043470A1 (en) * 2000-10-31 2004-03-04 Yonghong Xiao Regulation of human histone deacetylase
AU2002243231A1 (en) * 2000-11-21 2002-07-24 Wake Forest University Method of treating autoimmune diseases
DE10061162A1 (en) 2000-11-30 2002-07-11 Schering Ag Aryl-substituted indirubin derivatives, their preparation and use
AR035659A1 (en) * 2000-12-07 2004-06-23 Hoffmann La Roche HYDROXYAMIDES OF ACID (1-OXO-1,2,3,4-TETRAHIDRO-NAFTALEN-2-IL) -ALCANOICO, PROCESS FOR THE MANUFACTURE OF THESE COMPOUNDS, PHARMACEUTICAL COMPOSITIONS CONTAINING THESE COMPOUNDS AND USES OF THE SAME
US6376508B1 (en) 2000-12-13 2002-04-23 Academia Sinica Treatments for spinal muscular atrophy
US20040005574A1 (en) * 2002-07-08 2004-01-08 Leonard Guarente SIR2 activity
US20040077046A1 (en) * 2000-12-20 2004-04-22 Dalia Cohen Histone deacetylase-related gene and protein
US6720445B2 (en) 2000-12-21 2004-04-13 Beacon Laboratories, Inc. Acetyloxymethyl esters and methods for using the same
PA8535601A1 (en) 2000-12-21 2002-11-28 Pfizer BENZIMIDAZOL AND PIRIDILIMIDAZOL DERIVATIVES AS LIGANDOS FOR GABAA
US6562995B1 (en) * 2000-12-21 2003-05-13 Beacon Laboratories, Inc. Delta dicarbonyl compounds and methods for using the same
KR100456047B1 (en) 2000-12-22 2004-11-08 이인원 Apicidin-derivatives, their synthetic methods, and antitumor compositions containing the same
IL156517A0 (en) 2000-12-22 2004-01-04 Wyeth Corp Heterocyclindazole and azaindazole compounds as 5-hydroxytryptamine-6 ligands
AR035513A1 (en) * 2000-12-23 2004-06-02 Hoffmann La Roche DERIVATIVES OF TETRAHYDROPIRIDINE, PROCESS TO PREPARE THEM, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM, AND USE OF SUCH COMPOUNDS IN THE PREPARATION OF MEDICINES
US6756200B2 (en) * 2001-01-26 2004-06-29 The Johns Hopkins University School Of Medicine Aberrantly methylated genes as markers of breast malignancy
AR035417A1 (en) * 2001-01-27 2004-05-26 Hoffmann La Roche TRICYCLE DERIVATIVES OF LACTAMA AND SULTAMA, PROCESSES FOR THEIR DEVELOPMENT, DRUGS THAT CONTAIN THEM, AND THE USE OF SUCH COMPOUNDS IN THE PREPARATION OF DRUGS
US7022728B2 (en) * 2001-03-09 2006-04-04 Abbott Laboratories Benzimidazoles that are useful in treating male sexual dysfunction
US6495719B2 (en) * 2001-03-27 2002-12-17 Circagen Pharmaceutical Histone deacetylase inhibitors
US7312247B2 (en) 2001-03-27 2007-12-25 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors
US8026280B2 (en) 2001-03-27 2011-09-27 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors
SE0101387D0 (en) * 2001-04-20 2001-04-20 Astrazeneca Ab Novel compounds
AR035455A1 (en) * 2001-04-23 2004-05-26 Hoffmann La Roche TRICYCLE DERIVATIVES OF ALQUILHIDROXAMATO, PROCESSES FOR THEIR DEVELOPMENT, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM, AND THE USE OF SUCH COMPOUNDS IN THE PREPARATION OF MEDICINES
US6905669B2 (en) 2001-04-24 2005-06-14 Supergen, Inc. Compositions and methods for reestablishing gene transcription through inhibition of DNA methylation and histone deacetylase
US7244853B2 (en) * 2001-05-09 2007-07-17 President And Fellows Of Harvard College Dioxanes and uses thereof
US7030150B2 (en) * 2001-05-11 2006-04-18 Trimeris, Inc. Benzimidazole compounds and antiviral uses thereof
US7063973B2 (en) * 2001-06-14 2006-06-20 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
US6784173B2 (en) * 2001-06-15 2004-08-31 Hoffmann-La Roche Inc. Aromatic dicarboxylic acid derivatives
AR034897A1 (en) * 2001-08-07 2004-03-24 Hoffmann La Roche N-MONOACILATED DERIVATIVES OF O-PHENYLENDIAMINS, THEIR HETEROCICLICAL ANALOGS OF SIX MEMBERS AND THEIR USE AS PHARMACEUTICAL AGENTS
US6897220B2 (en) 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
EP1293205A1 (en) * 2001-09-18 2003-03-19 G2M Cancer Drugs AG Valproic acid and derivatives thereof for the combination therapy of human cancers, for the treatment of tumour metastasis and minimal residual disease
US6706686B2 (en) 2001-09-27 2004-03-16 The Regents Of The University Of Colorado Inhibition of histone deacetylase as a treatment for cardiac hypertrophy
WO2003032921A2 (en) * 2001-10-16 2003-04-24 Sloan-Kettering Institute For Cancer Research Treatment of neurodegenerative diseases and cancer of the brain
BR0213932A (en) * 2001-11-06 2004-08-31 Novartis Ag Cyclooxygenase-2 inhibitor / histone deacetylase inhibitor combination
US7514406B2 (en) * 2001-11-27 2009-04-07 Fred Hutchinson Cancer Research Center Methods for inhibiting deacetylase activity
US6972281B2 (en) 2002-02-07 2005-12-06 University Of Ottawa Histogranin-like peptides and non-peptides, processes for their preparation and uses thereof
AU2003215112A1 (en) * 2002-02-07 2003-09-02 Axys Pharmaceuticals Novel bicyclic hydroxamates as inhibitors of histone deacetylase
CA2632078C (en) * 2002-03-04 2012-08-14 Sloan-Kettering Institute For Cancer Research Methods of inducing terminal differentiation
US7148257B2 (en) * 2002-03-04 2006-12-12 Merck Hdac Research, Llc Methods of treating mesothelioma with suberoylanilide hydroxamic acid
CN101080499A (en) * 2002-03-07 2007-11-28 约翰霍普金斯大学 Genomic screen for epigenetically silenced tumor suppressor genes
EP1488005A1 (en) * 2002-03-26 2004-12-22 Exhonit Therapeutics SA Histone deacetylase: novel molecular target of neurotoxicity
US20040077591A1 (en) * 2002-03-28 2004-04-22 The Brigham And Women's Hospital, Inc. Histone deacetylase inhibitors for the treatment of multiple sclerosis, amyotrophic lateral sclerosis and Alzheimer's Disease
US6841565B1 (en) 2002-03-29 2005-01-11 The Ohio State University Treatment of patients with chronic lymphocytic leukemia
AU2003226408B2 (en) * 2002-04-15 2007-06-14 Sloan-Kettering Institute For Cancer Research Combination therapy for the treatment of cancer
US6809118B2 (en) 2002-07-25 2004-10-26 Yih-Lin Chung Methods for therapy of radiation cutaneous syndrome
US20040018522A1 (en) * 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
US7351542B2 (en) * 2002-05-20 2008-04-01 The Regents Of The University Of California Methods of modulating tubulin deacetylase activity
EP1511729A4 (en) * 2002-05-22 2006-09-06 Errant Gene Therapeutics Llc Histone deacetylase inhibitors based on alphachalcogenmethylcarbonyl compounds
AU2003243285A1 (en) * 2002-05-22 2003-12-12 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors based on trihalomethylcarbonyl compounds
CA2486303C (en) * 2002-05-22 2013-04-30 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
WO2003100089A1 (en) * 2002-05-23 2003-12-04 The Regents Of The University Of California Methods of modulating mitochondrial nad-dependent deacetylase
US20040002447A1 (en) * 2002-06-04 2004-01-01 Regents Of The University Of California Induction of insulin expression
US20050084967A1 (en) * 2002-06-28 2005-04-21 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
US20040072770A1 (en) * 2002-07-03 2004-04-15 Besterman Jeffrey M. Methods for specifically inhibiting histone deacetylase-7 and 8
SE0202157D0 (en) * 2002-07-09 2002-07-09 Biovitrum Ab Methods for identification of compounds modulating insulin resistance
WO2004009771A2 (en) * 2002-07-24 2004-01-29 Advanced Stent Technologies, Inc. Stents capable of controllably releasing histone deacetylase inhibitors
JP3905005B2 (en) * 2002-09-18 2007-04-18 富士通株式会社 Portable device and semiconductor integrated circuit device
US20040077083A1 (en) * 2002-10-17 2004-04-22 Isis Pharmaceuticals Inc. Antisense modulation of histone deacetylase 4 expression
US20040077084A1 (en) * 2002-10-17 2004-04-22 Isis Pharmaceuticals Inc. Antisense modulation of histone deacetylase 4 expression
EP1608628A2 (en) 2003-03-17 2005-12-28 Takeda San Diego, Inc. Histone deacetylase inhibitors
US20050020557A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors
US7271195B2 (en) * 2003-06-10 2007-09-18 Kalypsys, Inc. Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
US7842835B2 (en) * 2003-07-07 2010-11-30 Georgetown University Histone deacetylase inhibitors and methods of use thereof
US20050037992A1 (en) * 2003-07-22 2005-02-17 John Lyons Composition and method for treating neurological disorders
US20050032794A1 (en) * 2003-08-05 2005-02-10 Padia Janak K. Diamine derivatives of quinone and uses thereof
US20050059682A1 (en) * 2003-09-12 2005-03-17 Supergen, Inc., A Delaware Corporation Compositions and methods for treatment of cancer
DE602004027824D1 (en) * 2003-09-25 2010-08-05 Astellas Pharma Inc ANTITUMORAL AGENT WITH A HISTON DEACETYLASE INHIBITOR AND A TOPOISOMERASE II HEMMER
US20080132539A1 (en) * 2003-12-25 2008-06-05 Junji Kakuchi Hydroxamic Acid Derivative And Age Generation Inhibitor Containing The Derivative

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE CAPLUS [Online] XP002995463 Database accession no. 2001:526062 & WO 01 51473 A1 *

Cited By (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8343988B2 (en) 2002-03-13 2013-01-01 Janssen Pharmaceutica, N.V Inhibitors of histone deacetylase
US8524711B2 (en) 2002-03-13 2013-09-03 Janssen Pharmaceutica N.V. Amino-derivatives as novel inhibitors of histone deacetylase
US8071615B2 (en) 2002-03-13 2011-12-06 Janssen Pharmaceutica N.V. Carbonylamino-derivatives as novel inhibitors of histone deacetylase
US8501737B2 (en) 2002-03-13 2013-08-06 Janssen Pharmaceutica N.V. Piperazinyl-, piperidinyl- and morpholinyl-derivatives as novel inhibitors of histone deacetylase
US8163733B2 (en) 2002-03-13 2012-04-24 Janssen Pharmaceutica N.V. Sulfonylamino-derivatives as novel inhibitors of histone deacetylase
US8114999B2 (en) 2002-03-13 2012-02-14 Janssen Pharmaceutica N.V. Aminocarbonyl-derivatives as novel inhibitors of histone deacetylase
US8455498B2 (en) 2002-03-13 2013-06-04 Janssen Pharmaceutica N.V. Inhibitors of histone deacetylase
US9556161B2 (en) 2002-03-13 2017-01-31 Janssen Pharmaceutica Nv Inhibitors of histone deacetylase
US8268833B2 (en) 2002-03-13 2012-09-18 Janssen Pharmaceutica, N.V. Inhibitors of histone deacetylase
US8501446B2 (en) 2004-03-30 2013-08-06 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
US8188281B2 (en) 2004-03-30 2012-05-29 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
US7507826B2 (en) 2004-03-30 2009-03-24 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
US8987454B2 (en) 2004-03-30 2015-03-24 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
US8722889B2 (en) 2004-03-30 2014-05-13 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of JAK and other protein kinases
US7973069B2 (en) 2004-07-14 2011-07-05 Ptc Therapeutics, Inc. Methods for treating hepatitis C
US8426416B2 (en) 2004-07-28 2013-04-23 Janssen Pharmaceutica, N.V. Substituted propenyl piperazine derivatives as novel inhibitors of histone deacetylase
US7947830B2 (en) 2004-07-28 2011-05-24 Janssen Pharmaceutica N.V. Substituted propenyl piperazine derivatives as novel inhibitors of histone deacetylase
US9636341B2 (en) 2004-07-28 2017-05-02 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US10799469B2 (en) 2005-02-03 2020-10-13 Topotarget Uk Limited Combination therapies using HDAC inhibitors
US10285959B2 (en) 2005-02-03 2019-05-14 Topotarget Uk Limited Combination therapies using HDAC inhibitors
US7772245B2 (en) 2005-02-14 2010-08-10 Miikana Therapeutics, Inc. Inhibitors of histone deacetylase
EP1863811A1 (en) * 2005-03-21 2007-12-12 S*Bio Pte Ltd Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
US8901149B2 (en) 2005-03-21 2014-12-02 Mei Pharma, Inc. Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
US9266882B2 (en) 2005-03-21 2016-02-23 Mei Pharma, Inc. Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
EP1863811A4 (en) * 2005-03-21 2008-11-12 S Bio Pte Ltd Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
US8648092B2 (en) 2005-03-21 2014-02-11 MEI Pharma Imidazo[1,2-a]pyridine derivatives: preparation and pharmaceutical applications
US7666880B2 (en) 2005-03-21 2010-02-23 S*Bio Pte Ltd. Imidazo[1,2-A]pyridine derivatives: preparation and pharmaceutical applications
US9856211B2 (en) 2005-05-13 2018-01-02 Topotarget Uk Limited Pharmaceutical formulations of HDAC inhibitors
US8835501B2 (en) 2005-05-13 2014-09-16 Topotarget Uk Limited Pharmaceutical formulations of HDAC inhibitors
US9957227B2 (en) 2005-05-13 2018-05-01 Topotarget Uk Limited Pharmaceutical formulations of HDAC inhibitors
US8138198B2 (en) 2005-05-18 2012-03-20 Angibaud Patrick Rene Substituted aminopropenyl piperidine or morpholine derivatives as novel inhibitors of histone deacetylase
US7741494B2 (en) 2005-07-14 2010-06-22 Takeda San Diego, Inc. Histone deacetylase inhibitors
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
JP2009503117A (en) * 2005-08-04 2009-01-29 サートリス ファーマシューティカルズ, インコーポレイテッド Benzimidazole derivatives as sirtuin modulators
US7709468B2 (en) 2005-09-02 2010-05-04 Abbott Laboratories Imidazo based heterocycles
JP2009507829A (en) * 2005-09-08 2009-02-26 エス*バイオ プライベート リミティッド Heterocyclic compounds
US7884105B2 (en) 2005-10-27 2011-02-08 Janssen Pharmaceutica, N.V. Squaric acid derivatives as inhibitors of histone deacetylase
US9603926B2 (en) 2005-11-10 2017-03-28 Topotarget Uk Limited Histone deacetylase (HDAC) inhibitors for the treatment of cancer
US8828392B2 (en) 2005-11-10 2014-09-09 Topotarget Uk Limited Histone deacetylase (HDAC) inhibitors (PXD101) for the treatment of cancer alone or in combination with chemotherapeutic agent
US8445681B2 (en) 2005-11-15 2013-05-21 Vertex Pharmaceuticals Incorporated Azaindazoles useful as inhibitors of kinases
US8017781B2 (en) 2005-11-15 2011-09-13 Vertex Pharmaceuticals Incorporated Azaindazoles useful as inhibitors of kinases
JP2009521424A (en) * 2005-12-19 2009-06-04 メチルジーン インコーポレイテッド Histone deacetylase inhibitors for enhancing antifungal activity
US9120790B2 (en) 2006-01-17 2015-09-01 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of Janus kinases
US8450489B2 (en) 2006-01-17 2013-05-28 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of janus kinases
US7767816B2 (en) 2006-01-17 2010-08-03 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of janus kinases
US8163917B2 (en) 2006-01-17 2012-04-24 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of Janus kinases
US8822681B2 (en) 2006-01-17 2014-09-02 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of janus kinases
US8119650B2 (en) 2006-01-19 2012-02-21 Janssen Pharmaceutica N.V. Aminophenyl derivatives as novel inhibitors of histone deacetylase
US8664223B2 (en) 2006-01-19 2014-03-04 Janssen Pharmaceutica N.V Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8101616B2 (en) 2006-01-19 2012-01-24 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8114876B2 (en) 2006-01-19 2012-02-14 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8163765B2 (en) 2006-01-19 2012-04-24 Janssen Pharmaceutica N.V. Substituted indolyl-alkyl-amino-derivatives as inhibitors of histone deacetylase
US9078896B2 (en) 2006-01-19 2015-07-14 Janssen Pharmaceutica, N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8530489B2 (en) 2006-12-21 2013-09-10 Vertex Pharmaceuticals Incorporated 5-cyano-4-(pyrrolo [2,3B] pyridine-3-yl)-pyrimidine derivatives useful as protein kinase inhibitors
US8962642B2 (en) 2006-12-21 2015-02-24 Vertex Pharmaceuticals Incorporated 5-cyano-4- (pyrrolo [2,3B] pyridine-3-yl) -pyrimidine derivatives useful as protein kinase inhibitors
US8247421B2 (en) 2006-12-21 2012-08-21 Vertex Pharmaceuticals Incorporated 5-cyano-4-(pyrrolo [2,3B] pyridine-3-yl)-pyrimidine derivatives useful as protein kinase inhibitors
US7723332B2 (en) 2007-03-06 2010-05-25 Wyeth Llc Aryl sulfonamides useful for modulation of the progesterone receptor
US8138168B1 (en) 2007-09-26 2012-03-20 Takeda Pharmaceutical Company Limited Renin inhibitors
US8476289B2 (en) 2008-03-27 2013-07-02 Janssen Pharmaceutica Nv Aza-bicyclohexyl substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylace
US8518911B2 (en) 2008-08-05 2013-08-27 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyridines as MARK inhibitors
US9808459B2 (en) 2009-06-17 2017-11-07 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10874673B2 (en) 2009-06-17 2020-12-29 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10039762B2 (en) 2009-06-17 2018-08-07 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US9096518B2 (en) 2009-06-22 2015-08-04 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
US8624040B2 (en) 2009-06-22 2014-01-07 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
WO2011003365A1 (en) * 2009-07-10 2011-01-13 天津尚德药缘科技有限公司 Histone deacetylase inhibitors with branched structure synthesized through click chemistry
US8987273B2 (en) 2010-07-28 2015-03-24 Bayer Intellectual Property Gmbh Substituted imidazo[1,2-B]pyridazines
US8871774B2 (en) 2010-12-16 2014-10-28 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US8778931B2 (en) 2010-12-22 2014-07-15 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
US9206185B2 (en) 2011-04-07 2015-12-08 Bayer Intellectual Property Gmbh Imidazopyridazines as Akt kinase inhibitors
US9908878B2 (en) 2011-08-01 2018-03-06 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US9051319B2 (en) 2011-08-01 2015-06-09 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10875855B2 (en) 2011-08-01 2020-12-29 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US9844553B2 (en) 2011-09-22 2017-12-19 Merck Sharp & Dohme Corp. Pyrazolopyridyl compounds as aldosterone synthase inhibitors
US9351973B2 (en) 2011-09-22 2016-05-31 Merck Sharp & Dohme Corp. Pyrazolopyridyl compounds as aldosterone synthase inhibitors
WO2013043520A1 (en) 2011-09-22 2013-03-28 Merck Sharp & Dohme Corp. Triazolopyridyl compounds as aldosterone synthase inhibitors
WO2014055595A1 (en) 2012-10-05 2014-04-10 Merck Sharp & Dohme Corp. Indoline compounds as aldosterone synthase inhibitiors related applications
US9771361B2 (en) 2013-11-13 2017-09-26 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US10023569B2 (en) 2013-11-13 2018-07-17 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US11345700B2 (en) 2013-11-13 2022-05-31 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US10640501B2 (en) 2013-11-13 2020-05-05 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US10016401B2 (en) 2014-09-17 2018-07-10 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10231957B2 (en) 2014-09-17 2019-03-19 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10533004B2 (en) 2015-05-13 2020-01-14 Vertex Pharmaceuticals Incorporated Methods of preparing inhibitors of influenza viruses replication
US10273233B2 (en) 2015-05-13 2019-04-30 Vertex Pharmaceuticals Incorporated Inhibitors of influenza viruses replication
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
US11891382B2 (en) 2017-04-26 2024-02-06 Basilea Pharmaceutica International AG Processes for the preparation of furazanobenzimidazoles and crystalline forms thereof
US11168093B2 (en) 2018-12-21 2021-11-09 Celgene Corporation Thienopyridine inhibitors of RIPK2

Also Published As

Publication number Publication date
CA2518318A1 (en) 2004-09-30
WO2004082638A3 (en) 2005-05-06
US7381825B2 (en) 2008-06-03
JP2006520796A (en) 2006-09-14
US20040254220A1 (en) 2004-12-16
US7375228B2 (en) 2008-05-20
EP1608628A2 (en) 2005-12-28
US7169801B2 (en) 2007-01-30
US20040266769A1 (en) 2004-12-30
US20050137232A1 (en) 2005-06-23
KR20050122210A (en) 2005-12-28

Similar Documents

Publication Publication Date Title
US7381825B2 (en) Histone deacetylase inhibitors
US7399884B2 (en) Histone deacetylase inhibitors
US7250514B1 (en) Histone deacetylase inhibitors
US20050159470A1 (en) Histone deacetylase inhibitors
US20050137234A1 (en) Histone deacetylase inhibitors
US7642275B2 (en) Histone deacetylase inhibitors
US7642253B2 (en) Histone deacetylase inhibitors
US20070173527A1 (en) Histone deacetylase inhibitors
US7741494B2 (en) Histone deacetylase inhibitors
EP1753730A1 (en) Dipeptidyl peptidase inhibitors
US20100093767A1 (en) Mitotic Kinase Inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2518318

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006507336

Country of ref document: JP

Ref document number: 1020057017527

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004757631

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20048118722

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004757631

Country of ref document: EP

Ref document number: 1020057017527

Country of ref document: KR

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)