US20080114037A1 - Histone deacetylase inhibitors - Google Patents

Histone deacetylase inhibitors Download PDF

Info

Publication number
US20080114037A1
US20080114037A1 US11/932,727 US93272707A US2008114037A1 US 20080114037 A1 US20080114037 A1 US 20080114037A1 US 93272707 A US93272707 A US 93272707A US 2008114037 A1 US2008114037 A1 US 2008114037A1
Authority
US
United States
Prior art keywords
alkyl
hetero
cycloalkyl
bicycloaryl
bicycloalkyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/932,727
Inventor
Jerome C. Bressi
Anthony R. Gangloff
Lily Kwok
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda California Inc
Original Assignee
Takeda San Diego 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 Takeda San Diego Inc filed Critical Takeda San Diego Inc
Priority to US11/932,727 priority Critical patent/US20080114037A1/en
Publication of US20080114037A1 publication Critical patent/US20080114037A1/en
Assigned to TAKEDA SAN DIEGO, INC. reassignment TAKEDA SAN DIEGO, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRESSI, JEROME C, GANGLOFF, ANTHONY R, KWOK, LILY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/16Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • C07D249/18Benzotriazoles
    • 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
    • C07D401/12Heterocyclic 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 chain containing hetero atoms as chain links
    • 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
    • C07D403/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to compounds that may be used to inhibit histone deacetylases (HDACs), as well as compositions of matter and kits comprising these compounds.
  • HDACs histone deacetylases
  • the invention also relates to methods for inhibiting HDACs and treatment methods using compounds according to the present invention.
  • the present invention relates to compounds, compositions of matter, kits and methods used to inhibit Class I HDACs, such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • 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 H1, H2A, H 2 B, H3, and H4.
  • the amino acid sequences of H2A, H 2 B, H3, and H4 show remarkable conservation between species, wherein H1 varies somewhat and in some cases is replaced by another histone, e.g., H5.
  • H2A, H 2 B, 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.
  • 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.
  • 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.
  • the —(CH 2 ) 4 —NH 2 side chain may be acetylated, for example by an acetyltransferase enzyme to give the amide —(CH 2 ) 4 —NHC( ⁇ O)CH 3 .
  • Methylation, acetylation, and phosphorylation of amino termini of histones that extend from the nucleosomal core affect chromatin structure and gene expression. Spencer and Davie 1999. Gene 240:11-12.
  • Acetylation and deacetylation of histones are 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 acetyltransferases (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 acetyltransferases
  • deacetylases histone deacetylases or HDACs
  • HDACs histone deacetylases
  • 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 Inst. (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.
  • 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. 1 (3):194-202).
  • HDAC inhibitors are useful in the treatment or prevention of protozoal diseases (U.S. Pat. No. 5,922,837) and psoriasis (PCT Publication No. WO 02/26696).
  • the present invention relates to compounds that have activity for inhibiting histone deacetylases (HDACs).
  • HDACs histone deacetylases
  • the present invention also provides compositions, articles of manufacture and kits comprising these compounds.
  • a pharmaceutical composition that comprises an 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, intraperitoneally, 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 co-administered in slow release dosage forms.
  • the invention is also directed to kits and other articles of manufacture for treating disease states associated with one or more 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 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 one or more HDAC.
  • the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology 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 subject that is converted to the compound in vivo where it inhibits one or more HDAC.
  • a method of inhibiting one or more HDAC comprises contacting an HDAC with a compound according to the present invention.
  • a method of inhibiting one or more HDAC comprises causing a compound according to the present invention to be present in a subject in order to inhibit the HDAC in vivo.
  • a method of inhibiting an HDAC comprises administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits the HDAC in vivo.
  • the compounds of the present invention may be the first or second compounds.
  • a therapeutic method comprises administering a compound according to the present invention.
  • a method of treating a condition in a patient which is known to be mediated by one or more 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 treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state comprising: causing a compound according to the present invention to be present in a subject in a therapeutically effective amount for the disease state.
  • a method for treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state 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.
  • the compounds of the present invention may be the first or second compounds.
  • a method for treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state 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 using a compound according to the present invention in order to manufacture a medicament for use in the treatment of a disease state that is known to be mediated by one or more HDAC, or that is known to be treated by HDAC inhibitors.
  • the present invention is intended to encompass all pharmaceutically acceptable ionized forms (e.g., salts) and solvates (e.g., hydrates) of the compounds, regardless of whether such ionized forms and solvates are specified since it is well know in the art to administer pharmaceutical agents in an ionized or solvated form. It is also noted that unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all possible stereoisomers (e.g., enantiomers or diastereomers depending on the number of chiral centers), independent of whether the compound is present as an individual isomer or a mixture of isomers.
  • prodrugs may also be administered which are altered in vivo and become a compound according to the present invention.
  • 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 to a compound according to the present invention.
  • certain compounds of the present invention may be altered in vivo prior to inhibiting kinases and thus may themselves be prodrugs for another compound.
  • Such prodrugs of another compound may or may not themselves independently have kinase inhibitory activity.
  • FIG. 1 illustrates residues 1-482 of HDAC1 and a Flag tag at both the N- and C-terminus (SEQ ID NO: 1).
  • FIG. 2 illustrates the DNA sequence (SEQ ID NO: 2) that was used to encode SEQ ID NO: 1.
  • FIG. 3 illustrates residues 1-488 of HDAC2 and a 6-histidine tag at the C-terminus (SEQ ID NO: 3).
  • FIG. 4 illustrates the DNA sequence (SEQ ID NO: 4) that was used to encode SEQ ID NO: 3.
  • FIG. 5 illustrates residues 73-845 of HDAC6 and a 6-histidine tag at the C-terminus (SEQ ID NO: 5).
  • FIG. 6 illustrates the DNA sequence (SEQ ID NO: 6) that was used to encode SEQ ID NO: 5.
  • FIG. 7 illustrates residues 1-377 of HDAC8 and a 6-histidine tag at the N-terminus (SEQ ID NO: 7).
  • FIG. 8 illustrates the DNA sequence (SEQ ID NO: 8) that was used to encode SEQ ID NO: 7.
  • Alicyclic means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one, two or more double or triple bonds. Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. The nitrogen atoms can be optionally quarternerized or oxidized and the sulfur atoms can be optionally oxidized.
  • alicyclic moieties include, but are not limited to moieties with C 3-8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cyclooctadiene.
  • C 3-8 rings such as cyclopropyl, cyclohexane, cyclopentane, cyclopentene, cyclopentadiene, cyclohexane, cyclohexene, cyclohexadiene, cycloheptane, cycloheptene, cycloheptadiene, cyclooctane, cyclooctene, and cycloo
  • “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, two or more double or triple bonds.
  • Alkoxy means an oxygen moiety having a further alkyl substituent.
  • the alkoxy groups of the present invention can be optionally substituted.
  • 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.
  • oxaalkyl oxygen
  • aminoalkyl nitrogen atoms between the carbon atoms.
  • C X alkyl and C X-Y 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 6-10 )aryl(C 1-3 )alkyl includes, benzyl, phenethyl, 1-phenylethyl, 3-phenylpropyl, 2-thienylmethyl, 2-pyridinylmethyl and the like).
  • alkenyl means a straight or branched, carbon chain that contains at least one carbon-carbon double bond. Examples of alkenyl include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, and the like.
  • Alkynyl means a straight or branched, carbon chain that contains at least one carbon-carbon triple bond. Examples of alkynyl include ethynyl, propargyl, 3-methyl-1-pentynyl, 2-heptynyl and the like.
  • Alkylene unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical.
  • C X alkylene and C X-Y alkylene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 1-6 alkylene includes methylene (—CH 2 —), ethylene (—CH 2 CH 2 —), trimethylene (—CH 2 CH 2 CH 2 —), tetramethylene (—CH 2 CH 2 CH 2 CH 2 —) 2-butenylene (—CH 2 CH ⁇ CHCH 2 —), 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.
  • Alkenylene means a straight or branched, divalent carbon chain having one or more carbon-carbon double bonds. Examples of alkenylene include ethene-1,2-diyl, propene-1,3-diyl, methylene-1,1-diyl, and the like.
  • Alkynylene means a straight or branched, divalent carbon chain having one or more carbon-carbon triple bonds. Examples of alkynylene include ethyne-1,2-diyl, propyne-1,3-diyl, and the like.
  • Alkylidene means a straight or branched saturated or unsaturated, aliphatic radical connected to the parent molecule by a double bond.
  • C X alkylidene and C X-Y alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain.
  • C 1-6 alkylidene includes methylene ( ⁇ CH 2 ), ethylidene ( ⁇ CHCH 3 ), isopropylidene ( ⁇ C(CH 3 ) 2 ), propylidene ( ⁇ CHCH 2 CH 3 ), allylidene ( ⁇ CH—CH ⁇ CH 2 ), and the like).
  • “Amido” means the radical —NR a C(O)R b where the point of attachment to the molecule is at the nitrogen, and R a and R b are further substituents attached to the nitrogen and the carbon of the carbonyl, respectively.
  • Amino means a nitrogen moiety having two further substituents where, for example, a hydrogen or carbon atom is attached to the nitrogen.
  • representative amino groups include —NH 2 , —NHCH 3 , —N(CH 3 ) 2 , —NHC 1-10 -alkyl, —N(C 1-10 -alkyl) 2 , —NHaryl, —NHheteroaryl, —N(aryl) 2 , —N(heteroaryl) 2 , and the like.
  • the two substituents together with the nitrogen may also form a ring.
  • 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.
  • 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 2 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 polycyclic ring assembly wherein each ring is aromatic or when fused with one or more rings forms an aromatic ring assembly. If one or more ring atoms is not carbon (e.g., N, S), the aryl is a heteroaryl. C X aryl and C X-Y 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.
  • C X bicycloaryl and C X-Y 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.
  • “Bridging ring” as used herein refers to a ring that is bonded to another ring to form a compound having a bicyclic structure where two ring atoms that are common to both rings are not directly bound to each other.
  • Non-exclusive examples of common compounds having a bridging ring include borneol, norbornane, 7-oxabicyclo[2.2.1]heptane, and the like.
  • One or both rings of the bicyclic system may also comprise heteroatoms.
  • Carbamoyl means the radical —OC(O)NR a R b where R a and R b are each independently two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
  • Carbocycle means a ring consisting of carbon atoms.
  • Carbocyclic ketone derivative means a carbocyclic derivative wherein the ring contains a —CO— moiety.
  • 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, aldehydes, amides, esters, and ketones.
  • Carboxamido means the radical —C(O)NR a R b where the point of attachment to the molecule is at the carbon of the carbonyl, and R a and R b are each independently two further substituents on the nitrogen.
  • Carboxy means the radical —CO 2 —. 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 X-Y )cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly.
  • (C 3-10 )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-1-yl, and the like.
  • Cycloalkylene means a divalent saturated or partially unsaturated, monocyclic or polycyclic ring assembly.
  • (C X )cycloalkylene and (C X-Y )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.
  • fused ring refers to a ring that is bonded to another ring to form a compound having a bicyclic structure when the ring atoms that are common to both rings are directly bound to each other.
  • Non-exclusive examples of common fused rings include decalin, naphthalene, anthracene, phenanthrene, indole, furan, benzofuran, quinoline, and the like.
  • Compounds having fused ring systems may be saturated, partially saturated, carbocyclics, heterocyclics, aromatics, heteroaromatics, and the like.
  • 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 (e.g., halo-substituted (C 1-3 )alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-1,1-dichloroethyl, and the like).
  • Heteroatom refers to an atom that is not a carbon atom. Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, and sulfur.
  • Heteroatom moiety includes a moiety where the atom by which the moiety is attached is not a carbon.
  • heteroatom moieties include —N ⁇ , —NR c —, —N + (O—) ⁇ , —O—, —S— or —S(O) 2 —, wherein R c is further substituent.
  • Heterobicycloalkyl means bicycloalkyl, as defined in this application, provided that one or more of the atoms within the ring is a heteroatom.
  • hetero(C 9-12 )bicycloalkyl as used 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.
  • Heterocycloalkylene means cycloalkylene, as defined in this application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom.
  • Heteroaryl means a cyclic aromatic group having five or six ring atoms, wherein at least one ring atom is a heteroatom and the remaining ring atoms are carbon.
  • the nitrogen atoms can be optionally quarternized and the sulfur atoms can be optionally oxidized.
  • Heteroaryl groups of this invention include, but are not limited to, those derived from furan, imidazole, isothiazole, isoxazole, oxadiazole, oxazole, 1,2,3-oxadiazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrroline, thiazole, 1,3,4-thiadiazole, triazole and tetrazole.
  • Heteroaryl also includes, but is not limited to, bicyclic or tricyclic rings, wherein the heteroaryl ring is fused to one or two rings independently selected from the group consisting of an aryl ring, a cycloalkyl ring, a cycloalkenyl ring, and another monocyclic heteroaryl or heterocycloalkyl ring.
  • bicyclic or tricyclic heteroaryls include, but are not limited to, those derived from benzo[b]furan, benzo[b]thiophene, benzimidazole, imidazo[4,5-c]pyridine, quinazoline, thieno[2,3-c]pyridine, thieno[3,2-b]pyridine, thieno[2,3-b]pyridine, indolizine, imidazo[1,2a]pyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[1,5-a]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrimidine, imidazo[1,2-c]pyrimidine, imidazo[1,5-a]pyrimidine,
  • the bicyclic or tricyclic heteroaryl rings can be attached to the parent molecule through either the heteroaryl group itself or the aryl, cycloalkyl, cycloalkenyl or heterocycloalkyl group to which it is fused.
  • the heteroaryl groups of this invention can be substituted or unsubstituted.
  • Heterobicycloaryl means bicycloaryl, as defined in this application, provided that one or more of the atoms within the ring is a heteroatom.
  • hetero(C 4-12 )bicycloaryl as used in this application includes, but is not limited to, 2-amino-4-oxo-3,4-dihydropteridin-6-yl, tetrahydroisoquinolinyl, and the like.
  • Heterocycloalkyl means cycloalkyl, as defined in this application, provided that one or more of the atoms forming the ring is a heteroatom selected, independently from N, O, or S,
  • Non-exclusive examples of heterocycloalkyl include piperidyl, 4-morpholyl, 4-piperazinyl, pyrrolidinyl, perhydropyrrolizinyl, 1,4-diazaperhydroepinyl, 1,3-dioxanyl, 1,4-dioxanyl and the like.
  • Haldroxy or “hydroxyl” means the radical —OH.
  • IC 50 means the molar concentration of an inhibitor that produces 50% inhibition of the target enzyme.
  • Iminoketone derivative means a derivative comprising the moiety —C(NR)—, wherein R comprises a hydrogen or carbon atom attached 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 “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 n-1 enantiomeric pairs, where n is the number of chiral centers.
  • Compounds with more than one chiral center may exist as ether an individual diastereomer or as a mixture of diastereomers, termed a “diastereomeric mixture.”
  • 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).
  • Leaving group means a moiety that can be displaced by another moiety, such as by nucleophilic attack, during a chemical reaction. Leaving groups are well known in the art and include, for example, halides and OSO 2 R′ where R′ is, for example, alkyl, haloalkyl, or aryl optionally substituted by halo, alkyl, alkoxy, amino, and the like. Non-limiting examples of leaving groups include chloro, bromo, iodo, mesylate, tosylate, and other similar groups.
  • Niro means the radical —NO 2 .
  • Oxo means the radical ⁇ O.
  • 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 2-6 )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 compounds 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, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzen
  • 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.
  • 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, 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.
  • 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.
  • Ring means a carbocyclic or a heterocyclic system.
  • “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, (C 1-10 )alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocyclyl, carboxyl, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, oxo, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted.
  • “Sulfinyl” means the radical —SO—. 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 —SO 2 —. 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 —CS—. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including thioacids, thioamides, thioesters, and thioketones.
  • Treatment or “treating” means any administration of a compound of the present invention and includes:
  • a (C 1 )alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom.
  • a (C 1 )alkyl comprises methyl (i.e., —CH 3 ) as well as —CR a R b R c , where R a , R b , and R c , may each independently be hydrogen or any other substituent where the atom attached to the carbon is a heteroatom or cyano.
  • CF 3 , CH 2 OH and CH 2 CN for example, are all (C 1 )alkyls.
  • the present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit histone deacetylases (HDACs) and, in particular, Class I HDACs such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • HDACs histone deacetylases
  • Class I HDACs such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • HDACs have been categorized into three 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.
  • NP_004955 Histone deacetylase a regulator of transcription, Wolffe, A. P., Science 272 (5260), 371-372 (1996) 2 NP_001518 Isolation and mapping of a human gene (RPD3L1) that is homologous to RPD3, a transcription factor in Saccharomyces cerevisiae; Furukawa, Y., Kawakami, T., Sudo, K., Inazawa, J., Matsumine, A., Akiyama, T. and Nakamura, Y., Cytogenet. Cell Genet.
  • HDAC11 a Novel Member of the Human Histone Deacetylase Family, Gao, L., Cueto, M. A., Asselbergs, F. and Atadja, P., J. Biol. Chem. 277 (28), 25748-25755 (2002)
  • Sir2-like proteins metabolize NAD and may have protein ADP- ribosyltransferase activity; Frye, R. A.; Biochem. Biophys. Res. Commun. 260 (1), 273-279 (1999) Sirtuin 2 NP_085096/ A ‘double adaptor’ method for improved shotgun NP_036369 library construction; Andersson, B., Wentland, M. A., Spainfrente, J. Y., Liu, W. and Gibbs, R.
  • Sir2-like proteins metabolize NAD and may have protein ADP- ribosyltransferase activity; Frye, R. A.; Biochem. Biophys. Res. Commun.
  • Class I HDACs are Class I HDACs. All Class I HDACs appear to be sensitive to inhibition by trichostatin A (TSA). Of particular note HDAC2 and HDAC8, proteins whose crystal structures Applicants determined and used in conjunction with arriving at the present invention.
  • TSA trichostatin A
  • HDAC2 is a 488 residue, 55 kDa 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 HDAC2 is described in GenBank Accession Number NM 001527, Furukawa, Y. et al., Cryogenet. Cell Genet., 73 (1-2), 130-133 (1996).
  • Zn 2+ is likely native to the protein and required for HDAC2 activity.
  • HDAC8 is a 377 residue, 42 kDa 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, J. J. et al., Biochem. J., 350 (Pt 1), 199-205 (2000).
  • Zn 2+ is likely native to the protein and required for HDAC8 activity.
  • the compounds of the present invention may also possess inhibitory activity for other HDAC family members and thus may be used to address disease states associated with these other family members.
  • HDAC2 was found to adopt an open-faced ⁇ / ⁇ structure consisting of 8 central parallel ⁇ -sheets sandwiched between 12 ⁇ -helices.
  • the ligand binding cleft lies almost in the plane of the central ⁇ -sheet, and is formed primarily by loops emanating from the carboxy-terminal ends of the ⁇ -strands comprising the sheet.
  • Residues which form loop regions extending between ⁇ -strand 1 and ⁇ -helix 1 and between ⁇ -helix 4 and ⁇ -helix 5 provide key surface interactions with bound ligands.
  • Residues which form loop regions extending between ⁇ -strand 3 and ⁇ -helix 6 and between ⁇ -strand 4 and ⁇ -helix 7 and between ⁇ -strand 8 and ⁇ -helix 10 play important roles in defining the shape of the ligand binding pocket, and are involved in a number of key interactions with the bound ligands.
  • 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 comprise:
  • HDAC inhibitors of the present invention comprise:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • m is 0, R 11 is hydrogen, and R 12 is selected from the group consisting of hydroxy, alkoxy, halo, amido, carboxamido, and (C 1-10 )alkylamido, each substituted or unsubstituted.
  • n is 0, p is 2, and R 12 is selected from the group consisting of hydroxy, alkoxy, and halo.
  • R 7 is hydrogen
  • R 12 is alkoxy
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • R 1 and R 2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C 1-10 )alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C 1-10 )alkyl, halo(C 1-10 )alkyl, carbonyl(C 1-3 )alkyl, thiocarbonyl(C 1-3 )alkyl, sulfonyl(C 1-3 )alkyl, sulfinyl(C 1-10 )alkyl, amino(C 1-10 )alkyl and imino(C 1-10 )alkyl, each substituted or unsubstituted.
  • R 1 is hydrogen or a substituent convertible in vivo to hydrogen.
  • R 2 is hydrogen or a substituent convertible in vivo to hydrogen.
  • R 3 is hydrogen or a substituent convertible in vivo to hydrogen.
  • R 4 is selected from the group consisting of hydrogen, halo, aryl and heteroaryl, each substituted or unsubstituted. In yet a further variation of each of the above embodiments and variations, R 4 is selected from the group consisting of phenyl, oxazolyl, thiazolyl, morpholinyl and thiomorpholinyl, each substituted or unsubstituted.
  • R 5 , R 6 , R 7 and R 20 are independently selected from the group consisting of carbonyl, oxo, amino, (C 1-10 )alkylamino, amido, carboxamido, cyano, alkoxy, (C 1-10 )alkyl, aryl, and heteroaryl, each substituted or unsubstituted.
  • R 5 and R 6 are taken together to form a substituted or unsubstituted ring.
  • the ring is an aryl or heteroaryl, each substituted or unsubstituted.
  • the ring is substituted with a substituent selected from the group consisting of halo, alkoxy, amino(C 1-10 )alkoxy, amino(C 1-10 )alkylamino, amino(C 1-10 )alkylsulfanyl, halo(C 1-10 )alkyl, aryl and heteroaryl, each substituted or unsubstituted.
  • the ring is substituted with a substituent selected from the group consisting of thiopheneyl, pyridinyl, furanyl and pyrimidinyl, each substituted or unsubstituted.
  • the ring is substituted with —O—CH 2 CH 2 —NR 13 R 14 .
  • the ring is substituted with —NH—CH 2 CH 2 —NR 13 R 14 .
  • the ring is substituted with —S—CH 2 CH 2 —NR 13 R 14 .
  • R 13 and R 14 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C 1-10 )alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C 1-10 )alkyl, halo(C 1-10 )alkyl, carbonyl(C 1-3 )alkyl, thiocarbonyl(C 1-3 )alkyl, sulfonyl(C 1-3 )alkyl, sulfinyl(C 1-3 )alkyl, amino(C 1-10 )alkyl, imino(C 1-3 )alkyl, (C 3-12 )cycloalkyl(C 1-5 )alkyl, hetero(C 3-12 )cycloalkyl(C 1-5 )alkyl, aryl(C 1-10 )alkyl, heteroary
  • R 13 and R 14 are each independently selected from the group consisting of hydrogen, (C 1-10 )alkyl, (C 3-12 )cycloalkyl, and heteroaryl, each substituted or unsubstituted.
  • R 13 and R 14 are taken together to form a ring selected from the group consisting of morpholinyl, pyrrolidinyl, piperazinyl and thiomorpholinyl, each substituted or unsubstituted.
  • R 8 , R 10 and R 19 are selected from the group consisting of hydrogen and substituted or unsubstituted (C 1-10 )alkyl.
  • L is a linker providing a 1-5 atom separation between the two ring atoms to which L is attached. In yet another variation of each of the above embodiments and variations, L is a linker providing a 1-3 atom separation between the two ring atoms to which L is attached. In a further variation of each of the above embodiments and variations, L is a substituted or unsubstituted alkylene. In yet a further variation of each of the above embodiments and variations, wherein L is —CH 2 —.
  • L is selected from the group consisting of —NR 15 —; —NR 15 —CH 2 —; —O—CH 2 —; —S—CH 2 —; —CH 2 —NR 15 —; —CH 2 —O— and —CH 2 —S—, wherein R 15 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C 1-10 )alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C 1-10 )alkyl, halo(C 1-10 )alkyl, carbonyl(C 1-3 )alkyl, thiocarbonyl(C 1-3 )alkyl, sulfonyl(C 1-3 )alkyl, sulfinyl(C 1-3 )alkyl, amino(C 1
  • a 1 is selected from the group consisting of aryl and heteroaryl, each substituted or unsubstituted. In yet another variation of each of the above embodiments and variations, A 1 is a substituted or unsubstituted phenylene. In a further variation of each of the above embodiments and variations, A 1 is a substituted or unsubstituted 1,4-phenylene.
  • a 1 is selected from the group consisting of a thiophenediyl, furandiyl, pyrrolediyl, thiazolediyl, oxazolediyl, imidazolediyl and pyridinediyl, each substituted or unsubstituted.
  • a 1 comprises:
  • the compounds of the present invention may be in the form of a pharmaceutically acceptable salt, biohydrolyzable ester, biohydrolyzable amide, biohydrolyzable carbamate, solvate, hydrate or prodrug thereof.
  • the compound optionally comprises a substituent that is convertible in vivo to a different substituent such as a hydrogen.
  • the compound may be present in a mixture of stereoisomers (including tautomers), or the compound may comprise a single stereoisomer.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient a compound according to any one of the above embodiments and variations.
  • the composition is a solid formulation adapted for oral administration.
  • the composition is a liquid formulation adapted for oral administration.
  • the composition is a tablet.
  • the composition is a liquid formulation adapted for parenteral administration.
  • compositions comprising a compound according to any one of the above embodiments and variations, wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, 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, and intrathecally.
  • kits comprising a compound of any one of the above embodiments and variations; and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the composition is to be administered, storage information for the composition, dosing information and instructions regarding how to administer the composition.
  • the kit comprises the compound in a multiple dose form.
  • an article of manufacture comprising a compound of any one of the above embodiments and variations; and packaging materials.
  • the packaging material comprises a container for housing the compound.
  • the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the compound.
  • the article of manufacture comprises the compound in a multiple dose form.
  • a therapeutic method comprising administering a compound of any one of the above embodiments and variations to a subject.
  • a method of inhibiting HDAC comprising contacting HDAC with a compound of any one of the above embodiments and variations.
  • a method of inhibiting HDAC comprising causing a compound of any one of the above embodiments and variations to be present in a subject in order to inhibit HDAC in vivo.
  • a method of inhibiting HDAC comprising administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo, the second compound being a compound according to any one of the above embodiments and variations.
  • a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state comprising causing a compound of any one of the above embodiments and variations to be present in a subject in a therapeutically effective amount for the disease state.
  • a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state comprising administering a compound of any one of the above embodiments and variations to a subject, wherein the compound is present in the subject in a therapeutically effective amount for the disease state.
  • a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state comprising administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo, the second compound being a compound according to any one of the above embodiments and variations.
  • the present invention relates to a method for treating cancer comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the cancer is selected from the group consisting of squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, non small-cell lung cancer, bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, small-cell lung cancer, glioma, colorectal cancer, genitourinary cancer and gastrointestinal cancer.
  • the present invention relates to a method for treating inflammation, inflammatory bowel disease, psoriasis, or transplant rejection, comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the present invention relates to a method for treating arthritis comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the present invention relates to a method for treating degenerative diseases of the eye comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the present invention relates to a method for treating multiple sclerosis, amyotrophic lateral sclerosis, thyroid neoplasm or Alzheimer's disease comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the present invention relates to a method for treating hyperproliferative skin diseases or inflammatory cutaneous disorders comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • the histone deacetylase is optionally a Class I histone deacetylase.
  • the histone deacetylase is HDAC2 and/or HDAC8.
  • the present invention relates to methods of making the HDAC inhibitors of the present invention, as well as intermediates useful for the preparation of such HDAC inhibitors.
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • the processes comprise:
  • X is halo. In one particular variation, X is bromo. In another particular variation, X is chloro.
  • the present invention relates to compounds useful in the preparation of the HDAC inhibitors of the present invention.
  • the compounds comprise:
  • the compounds comprise:
  • the compounds comprise:
  • the compounds comprise:
  • the compounds comprise:
  • the compounds comprise:
  • the compounds comprise:
  • the 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; other 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
  • other mineral acids and their corresponding salts such as sulfate, n
  • Further acid addition salts of the present invention include, but are not limited to: adipate, alginate, arginate, aspartate, bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, 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, man
  • 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 (benzathine), dicyclohexylamine, diethanolamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, iso-propylamine, lidocaine, lysine, meglumine, N-methyl
  • Compounds of the present invention that comprise basic nitrogen-containing groups may be quarternerized with such agents as (C 1-4 ) alkyl halides, e.g., methyl, ethyl, iso-propyl and tert-butyl chlorides, bromides and iodides; di(C 1-4 ) alkyl sulfates, e.g., dimethyl, diethyl and diamyl sulfates; (C 10-18 ) alkyl halides, e.g., decyl, dodecyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aryl(C 1-4 ) 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.
  • 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, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0° C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichloromethane
  • 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 that 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., 1,1-acyloxyalkylcarbonochloridate, para-nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et al. (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.
  • Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, 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.
  • the solubility of the compound is usually dependent upon the character of the particular salt form thereof, which it utilized.
  • 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 absorption of the compound.
  • 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.
  • diastereomers can typically be separated by chromatography or by separation/resolution techniques based upon differences in solubility.
  • separation/resolution techniques 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).
  • compositions Comprising HDAC Inhibitors
  • compositions and administration methods may be used in conjunction with the compounds of the present invention.
  • Such compositions may include, in addition to the compounds of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents.
  • the compositions may include active agents in addition to the compounds of the present invention.
  • These additional active agents may include additional compounds according to the invention, and/or one or more other pharmaceutically active agents.
  • compositions may be in gaseous, liquid, semi-liquid or solid form, formulated in a manner suitable for the route of administration to be used.
  • routes of administration for oral administration, capsules and tablets are typically used.
  • parenteral administration reconstitution of a lyophilized powder, prepared as described herein, is typically used.
  • compositions comprising compounds of the present invention may be administered or co-administered orally, parenterally, intraperitoneally, 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 compounds and/or compositions according to the invention may also be administered or co-administered in slow release dosage forms.
  • HDAC inhibitors and compositions comprising them may be administered or co-administered in any conventional dosage form.
  • Co-administration in the context of this invention is intended to mean the administration of more than one therapeutic agent, one of which includes a HDAC inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome.
  • Such co-administration 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,
  • 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.
  • cosolvents such as dimethylsulfoxide (DMSO)
  • surfactants such as TWEEN
  • dissolution in aqueous sodium bicarbonate such as sodium bicarbonate
  • 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 therapeutically active compounds and derivatives thereof 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, polyinylpyrrolidine, 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, polyinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to
  • 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 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 compounds 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 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.
  • compounds 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 monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene 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, polyethylene glycol 4000 and cellulose acetate phthalate.
  • the salt of the 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 compounds 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 compounds 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 examples 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. Examples of 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 acids 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.
  • flavoring agents 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 e.g., 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.
  • 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 compounds of the present invention by parenteral administration generally characterized by subcutaneous, intramuscular or intravenous injection.
  • 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 examples 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
  • thickening and solubilizing agents such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • Examples 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 examples include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection.
  • 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.
  • concentration of an 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 an 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 known and practiced in the art.
  • Injectables may be designed for local and systemic administration.
  • 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 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 the 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.
  • 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 compounds 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 compound 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 the 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 dosage or multiple dosages of the inhibitor.
  • 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 of 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 have diameters of less than 50 microns, preferably less than 10 microns.
  • the 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 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, (polyoxyethylene 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.
  • 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.
  • 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
  • the invention is also directed to kits and other articles of manufacture for treating diseases associated with HDACs. It is noted that diseases are intended to cover all conditions for which the HDACs possess activity that contributes to the pathology and/or symptomatology of the condition.
  • a kit comprising a composition comprising at least one 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 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.
  • 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.
  • kits can be used together in a single package to market a single dosage form.
  • tablets may be contained in a bottle that is in turn contained within a box.
  • the kit includes directions for the administration of the separate components.
  • the kit 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.
  • 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.
  • 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 growth 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 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.
  • therapeutic agents that may be used in combination with HDAC inhibitors include, but are not limited to, anticancer 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.
  • 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.
  • 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, epirubicin, idarubicin and anthracenedione), mitomycin C, bleomycin, dactinomycin, plicatomycin.
  • anthracyclines e.g. doxorubicin, daunorubicin, epirubicin, idarubicin and anthracenedione
  • mitomycin C e.g. doxorubicin, daunorubicin, epirubicin, idarubicin and anthracenedione
  • mitomycin C e.g. doxorubicin, daunorubicin, epirubicin, idarubicin and anthracenedione
  • mitomycin C e.g. doxorubicin
  • 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 chemotherapeutic 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. 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.
  • 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 therapy including a HDAC inhibitor and a hormonal agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these 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 II, 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.
  • IL-2 interleukin 2
  • IL-4 interleukin 4
  • IL-12 interleukin 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 other than cytokines may also be used in conjunction with a HDAC inhibitor to inhibit abnormal cell growth.
  • 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 CD2O 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 Wilms 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.
  • TAA tumor-associated antigens
  • GM2 gangliosides
  • PSA prostate specific antigen
  • AFP alpha-fetoprotein
  • CEA carcinoembryonic antigen
  • breast, lung, gastric, and pancreas cancer s melanoma associated antigens
  • MART-1 gp 100, 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.
  • 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.) and 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.
  • separation/resolution techniques 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).
  • Compounds according to the present invention can also 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.
  • a pharmaceutically acceptable base addition salt of a compound can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds are set forth in the definitions section of this application.
  • the salt forms of the compounds can be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds can be prepared from the corresponding base addition salt or acid addition salt form.
  • a compound in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like).
  • a compound in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
  • 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, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 C.
  • an oxidizing agent e.g., trifluoroperacetic acid, permaleic acid, perbenzoic acid, peracetic acid, meta-chloroperoxybenzoic acid, or the like
  • a suitable inert organic solvent e.g., a halogenated hydrocarbon such as dichloromethane
  • the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
  • Compounds in an unoxidized form can be prepared from N-oxides of compounds by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80° C.
  • a reducing agent e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like
  • an inert organic solvent e.g., acetonitrile, ethanol, aqueous dioxane, or the like
  • Prodrug derivatives of the compounds can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al. (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985).
  • appropriate prodrugs can be prepared by reacting a non-derivatized compound with a suitable carbamylating agent (e.g., 1,1-acyloxyalkylcarbonochloridate, para-nitrophenyl carbonate, or the like).
  • Protected derivatives of the compounds can be made by methods known to those of ordinary skill in the art. A detailed description of the 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.
  • Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds according to the present invention can also be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of compounds, dissociable complexes are preferred (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities.
  • the diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility.
  • the optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization.
  • a more detailed description of the techniques applicable to the resolution of 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).
  • IBCF isobutyl chloroformate
  • i-PrOH isopropanol
  • L liters
  • M molar
  • mCPBA metal-chloroperbenzoic acid
  • Me methyl
  • MeOH methanol
  • MOPS Metalpholinepropanesulfonic acid
  • mp melting point
  • NaOAc sodium acetate
  • OMe methoxy
  • psi pounds per square inch
  • RP reverse phase
  • RT ambient temperature
  • SPA Scintillation Proximity Assay
  • TBAF tetra-n-butylammonium fluoride
  • TBS t-butyldimethylsilyl
  • tBu tert-butyl
  • MS mass spectra
  • 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).
  • ESI electrospray ionization
  • UV detector (220 and 254 nm
  • ELSD evaporative light scattering detector
  • 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).
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as the Aldrich Chemical Company (Milwaukee, Wis.), Bachem (Torrance, Calif.), Sigma (St. Louis, Mo.), or may be prepared by methods well known to a person of ordinary skill in the art, following procedures described in such standard references as Fieser and Fieser's Reagents for Organic Synthesis, vols. 1-17, John Wiley and Sons, New York, N.Y., 1991; Rodd's Chemistry of Carbon Compounds, vols. 1-5 and supps., Elsevier Science Publishers, 1989; Organic Reactions, vols.
  • a solution of B (0.5 mmol) in DMF (2.5 mL) is treated with an appropriate heterocycle A (0.5 mmol) and solid K 2 CO 3 (0.6 mmol).
  • the reaction is poured into water (10 mL) and the resulting solid isolated by filtration.
  • the filter cake can be rinsed with water and allowed to dry in vacuo to yield a mixture of regioisomeric alkylation products C, D and E, which can be separated by flash chromatography. If no solid is formed, the DMF/water mixture can be extracted with ethylacetate.
  • the combined extracts can be dried (MgSO 4 ), concentrated in vacuo and purified by flash chromatography.
  • nitrobenzenes J′ and K′ may be prepared as described in connection with Schemes 1 and 2.
  • Chiral components can be separated and purified using any of a variety of techniques known to those skilled in the art.
  • chiral components can be purified using supercritical fluid chromatography (SFC).
  • SFC supercritical fluid chromatography
  • chiral analytical SFC/MS analyses are conducted using a Berger analytical SFC system (AutoChem, Newark, Del.) which consists of a Berger SFC dual pump fluid control module with a Berger FCM 1100/1200 supercritical fluid pump and FCM 1200 modifier fluid pump, a Berger TCM 2000 oven, and an Alcott 718 autosampler.
  • the integrated system can be controlled by BI-SFC Chemstation software version 3.4.
  • Detection can be accomplished with a Waters ZQ 2000 detector operated in positive mode with an ESI interface and a scan range from 200-800 Da with 0.5 second per scan.
  • Chromatographic separations can be performed on a ChiralPak AD-H, ChiralPak AS-H, ChiralCel OD-H, or ChiralCel OJ-H column (5 ⁇ , 4.6 ⁇ 250 mm; Chiral Technologies, Inc. West Chester, Pa.) with 10 to 40% methanol as the modifier and with or without ammonium acetate (10 mM).
  • Any of a variety of flow rates can be utilized including, for example, 1.5 or 3.5 mL/min with an inlet pressure set at 100 bar.
  • sample injection conditions can be used including, for example, sample injections of either 5 or 10 ⁇ L in methanol at 0.1 mg/mL in concentration.
  • preparative chiral separations are performed using a Berger MultiGram II SFC purification system.
  • samples can be loaded onto a ChiralPak AD column (21 ⁇ 250 mm, 10 ⁇ ).
  • the flow rate for separation can be 70 mL/min, the injection volume up to 2 mL, and the inlet pressure set at 130 bar. Stacked injections can be applied to increase the efficiency.
  • the present invention is further exemplified, but not limited by, the following examples that describe the synthesis of particular compounds according to the invention.
  • ESI-MS m/z 343.4 (M+H) + .
  • ESI-MS m/z 343.4 (M+H) + .
  • ESI-MS m/z 400.5 (M+H) + .
  • ESI-MS m/z 400.5 (M+H) + .

Abstract

Compounds, pharmaceutical compositions, kits and methods are provided for use with HDAC that comprise a compound selected from the group consisting of:
Figure US20080114037A1-20080515-C00001
wherein the substituents are as defined herein.

Description

    RELATED APPLICATION
  • This application is a continuation of U.S. application Ser. No. 11/457,260, filed Jul. 13, 2006, which claims the benefit of U.S. Provisional Application No. 60/699,139 filed Jul. 14, 2005; both applications are incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to compounds that may be used to inhibit histone deacetylases (HDACs), as well as compositions of matter and kits comprising these compounds. The invention also relates to methods for inhibiting HDACs and treatment methods using compounds according to the present invention. In particular, the present invention relates to compounds, compositions of matter, kits and methods used to inhibit Class I HDACs, such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • BACKGROUND OF THE 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. There are five main classes of histones H1, H2A, H2B, H3, and H4. The amino acid sequences of H2A, H2B, H3, and H4 show remarkable conservation between species, wherein H1 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.
  • 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.
  • 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 side chain may be acetylated, for example by an acetyltransferase enzyme to give the amide —(CH2)4—NHC(═O)CH3. Methylation, acetylation, and phosphorylation of amino termini of histones that extend from the nucleosomal core affect chromatin structure and gene expression. Spencer and Davie 1999. Gene 240:11-12.
  • Acetylation and deacetylation of histones are 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.
  • 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 acetyltransferases (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.
  • A growing number of histone deacetylases (HDACs) have been identified. HDACs function as part of large multi-protein 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 repressor function.
  • 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 Inst. (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, 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), and hyperproliferative skin disease such as cancerous and precancerous skin lesions, as well as inflammatory cutaneous disorders.
  • 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. 1 (3):194-202). In addition, HDAC inhibitors are useful in the treatment or prevention of protozoal diseases (U.S. Pat. No. 5,922,837) and psoriasis (PCT Publication No. WO 02/26696).
  • Accordingly, 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
  • The present invention relates to compounds that have activity for inhibiting histone deacetylases (HDACs). The present invention also provides compositions, articles of manufacture and kits comprising these compounds.
  • In one embodiment, a pharmaceutical composition is provided that comprises an 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, intraperitoneally, 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 co-administered in slow release dosage forms.
  • The invention is also directed to kits and other articles of manufacture for treating disease states associated with one or more HDAC.
  • 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.
  • 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.
  • 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. In still another embodiment, reactive intermediates are provided for use in conjunction with the synthetic schemes.
  • Also provided are methods for using compounds, compositions, kits and articles of manufacture according to the present invention.
  • In one embodiment, the compounds, compositions, kits and articles of manufacture are used to inhibit one or more HDAC.
  • In another embodiment, the compounds, compositions, kits and articles of manufacture are used to treat a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state.
  • In another embodiment, a compound is administered to a subject wherein HDAC activity within the subject is altered, preferably reduced.
  • In another embodiment, a prodrug of a compound is administered to a subject that is converted to the compound in vivo where it inhibits one or more HDAC.
  • In another embodiment, a method of inhibiting one or more HDAC is provided that comprises contacting an HDAC with a compound according to the present invention.
  • In another embodiment, a method of inhibiting one or more HDAC is provided that comprises causing a compound according to the present invention to be present in a subject in order to inhibit the HDAC in vivo.
  • In another embodiment, a method of inhibiting an 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 the HDAC in vivo. It is noted that the compounds of the present invention may be the first or second compounds.
  • In another embodiment, a therapeutic method is provided that comprises administering a compound according to the present invention.
  • In another embodiment, a method of treating a condition in a patient which is known to be mediated by one or more 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.
  • In another embodiment, a method is provided for treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology of the 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.
  • In another embodiment, a method is provided for treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology 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. It is noted that the compounds of the present invention may be the first or second compounds.
  • In another embodiment, a method is provided for treating a disease state for which one or more HDAC possesses activity that contributes to the pathology and/or symptomatology 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.
  • 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 a disease state that is known to be mediated by one or more HDAC, or that is known to be treated by HDAC inhibitors.
  • It is noted in regard to all of the above embodiments that the present invention is intended to encompass all pharmaceutically acceptable ionized forms (e.g., salts) and solvates (e.g., hydrates) of the compounds, regardless of whether such ionized forms and solvates are specified since it is well know in the art to administer pharmaceutical agents in an ionized or solvated form. It is also noted that unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all possible stereoisomers (e.g., enantiomers or diastereomers depending on the number of chiral centers), independent of whether the compound is present as an individual isomer or a mixture of isomers. Further, unless otherwise specified, recitation of a compound is intended to encompass all possible resonance forms and tautomers. With regard to the claims, the language “compound comprising the formula” is intended to encompass the compound and all pharmaceutically acceptable ionized forms and solvates, all possible stereoisomers, and all possible resonance forms and tautomers unless otherwise specifically specified in the particular claim.
  • It is further noted that prodrugs may also be administered which are altered in vivo and become a compound according to the present invention. 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 to a compound according to the present invention. It is also noted that certain compounds of the present invention may be altered in vivo prior to inhibiting kinases and thus may themselves be prodrugs for another compound. Such prodrugs of another compound may or may not themselves independently have kinase inhibitory activity.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 illustrates residues 1-482 of HDAC1 and a Flag tag at both the N- and C-terminus (SEQ ID NO: 1).
  • FIG. 2 illustrates the DNA sequence (SEQ ID NO: 2) that was used to encode SEQ ID NO: 1.
  • FIG. 3 illustrates residues 1-488 of HDAC2 and a 6-histidine tag at the C-terminus (SEQ ID NO: 3).
  • FIG. 4 illustrates the DNA sequence (SEQ ID NO: 4) that was used to encode SEQ ID NO: 3.
  • FIG. 5 illustrates residues 73-845 of HDAC6 and a 6-histidine tag at the C-terminus (SEQ ID NO: 5).
  • FIG. 6 illustrates the DNA sequence (SEQ ID NO: 6) that was used to encode SEQ ID NO: 5.
  • FIG. 7 illustrates residues 1-377 of HDAC8 and a 6-histidine tag at the N-terminus (SEQ ID NO: 7).
  • FIG. 8 illustrates the DNA sequence (SEQ ID NO: 8) that was used to encode SEQ ID NO: 7.
  • DEFINITIONS
  • Unless otherwise stated, the following terms used in the specification and claims shall have the following meanings for the purposes of this application.
  • “Alicyclic” means a moiety comprising a non-aromatic ring structure. Alicyclic moieties may be saturated or partially unsaturated with one, two or more double or triple bonds. Alicyclic moieties may also optionally comprise heteroatoms such as nitrogen, oxygen and sulfur. The nitrogen atoms can be optionally quarternerized or oxidized and the sulfur atoms can be optionally oxidized. Examples of alicyclic moieties include, but are not limited to moieties with C3-8 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, two or more double or triple bonds.
  • “Alkoxy” means an oxygen moiety having a further alkyl substituent. The alkoxy groups of the present invention can be optionally substituted.
  • “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 CX-Y 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, heteroarylalkyl) 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., (C6-10)aryl(C1-3)alkyl includes, benzyl, phenethyl, 1-phenylethyl, 3-phenylpropyl, 2-thienylmethyl, 2-pyridinylmethyl and the like).
  • “Alkenyl” means a straight or branched, carbon chain that contains at least one carbon-carbon double bond. Examples of alkenyl include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, and the like.
  • “Alkynyl” means a straight or branched, carbon chain that contains at least one carbon-carbon triple bond. Examples of alkynyl include ethynyl, propargyl, 3-methyl-1-pentynyl, 2-heptynyl and the like.
  • “Alkylene”, unless indicated otherwise, means a straight or branched, saturated or unsaturated, aliphatic, divalent radical. CX alkylene and CX-Y 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.
  • “Alkenylene” means a straight or branched, divalent carbon chain having one or more carbon-carbon double bonds. Examples of alkenylene include ethene-1,2-diyl, propene-1,3-diyl, methylene-1,1-diyl, and the like.
  • “Alkynylene” means a straight or branched, divalent carbon chain having one or more carbon-carbon triple bonds. Examples of alkynylene include ethyne-1,2-diyl, propyne-1,3-diyl, and the like.
  • “Alkylidene” means a straight or branched saturated or unsaturated, aliphatic radical connected to the parent molecule by a double bond. CX alkylidene and CX-Y alkylidene are typically used where X and Y indicate the number of carbon atoms in the chain. For example, C1-6 alkylidene includes methylene (═CH2), ethylidene (═CHCH3), isopropylidene (═C(CH3)2), propylidene (═CHCH2CH3), allylidene (═CH—CH═CH2), and the like).
  • “Amido” means the radical —NRaC(O)Rb where the point of attachment to the molecule is at the nitrogen, and Ra and Rb are further substituents attached to the nitrogen and the carbon of the carbonyl, respectively.
  • “Amino” means a nitrogen moiety having two further substituents where, for example, a hydrogen or carbon atom is attached to the nitrogen. For example, representative amino groups include —NH2, —NHCH3, —N(CH3)2, —NHC1-10-alkyl, —N(C1-10-alkyl)2, —NHaryl, —NHheteroaryl, —N(aryl)2, —N(heteroaryl)2, and the like. Optionally, the two substituents together with the nitrogen may also form a ring. 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 (C2-6)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).
  • “Aromatic” means a moiety wherein the constituent atoms make up an unsaturated ring system, all atoms in the ring system are sp2 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 polycyclic ring assembly wherein each ring is aromatic or when fused with one or more rings 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 CX-Y 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 CX-Y 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.
  • “Bridging ring” as used herein refers to a ring that is bonded to another ring to form a compound having a bicyclic structure where two ring atoms that are common to both rings are not directly bound to each other. Non-exclusive examples of common compounds having a bridging ring include borneol, norbornane, 7-oxabicyclo[2.2.1]heptane, and the like. One or both rings of the bicyclic system may also comprise heteroatoms.
  • “Carbamoyl” means the radical —OC(O)NRaRb where Ra and Rb are each independently two further substituents where a hydrogen or carbon atom is attached to the nitrogen.
  • “Carbocycle” means a ring consisting of carbon atoms.
  • “Carbocyclic ketone derivative” means a carbocyclic derivative wherein the ring contains a —CO— moiety.
  • “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, aldehydes, amides, esters, and ketones.
  • “Carboxamido” means the radical —C(O)NRaRb where the point of attachment to the molecule is at the carbon of the carbonyl, and Ra and Rb are each independently two further substituents on the nitrogen.
  • “Carboxy” means the radical —CO2—. 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. (CX)cycloalkyl and (CX-Y)cycloalkyl are typically used where X and Y indicate the number of carbon atoms in the ring assembly. For example, (C3-10)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-1-yl, and the like.
  • “Cycloalkylene” means a divalent saturated or partially unsaturated, monocyclic or polycyclic ring assembly. (CX)cycloalkylene and (CX-Y)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.
  • “Fused ring” as used herein refers to a ring that is bonded to another ring to form a compound having a bicyclic structure when the ring atoms that are common to both rings are directly bound to each other. Non-exclusive examples of common fused rings include decalin, naphthalene, anthracene, phenanthrene, indole, furan, benzofuran, quinoline, and the like. Compounds having fused ring systems may be saturated, partially saturated, carbocyclics, heterocyclics, aromatics, heteroaromatics, and the like.
  • “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 (e.g., halo-substituted (C1-3)alkyl includes chloromethyl, dichloromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, perfluoroethyl, 2,2,2-trifluoro-1,1-dichloroethyl, and the like).
  • “Heteroatom” refers to an atom that is not a carbon atom. Particular examples of heteroatoms include, but are not limited to nitrogen, oxygen, and sulfur.
  • “Heteroatom moiety” includes a moiety where the atom by which the moiety is attached is not a carbon. Examples of heteroatom moieties include —N═, —NRc—, —N+(O—)═, —O—, —S— or —S(O)2—, wherein Rc is further substituent.
  • “Heterobicycloalkyl” means bicycloalkyl, as defined in this application, provided that one or more of the atoms within the ring is a heteroatom. For example hetero(C9-12)bicycloalkyl as used 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.
  • “Heterocycloalkylene” means cycloalkylene, as defined in this application, provided that one or more of the ring member carbon atoms is replaced by a heteroatom.
  • “Heteroaryl” means a cyclic aromatic group having five or six ring atoms, wherein at least one ring atom is a heteroatom and the remaining ring atoms are carbon. The nitrogen atoms can be optionally quarternized and the sulfur atoms can be optionally oxidized. Heteroaryl groups of this invention include, but are not limited to, those derived from furan, imidazole, isothiazole, isoxazole, oxadiazole, oxazole, 1,2,3-oxadiazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrroline, thiazole, 1,3,4-thiadiazole, triazole and tetrazole. “Heteroaryl” also includes, but is not limited to, bicyclic or tricyclic rings, wherein the heteroaryl ring is fused to one or two rings independently selected from the group consisting of an aryl ring, a cycloalkyl ring, a cycloalkenyl ring, and another monocyclic heteroaryl or heterocycloalkyl ring. These bicyclic or tricyclic heteroaryls include, but are not limited to, those derived from benzo[b]furan, benzo[b]thiophene, benzimidazole, imidazo[4,5-c]pyridine, quinazoline, thieno[2,3-c]pyridine, thieno[3,2-b]pyridine, thieno[2,3-b]pyridine, indolizine, imidazo[1,2a]pyridine, quinoline, isoquinoline, phthalazine, quinoxaline, naphthyridine, quinolizine, indole, isoindole, indazole, indoline, benzoxazole, benzopyrazole, benzothiazole, imidazo[1,5-a]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrimidine, imidazo[1,2-c]pyrimidine, imidazo[1,5-a]pyrimidine, imidazo[1,5-c]pyrimidine, pyrrolo[2,3-b]pyridine, pyrrolo[2,3-c]pyridine, pyrrolo[3,2-c]pyridine, pyrrolo[3,2-b]pyridine, pyrrolo[2,3-d]pyrimidine, pyrrolo[3,2-d]pyrimidine, pyrrolo[2,3-b]pyrazine, pyrazolo[1,5-a]pyridine, pyrrolo[1,2-b]pyridazine, pyrrolo[1,2-c]pyrimidine, pyrrolo[1,2-a]pyrimidine, pyrrolo[1,2-a]pyrazine, triazo[1,5-a]pyridine, pteridine, purine, carbazole, acridine, phenazine, phenothiazene, phenoxazine, 1,2-dihydropyrrolo[3,2,1-hi]indole, indolizine, pyrido[1,2-a]indole and 2(1H)-pyridinone. The bicyclic or tricyclic heteroaryl rings can be attached to the parent molecule through either the heteroaryl group itself or the aryl, cycloalkyl, cycloalkenyl or heterocycloalkyl group to which it is fused. The heteroaryl groups of this invention can be substituted or unsubstituted.
  • “Heterobicycloaryl” means bicycloaryl, as defined in this application, provided that one or more of the atoms within the ring is a heteroatom. For example, hetero(C4-12)bicycloaryl as used in this application includes, but is not limited to, 2-amino-4-oxo-3,4-dihydropteridin-6-yl, tetrahydroisoquinolinyl, and the like.
  • “Heterocycloalkyl” means cycloalkyl, as defined in this application, provided that one or more of the atoms forming the ring is a heteroatom selected, independently from N, O, or S, Non-exclusive examples of heterocycloalkyl include piperidyl, 4-morpholyl, 4-piperazinyl, pyrrolidinyl, perhydropyrrolizinyl, 1,4-diazaperhydroepinyl, 1,3-dioxanyl, 1,4-dioxanyl and the like.
  • “Hydroxy” or “hydroxyl” means the radical —OH.
  • “IC50” means the molar concentration of an inhibitor that produces 50% inhibition of the target enzyme.
  • “Iminoketone derivative” means a derivative comprising the moiety —C(NR)—, wherein R comprises a hydrogen or carbon atom attached 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 “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 2n-1 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as ether an individual diastereomer 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).
  • “Leaving group” means a moiety that can be displaced by another moiety, such as by nucleophilic attack, during a chemical reaction. Leaving groups are well known in the art and include, for example, halides and OSO2R′ where R′ is, for example, alkyl, haloalkyl, or aryl optionally substituted by halo, alkyl, alkoxy, amino, and the like. Non-limiting examples of leaving groups include chloro, bromo, iodo, mesylate, tosylate, and other similar groups.
  • “Nitro” means the radical —NO2.
  • “Oxo” means the radical ═O.
  • “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 (C2-6)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 compounds 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, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic 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-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis(3-hydroxy-2-ene-1-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.
  • 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. 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, 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.
  • “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.
  • “Ring” means a carbocyclic or a heterocyclic system.
  • “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, (C1-10)alkyl, alkylene, alkylidene, amide, amino, aminoalkyl, aromatic, aryl, bicycloalkyl, bicycloaryl, carbamoyl, carbocyclyl, carboxyl, carbonyl group, cycloalkyl, cycloalkylene, ester, halo, heterobicycloalkyl, heterocycloalkylene, heteroaryl, heterobicycloaryl, heterocycloalkyl, oxo, hydroxy, iminoketone, ketone, nitro, oxaalkyl, and oxoalkyl moieties, each of which may optionally also be substituted or unsubstituted.
  • “Sulfinyl” means the radical —SO—. 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 —SO2—. 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 —CS—. It is noted that the thiocarbonyl radical may be further substituted with a variety of substituents to form different thiocarbonyl groups including thioacids, thioamides, thioesters, and thioketones.
  • “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 diseased (i.e., arresting further development of the pathology 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).
  • It is noted in regard to all of the definitions provided herein that the definitions should be interpreted as being open ended in the sense that further substituents beyond those specified may be included. Hence, a (C1)alkyl indicates that there is one carbon atom but does not indicate what are the substituents on the carbon atom. Hence, a (C1)alkyl comprises methyl (i.e., —CH3) as well as —CRaRbRc, where Ra, Rb, and Rc, may each independently be hydrogen or any other substituent where the atom attached to the carbon is a heteroatom or cyano. Hence, CF3, CH2OH and CH2CN, for example, are all (C1)alkyls.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to compounds, compositions, kits and articles of manufacture that may be used to inhibit histone deacetylases (HDACs) and, in particular, Class I HDACs such as HDAC1, HDAC2, HDAC6 and HDAC8.
  • 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.
  • HDACs have been categorized into three 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
    GenBank
    HDAC Accession Number Reference
    1 NP_004955 Histone deacetylase: a regulator of transcription,
    Wolffe, A. P., Science 272 (5260), 371-372 (1996)
    2 NP_001518 Isolation and mapping of a human gene (RPD3L1) that
    is homologous to RPD3, a transcription factor in
    Saccharomyces cerevisiae; Furukawa, Y., Kawakami, T.,
    Sudo, K., Inazawa, J., Matsumine, A., Akiyama, T.
    and Nakamura, Y., Cytogenet. Cell Genet. 73 (1-2),
    130-133 (1996)
    3 NP_003874 Isolation and characterization of cDNAs corresponding
    to an additional member of the human histone
    deacetylase gene family, Yang, W. M., Yao, Y. L., Sun, J. M.,
    Davie, J. R. and Seto, E., J. Biol. Chem. 272 (44),
    28001-28007 (1997)
    8 NP_060956 Buggy, J. J., Sideris, M. L., Mak, P., Lorimer, D. D.,
    McIntosh, B. and Clark, J. M.
    Biochem. J. 350 Pt 1, 199-205 (2000)
    11 NP_079103 Cloning and Functional Characterization of HDAC11,
    a Novel Member of the Human Histone Deacetylase
    Family, Gao, L., Cueto, M. A., Asselbergs, F. and
    Atadja, P., J. Biol. Chem. 277 (28), 25748-25755
    (2002)
  • TABLE 2
    CLASS II HDACs
    GenBank
    HDAC Accession Number Reference
    4 NP_006028 Transcriptional control. Sinful repression, Wolffe, A. P.,
    Nature 387 (6628), 16-17 (1997)
    5 NP_631944 Prediction of the coding sequences of unidentified
    human genes. IX. The complete sequences of 100 new
    cDNA clones from brain which can code for large
    proteins in vitro, Nagase, T., Ishikawa, K., Miyajima, N.,
    Tanaka, A., Kotani, H., Nomura, N. and Ohara, O.,
    DNA Res. 5 (1), 31-39 (1998)
    6 NP_006035 Transcriptional control. Sinful repression, Wolffe, A. P.,
    Nature 387 (6628), 16-17 (1997)
    7 NP_057680 Isolation of a novel histone deacetylase reveals that
    class I and class II deacetylases promote SMRT-
    mediated repression, Kao, H. Y., Downes, M.,
    Ordentlich, P. and Evans, R. M., Genes Dev. 14 (1),
    55-66 (2000)
    9 NP_478056 MEF-2 function is modified by a novel co-repressor,
    MITR, Sparrow, D. B., Miska, E. A., Langley, E.,
    Reynaud-Deonauth, S., Kotecha, S., Towers, N.,
    Spohr, G., Kouzarides, T. and Mohun, T. J., EMBO J.
    18 (18), 5085-5098 (1999)
    10 NP_114408 Isolation and characterization of mammalian
    HDAC10, a novel histone deacetylase, Kao, H. Y., Lee, C. H.,
    Komarov, A., Han, C. C. and Evans, R. M., J.
    Biol. Chem. 277 (1), 187-193 (2002)
  • TABLE 3
    CLASS III HDACs
    GenBank
    HDAC Accession Number Reference
    Sirtuin 1 NP_036370 Characterization of five human cDNAs with homology
    to the yeast SIR2 gene: Sir2-like proteins (sirtuins)
    metabolize NAD and may have protein ADP-
    ribosyltransferase activity; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 260 (1), 273-279 (1999)
    Sirtuin 2 NP_085096/ A ‘double adaptor’ method for improved shotgun
    NP_036369 library construction; Andersson, B., Wentland, M. A.,
    Ricafrente, J. Y., Liu, W. and Gibbs, R. A.; Anal.
    Biochem. 236 (1), 107-113 (1996)
    Sirtuin 3 NP_036371 Characterization of five human cDNAs with homology
    to the yeast SIR2 gene: Sir2-like proteins (sirtuins)
    metabolize NAD and may have protein ADP-
    ribosyltransferase activity; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 260 (1), 273-279 (1999)
    Sirtuin 4 NP_036372 Characterization of five human cDNAs with homology
    to the yeast SIR2 gene: Sir2-like proteins (sirtuins)
    metabolize NAD and may have protein ADP-
    ribosyltransferase activity; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 260 (1), 273-279 (1999)
    Sirtuin 5 NP_112534/ Characterization of five human cDNAs with homology
    NP_036373 to the yeast SIR2 gene: Sir2-like proteins (sirtuins)
    metabolize NAD and may have protein ADP-
    ribosyltransferase activity; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 260 (1), 273-279 (1999)
    Sirtuin 6 NP_057623 Phylogenetic classification of prokaryotic and
    eukaryotic Sir2-like proteins; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 273 (2), 793-798 (2000)
    Sirtuin 7 NP_057622 Phylogenetic classification of prokaryotic and
    eukaryotic Sir2-like proteins; Frye, R. A.; Biochem.
    Biophys. Res. Commun. 273 (2), 793-798 (2000)
  • Of particular note are Class I HDACs. All Class I HDACs appear to be sensitive to inhibition by trichostatin A (TSA). Of particular note HDAC2 and HDAC8, proteins whose crystal structures Applicants determined and used in conjunction with arriving at the present invention.
  • HDAC2 is a 488 residue, 55 kDa 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 HDAC2 is described in GenBank Accession Number NM 001527, Furukawa, Y. et al., Cryogenet. Cell Genet., 73 (1-2), 130-133 (1996). Zn2+ is likely native to the protein and required for HDAC2 activity.
  • HDAC8 is a 377 residue, 42 kDa 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, J. J. et al., Biochem. J., 350 (Pt 1), 199-205 (2000). Zn2+ is likely native to the protein and required for HDAC8 activity.
  • It is noted that the compounds of the present invention may also possess inhibitory activity for other HDAC family members and thus may be used to address disease states associated with these other family members.
  • Crystal Structure of Histone Deacetylase
  • Syrrx, Inc. (now Takeda San Diego, Inc.) in San Diego, Calif. solved the crystal structure for HDAC2. HDAC2 was found to adopt an open-faced α/β structure consisting of 8 central parallel β-sheets sandwiched between 12 α-helices. The ligand binding cleft lies almost in the plane of the central β-sheet, and is formed primarily by loops emanating from the carboxy-terminal ends of the β-strands comprising the sheet. Residues which form loop regions extending between β-strand 1 and α-helix 1 and between α-helix 4 and α-helix 5, provide key surface interactions with bound ligands. Residues which form loop regions extending between β-strand 3 and α-helix 6 and between β-strand 4 and α-helix 7 and between β-strand 8 and α-helix 10 play important roles in defining the shape of the ligand binding pocket, and are involved in a number of key interactions with the bound ligands.
  • 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/
    Figure US20080114037A1-20080515-P00001
    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.
  • Knowledge of the crystal structures was used to guide the design of the HDAC inhibitors provided herein.
  • HDAC Inhibitors
  • In one embodiment, HDAC inhibitors of the present invention comprise:
  • Figure US20080114037A1-20080515-C00002
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atom to which L is attached;
      • J1 is selected from the group consisting of —CR7R7′— and —NR19—;
      • J2 is selected from the group consisting of —CR20R20′ and —NR10—;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-2)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 are taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7, and R7′, R20 and R20′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R5, R6, R7 and R20, are each independently absent when the C to which they are bound is bound to L, and R5′, R6′, R7′ and R20′, are each independently absent when the C to which they are bound form part of a double bond; and
      • R8, R10 and R19 are each individually selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′, R20, R20′, R10 and R19 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7, R7′, R20, R20′ and R19 may be taken together to form a substituted or unsubstituted ring, or R19 and any one of R5, R5′, R6, R6′, R20 and R20′, may be taken together to form a substituted or unsubstituted ring, provided that R8, R10 and R19 are each independently absent when the N to which they are bound is bound to L, and R8, R10 and R11 are each independently absent when the N to which they are bound form part of a double bond.
  • In another embodiment, HDAC inhibitors of the present invention comprise:
  • Figure US20080114037A1-20080515-C00003
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5, R6 and R7 are each independently absent when the C to which they are bound is bound to L, and R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond; and
      • R8 and R10 are each individually selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R5 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 and R10 are each independently absent when the N to which they are bound is bound to L, and R8 and R10 are absent when the N to which they are bound form part of a double bond.
  • In another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00004
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond; and
      • R8 and R10 are each individually selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 and R10 are each independently absent when the N to which they are bound form part of a double bond.
  • In yet another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00005
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond;
      • R8 and R10 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 and R10 are each independently absent when the N to which they are bound form part of a double bond; and
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00006
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond;
      • R8 and R10 are independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 and R10 are absent when the N to which they are bound form part of a double bond; and
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and any one of R3, R5, R5′, R7, R7′, R8 and R10 may be taken together to form a substituted or unsubstituted ring.
  • In a further embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00007
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond;
      • R8 and R10 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 or R10 are each independently absent when the N to which it is bound form part of a double bond;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and any one of R3, R5, R5′, R7, R7′, R8 and R10 may be taken together to form a substituted or unsubstituted ring; and
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring.
  • In still a further embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00008
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7 and R7′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′ and R7′ are each independently absent when the C to which they are bound form part of a double bond;
      • R8 and R10 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′ and R10 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7 and R7′ may be taken together to form a substituted or unsubstituted ring, provided that R8 is absent when the N to which it is bound forms part of a double bond;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and any one of R5, R5′, R7, R7′, R8 and R10 may be taken together to form a substituted or unsubstituted ring; and
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring.
  • In yet a further embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00009
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R7 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00010
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • L is a linker providing a 0-6 atom separation between the two rings atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and R3 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00011
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and R3 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In yet another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00012
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R7 or R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In one variation of each of the above embodiments, m is 0, R11 is hydrogen, and R12 is selected from the group consisting of hydroxy, alkoxy, halo, amido, carboxamido, and (C1-10)alkylamido, each substituted or unsubstituted.
  • In yet another variation, n is 0, p is 2, and R12 is selected from the group consisting of hydroxy, alkoxy, and halo.
  • In still another variation, R7 is hydrogen, and R12 is alkoxy.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00013
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6 and R6′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, and R6′ may be taken together to form a substituted or unsubstituted ring, provided that R5′ and R6′ are each independently absent when the C to which they are bound form part of a double bond; and
      • R8, R10 and R19, are each individually selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R10 and R19 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′ and R19 may be taken together to form a substituted or unsubstituted ring, or R19 and any one of R5, R5′, R6 and R6′ may be taken together to form a substituted or unsubstituted ring, provided that R8, R10 and R19 are each independently absent when the N to which they are bound forms part of a double bond.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00014
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6 and R6′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6 and R6′ may be taken together to form a substituted or unsubstituted ring, provided that R5′ and R6′ are each independently absent when the C to which they are bound form part of a double bond;
      • R8, R10 and R19 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R10 and R19 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′ and R19 may be taken together to form a substituted or unsubstituted ring, or R19 and any one of R5, R5′, R6 and R6′ may be taken together to form a substituted or unsubstituted ring, provided that R8, R10 and R19 are each independently absent when the N to which they are bound forms part of a double bond.
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and any one of R3, R5, R5′, R8, R10 and R19 may be taken together to form a substituted or unsubstituted ring; and
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00015
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and R3 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R11 may be taken together to form a substituted or unsubstituted.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00016
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamide, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R11 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00017
  • wherein:
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7, R7′, R20 and R20′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′, R7′ and R20′, are each independently absent when the C to which they are bound form part of a double bond; and
      • R8 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R8 is absent when the N to which it is bound forms part of a double bond.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00018
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R5, R5′, R6, R6′, R7, R7′, R20 and R20′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R5′, R6′, R7′ and R20′, are each independently absent when the C to which they are bound form part of a double bond; and
      • R8 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R8 and any one of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R5 is absent when the N to which it is bound forms part of a double bond;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and any one of R3, R5, R5′, R6, R6′, R7, R7′, R20, R20′ and R8 may be taken together to form a substituted or unsubstituted ring; and
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00019
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 or one R9 and R3 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R11 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, HDAC inhibitors of the present invention comprise a compound selected from the group consisting of:
  • Figure US20080114037A1-20080515-C00020
  • wherein:
      • m is selected from the group consisting of 0, 1, 2, 3 and 4;
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R9 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two R9 may be taken together to form a substituted or unsubstituted ring;
      • R11 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R11 and R9 may be taken together to form a substituted or unsubstituted ring; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R11 may be taken together to form a substituted or unsubstituted ring.
  • In one variation of each of the above embodiments, R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-10)alkyl, amino(C1-10)alkyl and imino(C1-10)alkyl, each substituted or unsubstituted.
  • In another variation of each of the above embodiments and variations, R1 is hydrogen or a substituent convertible in vivo to hydrogen. In still another variation of each of the above embodiments and variations, R2 is hydrogen or a substituent convertible in vivo to hydrogen. In yet another variation of each of the above embodiments and variations, R3 is hydrogen or a substituent convertible in vivo to hydrogen.
  • In a further variation of each of the above embodiments and variations, R4 is selected from the group consisting of hydrogen, halo, aryl and heteroaryl, each substituted or unsubstituted. In yet a further variation of each of the above embodiments and variations, R4 is selected from the group consisting of phenyl, oxazolyl, thiazolyl, morpholinyl and thiomorpholinyl, each substituted or unsubstituted.
  • In still a further variation of each of the above embodiments and variations, R5, R6, R7 and R20 are independently selected from the group consisting of carbonyl, oxo, amino, (C1-10)alkylamino, amido, carboxamido, cyano, alkoxy, (C1-10)alkyl, aryl, and heteroaryl, each substituted or unsubstituted.
  • In another variation of each of the above embodiments and variations, R5 and R6 are taken together to form a substituted or unsubstituted ring. In one variation, the ring is an aryl or heteroaryl, each substituted or unsubstituted. In another variation, the ring is substituted with a substituent selected from the group consisting of halo, alkoxy, amino(C1-10)alkoxy, amino(C1-10)alkylamino, amino(C1-10)alkylsulfanyl, halo(C1-10)alkyl, aryl and heteroaryl, each substituted or unsubstituted. In yet another variation, the ring is substituted with a substituent selected from the group consisting of thiopheneyl, pyridinyl, furanyl and pyrimidinyl, each substituted or unsubstituted.
  • In still another variation, the ring is substituted with —O—CH2CH2—NR13R14. In a further variation, the ring is substituted with —NH—CH2CH2—NR13R14. In yet a further variation, the ring is substituted with —S—CH2CH2—NR13R14. In each of these variations, R13 and R14 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero (C8-12)bicycloaryl(C1-15)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R13 and R14 may be taken together to form a substituted or unsubstituted ring. In other embodiments, R13 and R14 are each independently selected from the group consisting of hydrogen, (C1-10)alkyl, (C3-12)cycloalkyl, and heteroaryl, each substituted or unsubstituted. In still other embodiments, R13 and R14 are taken together to form a ring selected from the group consisting of morpholinyl, pyrrolidinyl, piperazinyl and thiomorpholinyl, each substituted or unsubstituted.
  • In another variation of each of the above embodiments and variations, R8, R10 and R19 are selected from the group consisting of hydrogen and substituted or unsubstituted (C1-10)alkyl.
  • In still another variation of each of the above embodiments and variations, L is a linker providing a 1-5 atom separation between the two ring atoms to which L is attached. In yet another variation of each of the above embodiments and variations, L is a linker providing a 1-3 atom separation between the two ring atoms to which L is attached. In a further variation of each of the above embodiments and variations, L is a substituted or unsubstituted alkylene. In yet a further variation of each of the above embodiments and variations, wherein L is —CH2—. In another variation of each of the above embodiments and variations, L is selected from the group consisting of —NR15—; —NR15—CH2—; —O—CH2—; —S—CH2—; —CH2—NR15—; —CH2—O— and —CH2—S—, wherein R15 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted.
  • In still another variation of each of the above embodiments and variations, A1 is selected from the group consisting of aryl and heteroaryl, each substituted or unsubstituted. In yet another variation of each of the above embodiments and variations, A1 is a substituted or unsubstituted phenylene. In a further variation of each of the above embodiments and variations, A1 is a substituted or unsubstituted 1,4-phenylene. In still a further variation of each of the above embodiments and variations, A1 is selected from the group consisting of a thiophenediyl, furandiyl, pyrrolediyl, thiazolediyl, oxazolediyl, imidazolediyl and pyridinediyl, each substituted or unsubstituted.
  • In another variation of each of the above embodiments and variations, A1 comprises:
  • Figure US20080114037A1-20080515-C00021
  • wherein:
      • each X is independently selected from the group consisting of CR16 and N; and
      • each R16 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R16 is absent when the C to which it is bound is further bound to L or the carbonyl adjacent to A1.
  • Particular examples of compounds according to the present invention include, but are not limited to:
    • 4-((1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-acetamido-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((5-nitro-1H-indazol-1-yl)methyl)benzamide;
    • 4-((5-benzamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((5-nitro-2H-indazol-2-yl)methyl)benzamide;
    • ((5-amino-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-benzamido-2H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 2-Methoxyethyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate;
    • Methyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate;
    • 2-Methoxyethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate;
    • Methyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate;
    • Benzyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate;
    • Benzyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate;
    • N-(4-Amino-biphenyl-3-yl)-4-(5-nitro-indazol-2-ylmethyl)-benzamide;
    • N-(2-(4-((2-Aminophenyl)carbamoyl)benzyl)-2H-indazol-5-yl)morpholine-4-carboxamide;
    • 5-((2H-Indazol-2-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide;
    • N-(2-Aminophenyl)-5-((5-nitro-2H-indazol-2-yl)methyl)thiophene-2-carboxamide;
    • N-(1-(4-((2-Aminophenyl)carbamoyl)benzyl)-1H-indazol-5-yl)morpholine-4-carboxamide;
    • 2-Morpholinoethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate;
    • Pyridin-3-ylmethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate;
    • 5-((1H-Indazol-1-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide;
    • 4-((2H-Pyrazolo[3,4-b]pyridin-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(4-Amino-biphenyl-3-yl)-4-pyrazolo[3,4-b]pyridin-2-ylmethyl-benzamide;
    • N-(4-Amino-biphenyl-3-yl)-4-indazol-2-ylmethyl-benzamide;
    • N-(2-Aminophenyl)-4-((3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-Aminophenyl)-4-((3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • 4-((1H-indazol-3-ylamino)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-(4-((2-Aminophenyl)carbamoyl)benzyl)-2H-indazol-5-yl)morpholine-4-carboxamide;
    • Methyl 3-(4-((2H-indazol-2-yl)methyl)benzamido)-4-aminobenzoate;
    • N-(2-aminophenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-fluoro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-fluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-fluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-fluoro-2H-indazol-2-yl)methyl)benzamide;
    • 4-(1-(1H-indazol-1-yl)propan-2-yl)-N-(2-aminophenyl)benzamide;
    • 4-(2-(1H-indazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • 4-(2-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((4-chloro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-chloro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4,6-difluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4,6-difluoro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((7-fluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((7-fluoro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-fluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-fluoro-2H-indazol-2-yl)methyl)benzamide;
    • (R)-4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • (S)-4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((7-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((7-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5,6-difluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5,6-difluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-methoxy-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-chloro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-chloro-2H-indazol-2-yl)methyl)benzamide;
    • 4-((3-amino-5-(trifluoromethyl)-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((3-amino-5-(trifluoromethyl)-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((3-oxo-2,3-dihydro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-oxo-1H-indazol-2(3H)-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-methoxy-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-methoxy-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-ethyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-ethyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-hydroxy-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-hydroxy-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-phenyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-phenyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-methoxy-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3,5-dimethyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3,5-dimethyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((7-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((7-methoxy-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-methoxy-1H-indazol-1-yl)methyl)benzamide;
    • 4-((6-acetamido-3-methyl-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((6-acetamido-3-methyl-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((3-amino-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((6-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((6-acetamido-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methyl-7-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methyl-6-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methyl-6-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methyl-5-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methyl-5-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • 4-((3-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((5-hydroxy-6-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5,6-dimethoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5,6-dimethoxy-1H-indazol-1-yl)methyl)benzamide;
    • 4-((1H-benzo[d][1,2,3]triazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((2H-benzo[d][1,2,3]triazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(piperazin-1-yl)ethyl)-1H-pyrazole-4-carboxamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(4-aminopyrimidin-5-yl)benzamide;
    • 4-((5-acetamido-3-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-acetamido-3-methyl-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-acetamido-3-methyl-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-(1-(1H-indazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • 4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • 5-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(2-amino-5-(thiophen-2-yl)phenyl)benzamide;
    • N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4,6-difluoro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4,6-difluoro-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((6-methoxy-3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((6-methoxy-3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((6-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxylic acid;
    • N-(2-aminophenyl)-4-((3-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3,5-dimethyl-1H-pyrazol-1-yl)methyl)benzamide;
    • Ethyl 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxylate;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-2-ylmethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-phenyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-benzyl-1H-pyrazole-4-carboxamide;
    • N-(2-aminophenyl)-4-((4,5,6,7-tetrahydro-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4,5,6,7-tetrahydro-1H-indazol-1-yl)methyl)benzamide;
    • 4-((1H-pyrazol-3-ylamino)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((5-amino-1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((3-amino-1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-methyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-ethyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N,N-dimethyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-isopropyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-cyclopropyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(cyclopropylmethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(dimethylamino)ethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-hydroxyethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-(dimethylamino)propyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-3-ylmethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-4-ylmethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-morpholinoethyl)-1H-pyrazole-4-carboxamide;
    • N-(2-aminophenyl)-4-((5-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • (4-(2-aminophenylcarbamoyl)benzyl)-N-propyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-isobutyl-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-methoxyethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-methoxypropyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-hydroxypropyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-morpholinopropyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(piperidin-1-yl)ethyl)-1H-pyrazole-4-carboxamide;
    • 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-(4-methylpiperazin-1-yl)propyl)-1H-pyrazole-4-carboxamide;
    • 4-((1H-Pyrrolo[2,3-b]pyridin-1-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((1H-Indol-3-yl)methyl)-N-(2-aminophenyl)benzamide;
    • 4-((1H-indazol-1-yl)methyl)-N-(2-amino-5-fluorophenyl)benzamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(2-amino-5-fluorophenyl)benzamide;
    • 4-((1H-indazol-1-yl)methyl)-N-(2-amino-5-fluorophenyl)-3-methylbenzamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(2-amino-5-fluorophenyl)-3-methylbenzamide;
    • N-(2-aminophenyl)-3-methyl-4-((3-methyl-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-3-methyl-4-((3-methyl-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-cyano-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-cyano-2H-indazol-2-yl)methyl)benzamide;
    • 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazole-3-carboxylic acid;
    • 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazole-3-carboxylic acid;
    • N-(2-aminophenyl)-4-((5-cyano-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-cyano-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(pyridin-2-yl)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(pyridin-2-yl)-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(dimethylamino)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(dimethylamino)-2H-indazol-2-yl)methyl)benzamide;
    • 4-((1H-indazol-1-yl)methyl)-N-(2-amino-5-(furan-2-yl)phenyl)benzamide;
    • 4-((2H-indazol-2-yl)methyl)-N-(2-amino-5-(furan-2-yl)phenyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methoxy-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-methoxy-2H-indazol-2-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(methoxymethylamino)-1H-indazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(methoxymethylamino)-2H-indazol-2-yl)methyl)benzamide;
    • 4-((1H-pyrazol-1-yl)methyl)-N-(2-amino-5-fluorophenyl)benzamide;
    • N-(2-amino-5-fluorophenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • 4-((1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)-3-methylbenzamide;
    • N-(2-amino-5-fluorophenyl)-3-methyl-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-(furan-2-yl)-1H-pyrazol-1-yl)methyl)benzamide;
    • 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-pyrazole-4-carboxylic acid;
    • N-(2-aminophenyl)-4-((3-ethyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-ethyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((4-phenyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-3-methyl-4-((5-methyl-4-phenyl-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-cyano-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-(pyridin-2-yl)-1H-pyrazol-1-yl)methyl)benzamide;
    • 4-((1H-pyrazol-1-yl)methyl)-N-(2-amino-5-(furan-2-yl)phenyl)benzamide;
    • N-(2-amino-5-(furan-2-yl)phenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide;
    • (S)-4-(1-(1H-pyrazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • (R)-4-(1-(1H-pyrazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(pyridin-2-yl)-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-methoxy-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((3-(methoxymethylamino)-1H-pyrazol-1-yl)methyl)benzamide;
    • N-(2-aminophenyl)-4-((5-(methoxymethylamino)-1H-pyrazol-1-yl)methyl)benzamide; and
    • N-(2-aminophenyl)-4-((3-methoxy-1H-pyrazol-1-yl)methyl)benzamide.
  • It is noted that the compounds of the present invention may be in the form of a pharmaceutically acceptable salt, biohydrolyzable ester, biohydrolyzable amide, biohydrolyzable carbamate, solvate, hydrate or prodrug thereof. For example, the compound optionally comprises a substituent that is convertible in vivo to a different substituent such as a hydrogen.
  • It is further noted that the compound may be present in a mixture of stereoisomers (including tautomers), or the compound may comprise a single stereoisomer.
  • The present invention also provides a pharmaceutical composition comprising as an active ingredient a compound according to any one of the above embodiments and variations. In one particular variation, the composition is a solid formulation adapted for oral administration. In another particular variation, the composition is a liquid formulation adapted for oral administration. In yet another particular variation, the composition is a tablet. In still another particular variation, the composition is a liquid formulation adapted for parenteral administration.
  • In another of its aspects, there is provided a pharmaceutical composition comprising a compound according to any one of the above embodiments and variations, wherein the composition is adapted for administration by a route selected from the group consisting of orally, parenterally, intraperitoneally, 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, and intrathecally.
  • In yet another of its aspects, there is provided a kit comprising a compound of any one of the above embodiments and variations; and instructions which comprise one or more forms of information selected from the group consisting of indicating a disease state for which the composition is to be administered, storage information for the composition, dosing information and instructions regarding how to administer the composition. In one particular variation, the kit comprises the compound in a multiple dose form.
  • In still another of its aspects, there is provided an article of manufacture comprising a compound of any one of the above embodiments and variations; and packaging materials. In one variation, the packaging material comprises a container for housing the compound. In one particular variation, the container comprises a label indicating one or more members of the group consisting of a disease state for which the compound is to be administered, storage information, dosing information and/or instructions regarding how to administer the compound. In another variation, the article of manufacture comprises the compound in a multiple dose form.
  • In a further of its aspects, there is provided a therapeutic method comprising administering a compound of any one of the above embodiments and variations to a subject.
  • In another of its aspects, there is provided a method of inhibiting HDAC comprising contacting HDAC with a compound of any one of the above embodiments and variations.
  • In yet another of its aspects, there is provided a method of inhibiting HDAC comprising causing a compound of any one of the above embodiments and variations to be present in a subject in order to inhibit HDAC in vivo.
  • In a further of its aspects, there is provided a method of inhibiting HDAC comprising administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo, the second compound being a compound according to any one of the above embodiments and variations.
  • In another of its aspects, there is provided a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state, the method comprising causing a compound of any one of the above embodiments and variations to be present in a subject in a therapeutically effective amount for the disease state.
  • In yet another of its aspects, there is provided a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state, the method comprising administering a compound of any one of the above embodiments and variations to a subject, wherein the compound is present in the subject in a therapeutically effective amount for the disease state.
  • In a further of its aspects, there is provided a method of treating a disease state for which HDAC possesses activity that contributes to the pathology and/or symptomatology of the disease state, the method comprising administering a first compound to a subject that is converted in vivo to a second compound wherein the second compound inhibits HDAC in vivo, the second compound being a compound according to any one of the above embodiments and variations.
  • In still another embodiment, the present invention relates to a method for treating cancer comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof. In one variation, the cancer is selected from the group consisting of squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, non small-cell lung cancer, bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, small-cell lung cancer, glioma, colorectal cancer, genitourinary cancer and gastrointestinal cancer.
  • In another embodiment, the present invention relates to a method for treating inflammation, inflammatory bowel disease, psoriasis, or transplant rejection, comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • In a further embodiment, the present invention relates to a method for treating arthritis comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • In yet another embodiment, the present invention relates to a method for treating degenerative diseases of the eye comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • In still another embodiment, the present invention relates to a method for treating multiple sclerosis, amyotrophic lateral sclerosis, thyroid neoplasm or Alzheimer's disease comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • In a further embodiment, the present invention relates to a method for treating hyperproliferative skin diseases or inflammatory cutaneous disorders comprising administering a compound according to any one of the above embodiments and variations to a mammalian species in need thereof.
  • In each of the above embodiments and variations, the histone deacetylase is optionally a Class I histone deacetylase. In particular variations of each of the above embodiments and variations, the histone deacetylase is HDAC2 and/or HDAC8.
  • In still other of its aspects, the present invention relates to methods of making the HDAC inhibitors of the present invention, as well as intermediates useful for the preparation of such HDAC inhibitors. In one embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00022
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00023
      • under conditions that form a reaction product comprising a formula selected from the group consisting of
  • Figure US20080114037A1-20080515-C00024
      • wherein
      • X is a leaving group;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached; and
      • R5, R6 and R7 are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R6 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In another embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00025
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00026
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00027
      • wherein
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • R5, R6 and R7 are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R6 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00028
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00029
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00030
      • wherein
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • R5, R6 and R7 are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R6 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In yet another embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00031
      • with a compound comprising the formula

  • R—C(O)—X
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00032
      • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • X is a leaving group;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In a further embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00033
      • with a compound comprising the formula

  • R—C(O)—X
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00034
      • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • X is a leaving group;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In still a further embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00035
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00036
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00037
      • wherein
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In yet a further embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00038
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00039
      • under conditions that form a reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00040
      • wherein
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In another embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00041
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00042
      • under conditions that form a first reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00043
      •  and
      • reacting the first reaction product with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00044
      • under conditions that form a second reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00045
      • wherein
      • n is selected from the group consisting of 0, 1, 2, 3 and 4;
      • X is a leaving group;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
      • R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In another embodiment, the processes comprise:
      • reacting a compound comprising the formula
  • Figure US20080114037A1-20080515-C00046
      • with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00047
      • under conditions that form a first reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00048
      •  and
      • reacting the first reaction product with a compound comprising the formula
  • Figure US20080114037A1-20080515-C00049
      • under conditions that form a second reaction product comprising a formula
  • Figure US20080114037A1-20080515-C00050
      • wherein
      • X is a leaving group.
  • In one variation of the above embodiments, X is halo. In one particular variation, X is bromo. In another particular variation, X is chloro.
  • In yet other of its aspects, the present invention relates to compounds useful in the preparation of the HDAC inhibitors of the present invention. In one embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00051
  • wherein
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached; and
      • R5, R6 and R7 are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R6 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In another embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00052
  • wherein
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached; and
      • R5, R6 and R7 are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R6 and R7 may be taken together to form a substituted or unsubstituted ring.
  • In still another embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00053
  • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In one particular variation of the above embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00054
  • In yet another embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00055
  • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In a further embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00056
  • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
  • In still a further embodiment, the compounds comprise:
  • Figure US20080114037A1-20080515-C00057
  • wherein
      • p is selected from the group consisting of 0, 1, 2, 3 and 4;
      • A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
      • R is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
      • R7 is selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted; and
      • each R12 is independently selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R12 and R7 or R11 may be taken together to form a substituted or unsubstituted ring.
    Salts, Hydrates, and Prodrugs of HDAC Inhibitors
  • It should be recognized that the 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.
  • 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; other 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, bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, cyclopentanepropionate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, 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 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.
  • When the compounds of the present invention possess a free acid 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 (benzathine), dicyclohexylamine, diethanolamine, 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-(hydroxymethyl)methylamine (tromethamine). 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.
  • Compounds of the present invention that comprise basic nitrogen-containing groups may be quarternerized with such agents as (C1-4) alkyl halides, e.g., methyl, ethyl, iso-propyl and tert-butyl chlorides, bromides and iodides; di(C1-4) alkyl sulfates, e.g., dimethyl, diethyl and diamyl sulfates; (C10-18) alkyl halides, e.g., decyl, dodecyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aryl(C1-4) 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.
  • 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, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0° C. Alternatively, 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 that 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., 1,1-acyloxyalkylcarbonochloridate, para-nitrophenyl carbonate, or the like) or an acylating agent. Further examples of methods of making prodrugs are described in Saulnier et al. (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, 3rd 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 dioxin, 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. While the route of administration of the pharmaceutical composition is important, and various anatomical, physiological and pathological factors can critically affect bioavailability, the solubility of the compound is usually dependent upon the character of the 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 absorption of the compound.
  • Preparation of HDAC Inhibitors
  • 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 compounds of the present invention may also be synthesized by other synthetic routes that others may devise.
  • 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 (i.e., enantiomers and diastereomers). Unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all of the different possible stereoisomers.
  • 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).
  • Compositions Comprising HDAC Inhibitors
  • A wide variety of compositions and administration methods may be used in conjunction with the compounds of the present invention. Such compositions may include, in addition to the compounds of the present invention, conventional pharmaceutical excipients, and other conventional, pharmaceutically inactive agents. Additionally, the compositions may include active agents in addition to the compounds of the present invention. These additional active agents may include additional compounds according to the invention, and/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 compounds of the present invention may be administered or co-administered orally, parenterally, intraperitoneally, 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 compounds and/or compositions according to the invention may also be administered or co-administered in slow release dosage forms.
  • The HDAC inhibitors and compositions comprising them may be administered or co-administered in any conventional dosage form. Co-administration in the context of this invention is intended to mean the administration of more than one therapeutic agent, one of which includes a HDAC inhibitor, in the course of a coordinated treatment to achieve an improved clinical outcome. Such co-administration 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, disposable syringes or single or multiple dose vials made of glass, plastic or other suitable material.
  • When compounds 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.
  • Upon mixing or adding compounds 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.
  • 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 therapeutically active compounds and derivatives thereof 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.
  • In addition to one or more compounds 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, polyinylpyrrolidine, 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's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975. The composition or formulation to be administered will, in any event, contain a sufficient quantity of a 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 compounds 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%.
  • Salts, preferably sodium salts, of the 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.
  • Formulations for Oral Administration
  • 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.
  • In certain embodiments, compounds 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.
  • 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.
  • Examples of lubricants that may be used include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.
  • Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • Examples of glidants that may be used include, but are not limited to, colloidal silicon dioxide.
  • 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.
  • 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.
  • 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.
  • 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.
  • Examples of wetting agents that may be used include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • 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.
  • Examples of film coatings that may be used include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
  • If oral administration is desired, the salt of the 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.
  • 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.
  • 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 compounds 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.
  • Examples of pharmaceutically acceptable carriers that may be included in tablets comprising compounds 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.
  • Examples of liquid oral dosage forms that 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.
  • 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.
  • 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.
  • 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 acids and a source of carbon dioxide.
  • Coloring and flavoring agents may optionally be used in all of the above dosage forms.
  • Particular examples of preservatives that may be used include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • Particular examples of non-aqueous liquids that may be used in emulsions include mineral oil and cottonseed oil.
  • Particular examples of emulsifying agents that may be used include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • 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.
  • Particular examples of wetting agents that may be used include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • Particular examples of 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.
  • 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, e.g., 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.
  • 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.
  • Injectables, Solutions, and Emulsions
  • The present invention is also directed to compositions designed to administer the compounds of the present invention by parenteral administration, generally characterized by subcutaneous, intramuscular or intravenous injection. 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.
  • Examples of excipients that may be used in conjunction with injectables 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.
  • 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.
  • Examples 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.
  • Examples of aqueous vehicles that may optionally be used include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection.
  • 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.
  • 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 be used 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. 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.
  • 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 an 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 an 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 known 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 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 the 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.
  • 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.
  • Lyophilized Powders
  • The compounds 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 compound 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 the 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 inhibitor.
  • Topical Administration
  • The compounds 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 of 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 have diameters of less than 50 microns, preferably less than 10 microns.
  • The 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.
  • Formulations for Other Routes of Administrations
  • Depending upon the disease state being treated, other routes of administration, such as topical application, transdermal patches, and 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, (polyoxyethylene 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.
  • Examples of Formulations
  • 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.
  • ORAL FORMULATION
    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
  • TABLET FORMULATION
    Compound of the Present Invention  1%
    Microcrystalline Cellulose 73%
    Stearic Acid 25%
    Colloidal Silica   1%.
  • Kits Comprising HDAC Inhibitors
  • The invention is also directed to kits and other articles of manufacture for treating diseases associated with HDACs. It is noted that diseases are intended to cover all conditions for which the HDACs possess activity that contributes to the pathology and/or symptomatology of the condition.
  • In one embodiment, a kit is provided that comprises a composition comprising at least one 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.
  • In another embodiment, an article of manufacture is provided that comprises a composition comprising at least one 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.
  • 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 kit 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.
  • 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.
  • 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.
  • Combination Therapy
  • A wide variety of 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 growth resulting in undesirable benign conditions or tumor growth.
  • 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 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.
  • Examples of therapeutic agents that may be used in combination with HDAC inhibitors include, but are not limited to, anticancer 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. 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.
  • 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, epirubicin, idarubicin and anthracenedione), 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 chemotherapeutic 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. 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.
  • 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 therapy including a HDAC inhibitor and a hormonal agent may have therapeutic synergistic effects on cancer and reduce sides affects associated with these 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. 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 II, 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. 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.
  • 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 non-self. 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 (IL-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.
  • 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.
  • 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.
  • 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.
  • Another example of monoclonal antibodies against tumor antigens is RITUXAN® (Rituximab) that is raised against CD2O 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 Wilms 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.
  • 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, gp 100, 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. 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.
  • EXAMPLES Preparation of HDAC Inhibitors
  • 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.
  • 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 (i.e., enantiomers and diastereomers). Unless a particular stereochemistry is specified, recitation of a compound is intended to encompass all of the different possible stereoisomers.
  • 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.) and 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).
  • Compounds according to the present invention can also 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. Alternatively, a pharmaceutically acceptable base addition salt of a compound can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Inorganic and organic acids and bases suitable for the preparation of the pharmaceutically acceptable salts of compounds are set forth in the definitions section of this application. Alternatively, the salt forms of the compounds can be prepared using salts of the starting materials or intermediates.
  • The free acid or free base forms of the compounds can be prepared from the corresponding base addition salt or acid addition salt form. For example, a compound in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A compound in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc).
  • The 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, meta-chloroperoxybenzoic acid, or the like) in a suitable inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at approximately 0 C. Alternatively, the N-oxides of the compounds can be prepared from the N-oxide of an appropriate starting material.
  • Compounds in an unoxidized form can be prepared from N-oxides of compounds by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in an suitable inert organic solvent (e.g., acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80° C.
  • Prodrug derivatives of the compounds can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al. (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound with a suitable carbamylating agent (e.g., 1,1-acyloxyalkylcarbonochloridate, para-nitrophenyl carbonate, or the like).
  • Protected derivatives of the compounds can be made by methods known to those of ordinary skill in the art. A detailed description of the 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.
  • Compounds according to the present invention may 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 dioxin, tetrahydrofuran or methanol.
  • Compounds according to the present invention can also be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomer. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of compounds, dissociable complexes are preferred (e.g., crystalline diastereoisomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography or, preferably, by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of 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).
  • 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 three-letter abbreviations are generally used to designate amino acid residues, which are assumed to be in the L-configuration unless otherwise 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:
  • μL (microliters) Ac (acetyl)
    atm (atmosphere) ATP (Adenosine Triphophatase)
    BOC (tert-butyloxycarbonyl) BOP (bis(2-oxo-3-oxazolidinyl)phosphinic
    chloride)
    BSA (Bovine Serum Albumin) CBZ (benzyloxycarbonyl)
    CDI (1,1-carbonyldiimidazole) DCC (dicyclohexylcarbodiimide)
    DCE (dichloroethane) DCM (dichloromethane)
    DMAP (4-dimethylaminopyridine) DME (1,2-dimethoxyethane)
    DMF (N,N-dimethylformamide) DMPU (N,N′-dimethylpropyleneurea)
    DMSO (dimethylsulfoxide) EDCI (ethylcarbodiimide hydrochloride)
    EDTA (Ethylenediaminetetraacetic acid) Et (ethyl)
    Et2O (diethyl ether) EtOAc (ethyl acetate)
    FMOC (9-fluorenylmethoxycarbonyl) g (grams)
    h (hours) HOAc or AcOH (acetic acid)
    HOBT (1-hydroxybenzotriazole) HOSu (N-hydroxysuccinimide)
    HPLC (high pressure liquid chromatography) Hz (Hertz)
    i.v. (intravenous) IBCF (isobutyl chloroformate)
    i-PrOH (isopropanol) L (liters)
    M (molar) mCPBA (meta-chloroperbenzoic acid)
    Me (methyl) MeOH (methanol)
    mg (milligrams) MHz (megahertz)
    min (minutes) mL (milliliters)
    mM (millimolar) mmol (millimoles)
    mol (moles) MOPS (Morpholinepropanesulfonic acid)
    mp (melting point) NaOAc (sodium acetate)
    OMe (methoxy) psi (pounds per square inch)
    RP (reverse phase) RT (ambient temperature)
    SPA (Scintillation Proximity Assay) TBAF (tetra-n-butylammonium fluoride)
    TBS (t-butyldimethylsilyl) tBu (tert-butyl)
    TEA (triethylamine) TFA (trifluoroacetic acid)
    TFAA (trifluoroacetic anhydride) THF (tetrahydrofuran)
    TIPS (triisopropylsilyl) TLC (thin layer chromatography)
    TMS (trimethylsilyl) TMSE (2-(trimethylsilyl)ethyl)
    Tr (retention time)
  • All references to ether or Et2O are to diethyl ether; and brine refers to a saturated aqueous solution of NaCl. Unless otherwise indicated, all temperatures are expressed in ° C. (degrees Centigrade). All reactions are conducted under an inert atmosphere at RT unless otherwise noted.
  • 1H 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).
  • 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).
  • The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as the Aldrich Chemical Company (Milwaukee, Wis.), Bachem (Torrance, Calif.), Sigma (St. Louis, Mo.), or may be prepared by methods well known to a person of ordinary skill in the art, following procedures described in such standard references as Fieser and Fieser's Reagents for Organic Synthesis, vols. 1-17, John Wiley and Sons, New York, N.Y., 1991; Rodd's Chemistry of Carbon Compounds, vols. 1-5 and supps., Elsevier Science Publishers, 1989; Organic Reactions, vols. 1-40, John Wiley and Sons, New York, N.Y., 1991; March J.: Advanced Organic Chemistry, 4th ed., John Wiley and Sons, New York, N.Y.; and Larock: Comprehensive Organic Transformations, VCH Publishers, New York, 1989.
  • The entire disclosure of the documents cited throughout this application are incorporated herein by reference.
  • Synthetic Schemes for Compounds of the Present Invention
  • Compounds according to the present invention may be synthesized according to the reaction schemes shown below. Other reaction schemes could be readily devised by those skilled in the art. It should also be appreciated that a variety of different solvents, temperatures and other reaction conditions can be varied to optimize the yields of the reactions.
  • In the reactions described hereinafter it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see T. W. Greene and P. G. M. Wuts in “Protective Groups in Organic Chemistry” John Wiley and Sons, 1991.
  • General synthetic routes for producing compounds of the present invention are shown in the schemes below.
  • Figure US20080114037A1-20080515-C00058
  • A solution of B (0.5 mmol) in DMF (2.5 mL) is treated with an appropriate heterocycle A (0.5 mmol) and solid K2CO3 (0.6 mmol). After stirring at 23° C. for 18 h, the reaction is poured into water (10 mL) and the resulting solid isolated by filtration. The filter cake can be rinsed with water and allowed to dry in vacuo to yield a mixture of regioisomeric alkylation products C, D and E, which can be separated by flash chromatography. If no solid is formed, the DMF/water mixture can be extracted with ethylacetate. The combined extracts can be dried (MgSO4), concentrated in vacuo and purified by flash chromatography.
  • A solution of the alkylation products C, D and/or E (0.25 mmol) in dioxane (1 mL) is treated with aqueous LiOH (1 M, 1.0 mL). After stirring at 23° C. for 2 h, the reaction is neutralized with aqueous HCl (1 M, 2.0 mL) and the resulting solid isolated by filtration. The filter cake can be rinsed with water and allowed to dry in vacuo to yield the corresponding carboxylic acid.
  • A solution of the carboxylic acid formed above (0.1 mmol) in DMF (1 mL) is sequentially treated with EDC (0.12 mmol), HOBt (0.12 mmol), a substituted or unsubstituted 1,2-phenylenediamine (0.12 mmol) and NMM (0.3 mmol). After stirring at 23° C. for 4 h, the reaction is poured into water (10 mL) and the resulting solid isolated by filtration. The filter cake can be rinsed with water and allowed to dry in vacuo to yield the corresponding amide F, G and/or H.
  • Figure US20080114037A1-20080515-C00059
  • In relation to Scheme 2, the alkylation of I with B, hydrolysis of J and/or K, and coupling with the 1,2-phenylenediamine to provide L and/or M proceed as described in Scheme 1.
  • Figure US20080114037A1-20080515-C00060
  • In relation to Scheme 3, nitrobenzenes J′ and K′ may be prepared as described in connection with Schemes 1 and 2.
  • A solution of the nitrobenzene (J′ and/or K′) (1.0 mmol) in MeOH (10 mL) with Pd/C (10 wt %, 50 mg) is stirred vigorously under hydrogen for 4 h. The reaction is filtered and concentrated in vacuo to give the corresponding aniline that can be carried forward.
  • A solution of the aniline (1.0 mmol) in dichloromethane (10 mL) is treated sequentially with the appropriate acid chloride or chloroformate (RC(O)Cl), wherein R is, for example, an optionally substituted alkoxy, alkyl, heterocycloalkyl or aryl) (1.1 mmol) and triethylamine (2.2 mmol). After stirring at 23° C. for 4 h, the reaction is concentrated in vacuo to yield the corresponding acylation product (N and/or O). If necessary, purification can be carried out by flash chromatography or recrystallization.
  • Compounds N and/or O can be coupled with a substituted or unsubstituted 1,2-phenylenediamine to yield P and/or Q, as described in connection with Schemes 1 and 2.
  • Chiral components can be separated and purified using any of a variety of techniques known to those skilled in the art. For example, chiral components can be purified using supercritical fluid chromatography (SFC). In one particular variation, chiral analytical SFC/MS analyses are conducted using a Berger analytical SFC system (AutoChem, Newark, Del.) which consists of a Berger SFC dual pump fluid control module with a Berger FCM 1100/1200 supercritical fluid pump and FCM 1200 modifier fluid pump, a Berger TCM 2000 oven, and an Alcott 718 autosampler. The integrated system can be controlled by BI-SFC Chemstation software version 3.4. Detection can be accomplished with a Waters ZQ 2000 detector operated in positive mode with an ESI interface and a scan range from 200-800 Da with 0.5 second per scan. Chromatographic separations can be performed on a ChiralPak AD-H, ChiralPak AS-H, ChiralCel OD-H, or ChiralCel OJ-H column (5 μ, 4.6×250 mm; Chiral Technologies, Inc. West Chester, Pa.) with 10 to 40% methanol as the modifier and with or without ammonium acetate (10 mM). Any of a variety of flow rates can be utilized including, for example, 1.5 or 3.5 mL/min with an inlet pressure set at 100 bar. Additionally, a variety of sample injection conditions can be used including, for example, sample injections of either 5 or 10 μL in methanol at 0.1 mg/mL in concentration.
  • In another variation, preparative chiral separations are performed using a Berger MultiGram II SFC purification system. For example, samples can be loaded onto a ChiralPak AD column (21×250 mm, 10μ). In particular variations, the flow rate for separation can be 70 mL/min, the injection volume up to 2 mL, and the inlet pressure set at 130 bar. Stacked injections can be applied to increase the efficiency.
  • In each of the above reaction procedures or schemes, the various substituents may be selected from among the various substituents otherwise taught herein.
  • Descriptions of the syntheses of particular compounds according to the present invention based on the above reaction scheme are set forth herein.
  • Examples of HDAC Inhibitors
  • The present invention is further exemplified, but not limited by, the following examples that describe the synthesis of particular compounds according to the invention.
  • Examples 1 and 2 4-((1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide and 4-((2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00061
  • The title compounds were prepared using the procedure described in Scheme 2. In particular, a solution of methyl 4-bromomethylbenzoate (0.5 mmol) in DMF (2.5 mL) was treated with the indazole (0.5 mmol) and solid K2CO3 (0.6 mmol). After stirring at 23° C. for 18 h, the reaction was poured into water (10 mL) and the resulting solid was isolated by filtration. The filter cake was rinsed with water and allowed to dry in vacuo to yield a mixture of regioisomeric alkylation products, which were separated by flash chromatography.
  • A solution of the methyl ester (0.25 mmol) in dioxane (1 mL) was treated with aqueous LiOH (1 M, 1.0 mL). After stirring at 23° C. for 2 h, the reaction was neutralized with aqueous HCl (1 M, 2.0 mL) and the resulting solid was isolated by filtration. The filter cake was rinsed with water and allowed to dry in vacuo to yield the corresponding benzoic acid.
  • A solution of the appropriate benzoic acid (0.1 mmol) in DMF (1 mL) was sequentially treated with EDC (0.12 mmol), HOBt (0.12 mmol), 1,2-phenylenediamine (0.12 mmol) and NMM (0.3 mmol). After stirring at 23° C. for 4 h, the reaction was poured into water (10 mL) and the resulting solid was isolated by filtration. The filter cake was rinsed with water and allowed to dry in vacuo to yield the corresponding amide.
  • Example 1: 1H NMR (400 MHz, DMSO-d6) δ ppm 4.85 (s, 2H) 5.74 (s, 2H) 6.53-6.58 (m, 1H) 6.73 (dd, J=7.83, 1.26 Hz, 1H) 6.91-6.96 (m, 1H) 7.09-7.16 (m, 2H) 7.28-7.39 (m, 3H) 7.70 (d, J=8.34 Hz, 1H) 7.77 (d, J=8.34 Hz, 1H) 7.87 (d, J=8.08 Hz, 2H) 8.13 (s, 1H) 9.56 (s, 1H). ESI-MS: m/z 343.4 (M+H)+.
  • Example 2: 1H NMR (400 MHz, DMSO-D6) δ ppm 4.94 (s, 2H) 5.72 (s, 2H) 6.57 (t, J=7.58 Hz, 1H) 6.74-6.77 (m, 1H) 6.95 (td, J=7.58, 1.52 Hz, 1H) 7.00-7.05 (m, 1H) 7.13 (d, J=7.58 Hz, 1H) 7.22 (dd, J=8.21, 7.20 Hz, 1H) 7.41 (d, J=8.08 Hz, 2H) 7.58 (d, J=8.59 Hz, 1H) 7.71 (d, J=8.59 Hz, 1H) 7.93 (d, J=8.08 Hz, 2H) 8.51 (s, 1H) 9.62 (s, 1H). ESI-MS: m/z 343.4 (M+H)+.
  • Examples 3 and 4 4-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide and 4-((5-acetamido-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00062
  • The title compounds were prepared using a procedure analogous to that described in Examples 1 and 2 except that the nitrobenzene was reduced and the aniline acylated as further described in Scheme 2. Specifically, the alkylation with methyl 4-bromomethylbenzoate, hydrolysis of methyl benzoate, and coupling with 1,2-phenylenediamine proceed as described in connection with Examples 1 and 2. A solution of the nitrobenzene (1.0 mmol) in MeOH (10 mL) with Pd/C (10 wt %, 50 mg) was then stirred vigorously under hydrogen for 4 h. The reaction was filtered and concentrated in vacuo to give the corresponding aniline that was carried forward. A solution of the aniline (1.0 mmol) in dichloromethane (10 mL) was treated sequentially with the appropriate acid chloride or chloroformate (1.1 mmol) and triethylamine (2.2 mmol). After stirring at 23° C. for 4 h, the reaction was concentrated in vacuo to yield the corresponding acylation product. If necessary, purification can be carried out by flash chromatography or recrystallization.
  • Example 3: 1H NMR (400 MHz, DMSO-d6) δ ppm 2.02 (s, 3H) 4.85 (s, 2H) 5.69 (s, 2H) 6.49-6.60 (m, 1H) 6.73 (d, J=8.08 Hz, 1H) 6.87-6.99 (m, 2H) 7.11 (d, J=8.08 Hz, 1H) 7.29 (d, J=8.08 Hz, 2H) 7.39 (dd, J=9.09, 1.77 Hz, 1H) 7.61 (d, J=8.84 Hz, 1H) 7.87 (d, J=8.08 Hz, 2H) 8.06 (s, 1H) 8.12 (s, 1H) 9.56 (s, 1H). ESI-MS: m/z 400.5 (M+H)+.
  • Example 4: 1H NMR (400 MHz, DMSO-D6) δ ppm 2.03 (s, 3H) 4.87 (s, 2H) 5.67 (s, 2H) 6.45-6.65 (m, 1H) 6.74 (d, J=8.08 Hz, 1H) 6.86-7.02 (m, 1H) 7.13 (d, J=7.33 Hz, 1H) 7.20 (dd, J=9.35, 1.77 Hz, 1H) 7.39 (d, J=8.08 Hz, 2H) 7.52 (d, J=9.09 Hz, 1H) 7.92 (d, J=8.08 Hz, 2H) 8.12 (s, 1H) 8.41 (s, 1H) 9.61 (s, 1H) 9.85 (s, 1H). ESI-MS: m/z 400.5 (M+H)+.
  • Example 5 N-(2-aminophenyl)-4-((5-nitro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00063
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 5.86 (s, 2H) 6.96 (s, 1H) 7.05 (d, J=7.58 Hz, 1H) 7.13 (d, J=7.33 Hz, 1H) 7.25 (d, J=7.83 Hz, 1H) 7.37 (d, J=8.08 Hz, 2H) 7.92 (d, J=8.34 Hz, 2H) 7.96 (d, J=9.09 Hz, 1H) 8.24 (dd, J=9.35, 2.27 Hz, 1H) 8.47 (s, 1H) 8.85 (d, J=2.02 Hz, 1H) 9.99 (s, 1H). ESI-MS: m/z 388.4 (M+H)+.
  • Example 6 4-((5-benzamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00064
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.86 (s, 2H) 5.73 (s, 2H) 6.49-6.62 (m, 1H) 6.73 (d, J=7.83 Hz, 1H) 6.85-6.97 (m, 1H) 7.11 (d, J=7.83 Hz, 1H) 7.30 (d, J=8.08 Hz, 2H) 7.45-7.62 (m, 3H) 7.61-7.74 (m, 2H) 7.88 (d, J=8.34 Hz, 2H) 7.96 (d, J=6.82 Hz, 2H) 8.12 (s, 1H) 8.26 (s, 1H) 9.57 (s, 1H) 10.28 (s, 1H). ESI-MS: m/z 462.5 (M+H)+.
  • Example 7 4-((5-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00065
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.85 (bs, 4H) 5.59 (s, 2H) 6.55 (s, 1 H) 6.71-6.80 (m, 3H) 6.93 (d, J=6.32 Hz, 1H) 7.11 (d, J=6.82 Hz, 1H) 7.26 (d, J=7.58 Hz, 2H) 7.30-7.40 (m, 1H) 7.73-7.83 (m, 1H) 7.86 (d, J=7.58 Hz, 2H) 9.56 (s, 1H). ESI-MS: m/z 358.4 (M+H)+.
  • Example 8 N-(2-aminophenyl)-4-((5-nitro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00066
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.87 (br. s., 2H) 5.75-5.88 (m, 2H) 6.51-6.61 (m, 1H) 6.74 (d, J=7.33 Hz, 1H) 6.90-6.98 (m, 1H) 7.13 (d, J=7.58 Hz, 1H) 7.46 (d, J=8.08 Hz, 2H) 7.77 (d, J=9.60 Hz, 1H) 7.94 (d, J=8.08 Hz, 2H) 7.97-8.04 (m, 1H) 8.91 (d, 1H) 8.94-9.01 (m, 1H) 9.62 (s, 1H). ESI-MS: m/z 388.4 (M+H)+.
  • Example 9 4-((5-amino-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00067
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 5.76 (s, 2H) 6.84-6.95 (m, 1H) 7.01 (d, J=7.83 Hz, 1H) 7.08-7.20 (m, 2H) 7.24 (s, 1H) 7.44 (d, J=8.08 Hz, 2H) 7.69-7.79 (m, 2H) 7.96 (d, J=7.83 Hz, 2H) 8.64 (s, 1H) 9.96 (s, 1H). ESI-MS: m/z 358.4 (M+H)+
  • Example 10 4-((5-benzamido-2H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00068
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.87 (s, 2H) 5.61-5.76 (s, 2H) 6.56 (t, J=7.33 Hz, 1H) 6.75 (d, J=8.08 Hz, 1H) 6.88-7.00 (m, 1H) 7.13 (d, J=8.08 Hz, 1H) 7.41 (d, J=7.07 Hz, 2H) 7.45-7.63 (m, 3H) 7.83-8.02 (m, 4H) 8.26 (s, 1H) 8.49 (s, 1H) 9.61 (s, 1H) 10.20 (s, 1H). ESI-MS: m/z 462.5 (M+H)+.
  • Example 11 2-Methoxyethyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00069
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 3.52-3.60 (m, 2H) 4.13-4.22 (m, 2H) 4.86 (bs, 2H) 5.66 (s, 2H) 6.56 (m, 1H) 6.75 (m, 1H) 6.93 (d, J=6.82 Hz, 1H) 7.13 (m, 1H) 7.21 (d, J=2.02 Hz, 1H) 7.39 (d, J=7.83 Hz, 2H) 7.52 (s, 1H) 7.82-7.89 (m, 2H) 7.91 (s, 1H) 8.40 (s, 1H) 9.60 (s, 1H) 9.64 (s, 1H). ESI-MS: m/z 460.5 (M+H)+.
  • Example 12 Methyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00070
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 3.32 (s, 3H) 4.87 (s, 2H) 5.58-5.73 (m, 2H) 6.48-6.62 (m, 1H) 6.74 (d, J=8.08 Hz, 1H) 6.88-7.00 (m, 1H) 7.07-7.17 (m, 1H) 7.18-7.24 (m, 1H) 7.39 (dd, J=8.08, 4.55 Hz, 2H) 7.46-7.57 (m, 1H) 7.88-8.00 (m, 2H) 8.39 (d, J=6.32 Hz, 1H) 8.56 (s, 1H) 9.54 (s, 1H) 9.61 (s, 1H). ESI-MS: m/z 416.5 (M+H)+.
  • Example 13 2-Methoxyethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00071
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 3.31 (s, 3H) 3.47-3.61 (m, 2H) 4.12-4.23 (m, 2H) 4.85 (s, 2H) 5.69 (s, 2H) 6.55 (t, J=7.58 Hz, 1H) 6.69-6.78 (m, 1H) 6.86-7.01 (m, 1H) 7.11 (d, J=7.83 Hz, 1H) 7.28 (d, J=8.34 Hz, 2H) 7.33-7.45 (m, 1H) 7.61 (d, J=9.09 Hz, 1H) 7.87 (m, 3H) 8.05 (s, 1H) 9.56 (s, 1H) 9.71 (s, 1H). ESI-MS: m/z 460.5 (M+H)+.
  • Example 14 Methyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00072
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 3.65 (s, 3H) 5.70 (s, 2H) 6.90 (s, 1H) 6.99 (s, 1H) 7.11 (s, 1H) 7.23 (d, J=7.83 Hz, 1H) 7.31 (d, J=8.08 Hz, 2H) 7.38 (s, 1H) 7.61 (d, J=9.09 Hz, 1H) 7.89 (d, J=7.83 Hz, 3H) 8.06 (s, 1H) 9.61 (s, 1H) 9.91 (s, 1H). ESI-MS: m/z 416.5 (M+H)+.
  • Example 15 Benzyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00073
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 4.85 (s, 2H) 5.14 (s, 2H) 5.69 (s, 2H) 6.55 (s, 1H) 6.73 (d, J=8.08 Hz, 1H) 6.93 (s, 1H) 7.04-7.18 (m, 1H) 7.22-7.49 (m, 7H) 7.61 (s, 1H) 7.79-8.00 (m, 4H) 8.05 (s, 1H) 9.56 (s, 1H) 9.74 (s, 1H). ESI-MS: m/z 492.5 (M+H)+.
  • Example 16 Benzyl 2-(4-((2-aminophenyl)carbamoyl)benzyl)-2H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00074
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 5.14 (s, 2H) 5.68 (s, 2H) 6.81-6.94 (m, 1H) 6.95-7.05 (m, 1H) 7.10 (s, 1H) 7.22 (dd, J=9.35, 1.77 Hz, 2H) 7.29-7.47 (m, 7H) 7.51 (d, J=9.35 Hz, 2H) 7.94 (d, J=8.08 Hz, 2H) 8.41 (s, 1H) 9.68 (s, 1H) 9.94 (s, 1H). ESI-MS: m/z 492.5 (M+H)+.
  • Example 17 N-(4-Amino-biphenyl-3-yl)-4-(5-nitro-indazol-2-ylmethyl)-benzamide
  • Figure US20080114037A1-20080515-C00075
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 5.84 (s, 2H) 6.97 (s, 1H) 7.25 (s, 1H) 7.39 (s, 3H) 7.44-7.62 (m, 5H) 7.79 (s, 1H) 7.99 (s, 3H) 8.98 (s, 2H) 9.89 (s, 1H). ESI-MS: m/z 464.5 (M+H)+.
  • Example 18 N-(2-(4-((2-Aminophenyl)carbamoyl)benzyl)-2H-indazol-5-yl)morpholine-4-carboxamide
  • Figure US20080114037A1-20080515-C00076
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 3.34-3.45 (m, 4H) 3.59 (m, 4H), 6.86-6.98 (m, 1H), 7.06-7.16 (m, 1H) 7.20-7.30 (m, 1H) 7.38 (d, 2H) 7.46 (m, 1H) 7.78 (s, 1H) 7.91 (d, 2H) 8.36 (s, 1H) 8.40-8.48 (m, 1H) 9.61 (s, 1H). ESI-MS: m/z 471.5 (M+H)+.
  • Example 19 5-((2H-Indazol-2-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide
  • Figure US20080114037A1-20080515-C00077
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.88 (s, 2H) 5.89 (s, 2H) 6.54-6.60 (m, 1H) 6.75 (d, J=8.08 Hz, 1H) 6.94-6.99 (m, 1H) 7.02-7.10 (m, 2H) 7.22-7.30 (m, 2H) 7.62 (d, J=8.84 Hz, 1H) 7.72 (d, J=8.59 Hz, 1H) 7.83 (d, J=3.03 Hz, 1H) 8.51 (s, 1H) 9.68 (s, 1H). ESI-MS: m/z 349.4 (M+H)+.
  • Example 20 N-(2-Aminophenyl)-5-((5-nitro-2H-indazol-2-yl)methyl)thiophene-2-carboxamide
  • Figure US20080114037A1-20080515-C00078
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.89 (s, 2H) 6.01 (s, 2H) 6.55-6.59 (m, 1H) 6.75 (d, J=8.08 Hz, 1H) 6.95 (d, J=7.83 Hz, 1H) 7.07 (d, J=7.83 Hz, 1H) 7.30 (s, 1H) 7.82 (d, J=9.35 Hz, 2H) 8.01-8.05 (m, 1H) 8.93-8.97 (m, 2H) 9.70 (s, 1H). ESI-MS: m/z 394.4 (M+H)+.
  • Example 21 N-(1-(4-((2-Aminophenyl)carbamoyl)benzyl)-1H-indazol-5-yl)morpholine-4-carboxamide
  • Figure US20080114037A1-20080515-C00079
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 3.37-3.47 (m, 4H) 3.50-3.66 (m, 4H) 5.62 (s, 2H) 6.56 (m, 1H) 6.74 (d, J=8.59 Hz, 1H) 6.94 (m, 1H) 7.11 (m, 1H) 7.29 (d, J=8.08 Hz, 2H) 7.39 (dd, J=9.09, 1.77 Hz, 1H) 7.57 (d, J=9.09 Hz, 1H) 7.74-7.89 (m, 3H) 7.96-8.05 (m, 1H) 8.52 (s, 1H) 9.57 (s, 1H). ESI-MS: m/z 471.5 (M+H)+.
  • Example 22 2-Morpholinoethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00080
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 2.41 (m, 2H) 2.56 (m, 2H) 3.55 (m, 4H) 4.18 (m, 4H) 4.85 (s, 2H) 5.68 (s, 2H) 6.47-6.63 (m, 1H) 6.73 (d, J=8.08 Hz, 1H) 6.85-6.99 (m, 1H) 7.10 (s, 1H) 7.28 (d, J=8.34 Hz, 2H) 7.39 (d, J=8.84 Hz, 1H) 7.61 (d, J=9.09 Hz, 1H) 7.87 (d, J=8.08 Hz, 3H) 8.04 (s, 1H) 9.55 (s, 1H) 9.60-9.71 (m, 1H). ESI-MS: m/z 515.6 (M+H)+.
  • Example 23 Pyridin-3-ylmethyl 1-(4-((2-aminophenyl)carbamoyl)benzyl)-1H-indazol-5-ylcarbamate
  • Figure US20080114037A1-20080515-C00081
  • The title compound was prepared using a procedure analogous to that described in Examples 3 and 4. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.85 (bs, 2H) 5.18 (s, 2H) 5.69 (s, 2H) 6.48-6.59 (m, 1H) 6.73 (dd, J=8.08, 1.26 Hz, 1H) 6.87-6.98 (m, 1H) 7.11 (d, J=7.33 Hz, 1H) 7.28 (d, J=8.34 Hz, 2H) 7.34-7.47 (m, 2H) 7.62 (d, J=9.09 Hz, 2H) 7.77-7.94 (m, 3H) 8.03-8.08 (m, 1H) 8.54 (dd, J=4.80, 1.52 Hz, 1H) 8.65 (d, J=1.77 Hz, 1H) 9.56 (s, 1H) 9.78 (s, 1H). ESI-MS: m/z 493.5 (M+H)+.
  • Example 24 5-((1H-Indazol-1-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide
  • Figure US20080114037A1-20080515-C00082
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.86 (s, 2H) 5.90 (s, 2H) 6.52-6.59 (m, 1H) 6.74 (d, J=7.33 Hz, 1H) 6.91-6.99 (m, 1H) 7.05 (d, J=7.33 Hz, 1H) 7.13-7.23 (m, 2H) 7.42 (s, 1H) 7.79 (m, 3H) 8.15 (s, 1H) 9.62 (s, 1H). ESI-MS: m/z 349.4 (M+H)+.
  • Example 25 4-((2H-Pyrazolo[3,4-b]pyridin-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00083
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) of the bis-trifluoroacetic acid salt: δ ppm 6.11 (s, 2H) 6.79 (d, J=8.08 Hz, 1H) 6.92 (d, J=8.08 Hz, 1H) 7.02-7.10 (m, 1H) 7.19 (d, J=7.58 Hz, 1H) 7.63 (d, J=8.08 Hz, 2H) 7.79 (dd, J=8.08, 5.81 Hz, 1H) 7.97 (d, J=8.34 Hz, 2H) 9.11-9.19 (m, 1H) 9.23 (d, J=7.83 Hz, 1H) 9.40 (d, J=4.80 Hz, 1H) 9.86 (s, 1H). ESI-MS: m/z 344.4 (M+H)+.
  • Example 26 N-(4-Amino-biphenyl-3-yl)-4-pyrazolo[3,4-b]pyridin-2-ylmethyl-benzamide
  • Figure US20080114037A1-20080515-C00084
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) of the bis-trifluoroacetic acid salt: δ ppm 6.11 (s, 2H) 6.91 (d, J=8.34 Hz, 1H) 7.24 (t, J=7.33 Hz, 1H) 7.33-7.41 (m, 3H) 7.48-7.56 (m, 3H) 7.64 (d, J=8.34 Hz, 2H) 7.79 (dd, J=8.08, 5.81 Hz, 1H) 8.00 (d, J=8.34 Hz, 2H) 9.16 (br. s., 1H) 9.23 (d, J=7.83 Hz, 1H) 9.35-9.45 (m, 1H) 9.82 (s, 1H). ESI-MS: m/z 420.5 (M+H)+.
  • Example 27 N-(4-Amino-biphenyl-3-yl)-4-indazol-2-ylmethyl-benzamide
  • Figure US20080114037A1-20080515-C00085
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 5.74 (s, 2H) 6.98-7.06 (m, 1H) 7.08 (d, J=8.34 Hz, 1H) 7.18-7.25 (m, 1H) 7.28 (t, 1H) 7.35-7.48 (m, 5H) 7.58 (m, 4H) 7.71 (d, J=8.34 Hz, 1H) 7.98 (d, J=8.08 Hz, 2H) 8.53 (s, 1H) 10.02 (s, 1H). ESI-MS: m/z 419.5 (M+H)+.
  • Example 28 N-(2-Aminophenyl)-4-((3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00086
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 2.48 (s, 3H) 4.87 (s, 2H) 5.66 (s, 2H) 6.54-6.61 (m, 1H) 6.76 (dd, J=7.83, 1.26 Hz, 1H) 6.91-6.99 (m, 1H) 7.09-7.17 (m, 2H) 7.31 (d, J=8.34 Hz, 2H) 7.34-7.40 (m, 1H) 7.64 (d, J=8.34 Hz, 1H) 7.73 (d, J=8.08 Hz, 1H) 7.89 (d, J=8.34 Hz, 2H) 9.57 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 29 N-(2-Aminophenyl)-4-((3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00087
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 2.61 (s, 3H) 4.88 (s, 2H) 5.72 (s, 2H) 6.50-6.64 (m, 1H) 6.72-6.82 (m, 1H) 6.93-7.03 (m, 2H) 7.12-7.17 (m, 1H) 7.20-7.31 (m, 3H) 7.55 (d, J=8.59 Hz, 1H) 7.68 (d, J=8.59 Hz, 1H) 7.93 (d, J=8.08 Hz, 2H) 9.62 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 30 4-((1H-indazol-3-ylamino)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00088
  • The title compound was prepared using a procedure analogous to that described in Examples 1 and 2. 1H NMR (400 MHz, DMSO-D6) δ ppm 4.56 (d, J=6.15 Hz, 2H) 4.88 (s, 2H) 6.49-6.63 (m, 1H) 6.63-6.73 (m, 1H) 6.77 (d, J=7.98 Hz, 1H) 6.84-7.01 (m, 2H) 7.16 (d, J=7.48 Hz, 1H) 7.20-7.29 (m, 2H) 7.52 (d, J=8.14 Hz, 2H) 7.77 (d, J=8.14 Hz, 1H) 7.92 (d, J=8.14 Hz, 2H) 9.60 (s, 1H) 11.40 (s, 1H). ESI-MS: m/z 358.4 (M+H)+.
  • Example 31 4-((1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00089
  • 1H NMR (499 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 5.38 (s, 2H), 6.25-6.33 (m, 2H), 7.18-7.26 (m, 3H), 7.31-7.38 (m, 3H), 7.46-7.53 (m, 1H), 7.84-7.89 (m, 1H), 7.92-7.99 (m, 1H), 10.09 (s, 1H). ESI-MS: m/z 293.3 (M+H)+.
  • Example 32 N-(2-(4-((2-Aminophenyl)carbamoyl)benzyl)-2H-indazol-5-yl)morpholine-4-carboxamide
  • Figure US20080114037A1-20080515-C00090
  • 1H NMR (400 MHz, DMSO-D6) δ ppm 3.36-3.47 (m, 4H) 3.61 (m, 4H) 4.85 (s, 2H) 5.69 (s, 2H) 6.77 (m, 1H) 6.94 (m, 1H) 7.12 (m, 1H) 7.27 (m, 1H) 7.37 (d, 2H) 7.46 (m, 1H) 7.78 (s, 1H) 7.87 (d, 2H) 8.36 (s, 1H) 8.46 (m, 1H) 9.56 (s, 1H). ESI-MS: m/z 471.5 (M+H)+.
  • Example 33 Methyl 3-(4-((2H-indazol-2-yl)methyl)benzamido)-4-aminobenzoate
  • Figure US20080114037A1-20080515-C00091
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.73 (s, 14H), 5.72 (s, 10H), 5.81 (s, 10H), 6.74 (d, J=8.59 Hz, 6H), 6.98-7.07 (m, 6H), 7.17-7.26 (m, 6H), 7.41 (d, J=8.34 Hz, 10H), 7.51-7.61 (m, 11H), 7.67-7.77 (m, 11H), 7.94 (d, J=8.08 Hz, 10H), 8.52 (s, 6H), 9.60 (s, 1H). ESI-MS: m/z 401.4 (M+H)+.
  • Example 34 N-(2-aminophenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00092
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.00 (s, 3H), 4.87 (s, 2H), 5.29-5.33 (m, 2H), 6.54-6.60 (m, 1H), 6.73-6.78 (m, 1H), 6.92-6.97 (m, 1H), 7.13 (d, J=7.58 Hz, 1H), 7.25-7.30 (m, 3H), 7.58 (s, 1H), 7.91 (d, J=8.08 Hz, 2H), 9.60 (s, 1H). ESI-MS: m/z 307.4 (M+H)+.
  • Example 35 N-(2-aminophenyl)-4-((6-fluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00093
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.70 (s, 2H), 6.53-6.60 (m, 1H), 6.75 (d, J=8.08 Hz, 1H), 6.91-6.99 (m, 2H), 7.13 (d, J=8.08 Hz, 1H), 7.31 (d, J=10.36 Hz, 1H), 7.41 (d, J=8.08 Hz, 2H), 7.76-7.83 (m, 1H), 7.93 (d, J=8.08 Hz, 2H), 8.58 (s, 1H), 9.61 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 36 N-(2-aminophenyl)-4-((6-fluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00094
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.67-5.72 (m, 2H), 6.53-6.59 (m, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.91-6.96 (m, 1H), 6.99-7.06 (m, 1H), 7.11 (d, J=7.33 Hz, 1H), 7.33 (d, J=8.08 Hz, 2H), 7.59-7.66 (m, 1H), 7.81 (dd, J=8.84, 5.31 Hz, 1H), 7.88 (d, J=8.08 Hz, 2H), 8.13-8.16 (m, 1H), 9.57 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 37 N-(2-aminophenyl)-4-((5-fluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00095
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.75 (s, 2H), 6.55 (s, 1H), 6.74 (d, J=7.83 Hz, 1H), 6.92 (d, J=7.58 Hz, 1H), 7.10 (s, 1H), 7.23-7.33 (m, 3H), 7.49-7.59 (m, 1H), 7.75 (s, 1H), 7.87 (s, 2H), 8.11 (s, 1H), 9.57 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 38 N-(2-aminophenyl)-4-((5-fluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00096
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.68 (s, 2H), 6.52-6.59 (m, 1H), 6.71-6.77 (m, 1H), 6.91-6.98 (m, 1H), 7.09-7.17 (m, 2H), 7.37-7.48 (m, 3H), 7.62-7.68 (m, 1H), 7.93 (d, J=8.34 Hz, 2H), 8.50 (s, 1H), 9.60 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 39 4-(1-(1H-indazol-1-yl)propan-2-yl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00097
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 1.20 (d, J=6.82 Hz, 3H), 3.51-3.62 (m, 1H), 4.54-4.65 (m, 2H), 6.95 (s, 2H), 7.01-7.13 (m, 3H), 7.25 (d, J=7.58 Hz, 1H), 7.29-7.35 (m, 1H), 7.44 (d, J=8.34 Hz, 2H), 7.65-7.72 (m, 2H), 7.85 (d, J=8.08 Hz, 2H), 8.02 (s, 1H), 9.84-10.01 (m, 1H). ESI-MS: m/z 371.4 (M+H)+.
  • Example 40 4-(2-(1H-indazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00098
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.17-3.28 (m, 2H), 4.68 (t, J=7.07 Hz, 2H), 4.85 (s, 2H), 6.51-6.62 (m, 1H), 6.75 (dd, J=8.08, 1.26 Hz, 1H), 6.88-7.00 (m, 1H), 7.03-7.18 (m, 2H), 7.26-7.38 (m, 3H), 7.62 (d, J=8.34 Hz, 1H), 7.72 (d, J=8.08 Hz, 1H), 7.78-7.86 (m, 2H), 8.04 (s, 1H), 9.56 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 41 4-(2-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00099
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.34 (t, J=7.07 Hz, 2H), 4.72 (t, J=7.07 Hz, 2H), 4.85 (s, 2H), 6.51-6.62 (m, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.88-7.03 (m, 2H), 7.11 (d, J=7.33 Hz, 1H), 7.15-7.25 (m, 1H), 7.29 (d, J=8.08 Hz, 2H), 7.54-7.68 (m, 2H), 7.84 (d, J=8.08 Hz, 2H), 8.19-8.30 (m, 1H), 9.56 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 42 N-(2-aminophenyl)-4-((4-chloro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00100
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.85 (s, 2H), 5.74-5.81 (m, 2H), 6.52-6.59 (m, 1H), 6.71-6.77 (m, 1H), 6.90-6.97 (m, 1H), 7.11 (d, J=7.07 Hz, 1H), 7.23 (d, J=7.58 Hz, 1H), 7.31 (d, J=8.34 Hz, 2H), 7.35-7.42 (m, 1H), 7.74 (d, J=8.59 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 8.18-8.23 (m, 1H), 9.57 (s, 1H). ESI-MS: m/z 377.8 (M+H)+.
  • Example 43 N-(2-aminophenyl)-4-((4-chloro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00101
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.70-5.78 (m, 2H), 6.53-6.61 (m, 1H), 6.75 (dd, J=8.08, 1.26 Hz, 1H), 6.91-6.98 (m, 1H), 7.13 (d, J=7.07 Hz, 2H), 7.22 (dd, J=8.59, 7.07 Hz, 1H), 7.41-7.49 (m, 2H), 7.58 (d, J=8.59 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 8.67-8.71 (m, 1H), 9.61 (s, 1H). ESI-MS: m/z 377.8 (M+H)+.
  • Example 44 N-(2-aminophenyl)-4-((4,6-difluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00102
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.67-5.75 (m, 2H), 6.52-6.59 (m, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.91-6.96 (m, 1H), 7.00-7.07 (m, 1H), 7.12 (d, J=7.58 Hz, 1H), 7.34 (d, J=8.08 Hz, 2H), 7.60 (d, J=9.35 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 8.25-8.30 (m, 1H), 9.58 (s, 1H). ESI-MS: m/z 379.4 (M+H)+.
  • Example 45 N-(2-aminophenyl)-4-((4,6-difluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00103
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.67-5.74 (m, 2H), 6.52-6.61 (m, 1H), 6.75 (dd, J=7.83, 1.26 Hz, 1H), 6.88-6.98 (m, 2H), 7.13 (d, J=7.07 Hz, 1H), 7.25 (d, J=8.84 Hz, 1H), 7.44 (d, J=8.34 Hz, 2H), 7.93 (d, J=8.08 Hz, 2H), 8.82 (s, 1H), 9.61 (s, 1H). ESI-MS: m/z 379.4 (M+H)+.
  • Example 46 N-(2-aminophenyl)-4-((7-fluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00104
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.78 (s, 2H), 6.51-6.58 (m, 1H), 6.71-6.76 (m, 1H), 6.89-6.96 (m, 1H), 7.07-7.14 (m, 2H), 7.17-7.25 (m, 3H), 7.61 (d, J=7.83 Hz, 1H), 7.88 (d, J=8.08 Hz, 2H), 8.24 (d, J=2.53 Hz, 1H), 9.56 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 47 N-(2-aminophenyl)-4-((7-fluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00105
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.72-5.78 (m, 2H), 6.52-6.60 (m, 1H), 6.75 (dd, J=8.08, 1.26 Hz, 1H), 6.92-7.04 (m, 3H), 7.13 (d, J=7.33 Hz, 1H), 7.43 (d, J=8.08 Hz, 2H), 7.51-7.57 (m, 1H), 7.94 (d, J=8.08 Hz, 2H), 8.66 (d, J=2.78 Hz, 1H), 9.62 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 48 N-(2-aminophenyl)-4-((4-fluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00106
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.77 (s, 2H), 6.50-6.60 (m, 1H), 6.73 (d, J=8.34 Hz, 1H), 6.88-6.97 (m, 2H), 7.11 (d, J=7.58 Hz, 1H), 7.32 (d, J=8.08 Hz, 2H), 7.34-7.42 (m, 1H), 7.58 (d, J=8.34 Hz, 1H), 7.89 (d, J=7.83 Hz, 2H), 8.25 (s, 1H), 9.57 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 49 N-(2-aminophenyl)-4-((4-fluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00107
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.69-5.75 (m, 2H), 6.52-6.60 (m, 1H), 6.72-6.82 (m, 2H), 6.91-6.97 (m, 1H), 7.12 (d, J=7.33 Hz, 1H), 7.17-7.24 (m, 1H), 7.39-7.46 (m, 3H), 7.93 (d, J=8.08 Hz, 2H), 8.74 (s, 1H), 9.61 (s, 1H). ESI-MS: m/z 361.4 (M+H)+.
  • Example 50 (R)-4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00108
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.18 (d, J=7.07 Hz, 4H), 3.36 (d, J=5.05 Hz, 2H), 4.29 (q, 1H), 5.07 (s, 2H), 6.24 (d, J=7.07 Hz, 1H), 6.76 (s, 1H), 6.96 (s, 1H), 7.13 (s, 1H), 7.24 (d, J=8.59 Hz, 2H), 7.32 (s, 1H), 7.39-7.44 (m, 1H), 7.63 (d, J=8.08 Hz, 3H), 7.80 (d, J=8.84 Hz, 2H), 7.90 (d, J=8.34 Hz, 1H), 8.11 (d, J=8.34 Hz, 3H), 8.75 (s, 1H), 9.79 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 51 (S)-4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00109
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.18 (d, J=7.07 Hz, 4H), 3.36 (d, J=5.05 Hz, 2H), 4.29 (q, 1H), 5.07 (s, 2H), 6.24 (d, J=7.07 Hz, 1H), 6.76 (s, 1H), 6.96 (s, 1H), 7.13 (s, 1H), 7.24 (d, J=8.59 Hz, 2H), 7.32 (s, 1H), 7.39-7.44 (m, 1H), 7.63 (d, J=8.08 Hz, 3H), 7.80 (d, J=8.84 Hz, 2H), 7.90 (d, J=8.34 Hz, 1H), 8.11 (d, J=8.34 Hz, 3H), 8.75 (s, 1H), 9.79 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 52 N-(2-aminophenyl)-4-((7-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00110
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.51 (s, 3H), 4.86 (s, 2H), 5.62-5.71 (m, 2H), 6.51-6.59 (m, 1H), 6.73 (d, J=7.83 Hz, 1H), 6.89-6.97 (m, 1H), 7.04-7.14 (m, 2H), 7.15-7.25 (m, 3H), 7.56 (d, J=8.08 Hz, 1H), 7.87 (d, J=7.83 Hz, 2H), 9.56 (s, 1H). ESI-MS: m/z 375.4 (M+H)+.
  • Example 53 N-(2-aminophenyl)-4-((7-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00111
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.61 (s, 3H), 4.87 (s, 2H), 5.74 (s, 2H), 6.53-6.59 (m, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.91-7.02 (m, 3H), 7.12 (d, J=7.58 Hz, 1H), 7.29 (d, J=8.08 Hz, 2H), 7.51 (d, J=8.34 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 9.61 (s, 1H). ESI-MS: m/z 375.4 (M+H)+.
  • Example 54 N-(2-aminophenyl)-4-((4-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00112
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.53-2.58 (m, 3H), 4.86 (s, 2H), 5.65 (s, 2H), 6.51-6.59 (m, 1H), 6.74 (dd, J=7.83, 1.26 Hz, 1H), 6.84 (dd, J=10.86, 7.58 Hz, 1H), 6.90-6.97 (m, 1H), 7.11 (d, J=7.83 Hz, 1H), 7.25-7.36 (m, 3H), 7.48 (d, J=8.59 Hz, 1H), 7.88 (d, J=8.08 Hz, 2H), 9.56 (s, 1H). ESI-MS: m/z 375.4 (M+H)+.
  • Example 55 N-(2-aminophenyl)-4-((4-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00113
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.65-2.71 (m, 3H), 4.87 (s, 2H), 5.71 (s, 2H), 6.53-6.60 (m, 1H), 6.66-6.77 (m, 2H), 6.91-6.98 (m, 1H), 7.09-7.19 (m, 2H), 7.29 (d, J=8.08 Hz, 2H), 7.36 (d, J=8.59 Hz, 1H), 7.92 (d, J=8.34 Hz, 2H), 9.61 (s, 1H). ESI-MS: m/z 375.4 (M+H)+.
  • Example 56 N-(2-aminophenyl)-4-((5,6-difluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00114
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.45 (s, 3H), 4.86 (s, 2H), 5.60 (s, 2H), 6.51-6.60 (m, 1H), 6.74 (d, J=8.08 Hz, 1H), 6.89-6.98 (m, 1H), 7.12 (d, J=7.83 Hz, 1H), 7.32 (d, J=7.83 Hz, 2H), 7.78-7.84 (m, 1H), 7.85-7.91 (m, 3H), 9.56 (s, 1H). ESI-MS: m/z 393.4 (M+H)+.
  • Example 57 N-(2-aminophenyl)-4-((5,6-difluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00115
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.57 (s, 3H), 4.98 (s, 1H), 5.67 (s, 2H), 6.58 (s, 1H), 6.76 (s, 1H), 6.95 (s, 1H), 7.13 (s, 1H), 7.27 (s, 2H), 7.55 (s, 1H), 7.75 (s, 1H), 7.90 (s, 3H), 9.62 (s, 1H). ESI-MS: m/z 393.4 (M+H)+.
  • Example 58 N-(2-aminophenyl)-4-((6-methoxy-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00116
  • 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 3.31 (s, 3H), 4.85 (s, 2H), 5.68 (s, 2H), 6.56 (s, 1H), 6.72-6.79 (m, 2H), 6.93 (s, 1H), 7.11 (s, 1H), 7.20 (s, 1H), 7.30 (d, J=8.08 Hz, 2H), 7.61 (s, 1H), 7.88 (d, J=8.08 Hz, 2H), 7.98 (s, 1H), 9.56 (s, 1H). ESI-MS: m/z 373.4 (M+H)+.
  • Example 59 N-(2-aminophenyl)-4-((6-methoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00117
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.71-3.79 (m, 3H), 4.87 (s, 2H), 5.63 (s, 2H), 6.51-6.62 (m, 1H), 6.64-6.71 (m, 1H), 6.72-6.78 (m, 1H), 6.90 (d, J=1.77 Hz, 1H), 6.91-6.97 (m, 1H), 7.13 (d, J=7.83 Hz, 1H), 7.38 (d, J=8.34 Hz, 2H), 7.57 (d, J=9.09 Hz, 1H), 7.92 (d, J=8.08 Hz, 2H), 8.38 (s, 1H), 9.60 (s, 1H). ESI-MS: m/z 373.4 (M+H)+.
  • Example 60 N-(2-aminophenyl)-4-((3-chloro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00118
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.72 (s, 2H), 6.52-6.59 (m, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.90-6.97 (m, 1H), 7.11 (d, J=6.57 Hz, 1H), 7.22-7.30 (m, 1H), 7.35 (d, J=8.34 Hz, 2H), 7.47-7.54 (m, 1H), 7.68 (d, J=8.08 Hz, 1H), 7.82 (d, J=8.59 Hz, 1H), 7.89 (d, J=8.34 Hz, 2H), 9.57 (s, 1H). ESI-MS: m/z 377.8 (M+H)+.
  • Example 61 N-(2-aminophenyl)-4-((3-chloro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00119
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.73-5.82 (m, 2H), 6.51-6.61 (m, 1H), 6.74 (d, J=8.08 Hz, 1H), 6.90-6.99 (m, 1H), 7.10-7.19 (m, 2H), 7.29-7.37 (m, 3H), 7.53 (s, 1H), 7.57-7.68 (m, 2H), 7.93 (d, J=7.83 Hz, 2H), 9.61 (s, 1H). ESI-MS: m/z 377.8 (M+H)+.
  • Example 62 4-((3-amino-5-(trifluoromethyl)-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00120
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.47 (s, 2H), 6.53-6.60 (m, 1H), 6.73-6.80 (m, 3H), 6.90-6.97 (m, 1H), 7.13 (d, J=7.58 Hz, 1H), 7.20-7.25 (m, 1H), 7.25-7.34 (m, 3H), 7.90 (d, J=8.08 Hz, 2H), 8.17 (s, 1H), 9.59 (s, 1H). ESI-MS: m/z 425.5 (M+H)+.
  • Example 63 4-((3-amino-5-(trifluoromethyl)-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00121
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.85 (s, 2H), 5.45 (s, 2H), 5.85 (s, 2H), 6.56 (td, J=7.58, 1.26 Hz, 1H), 6.74 (dd, J=8.08, 1.26 Hz, 1H), 6.89-6.97 (m, 1H), 7.10 (s, 1H), 7.28 (d, J=8.08 Hz, 2H), 7.50-7.57 (m, 1H), 7.66 (d, J=8.84 Hz, 1H), 7.87 (d, J=8.08 Hz, 2H), 8.20 (s, 1H), 9.55 (s, 1H). ESI-MS: m/z 425.5 (M+H)+.
  • Example 64 N-(2-aminophenyl)-4-((3-oxo-2,3-dihydro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00122
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 5.46 (s, 2H), 6.92 (s, 1H), 7.01 (t, J=7.33 Hz, 2H), 7.12 (s, 1H), 7.25 (s, 1H), 7.33 (d, J=7.83 Hz, 3H), 7.55 (d, J=8.34 Hz, 1H), 7.64 (d, J=8.08 Hz, 1H), 7.90 (d, J=7.58 Hz, 2H), 9.92 (s, 1H). ESI-MS: m/z 359.3 (M+H)+.
  • Example 65 N-(2-aminophenyl)-4-((3-oxo-1H-indazol-2(3H)-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00123
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 5.51 (s, 2H), 6.89 (s, 1H), 7.02 (s, 2H), 7.15 (s, 1H), 7.27 (s, 1H), 7.37 (s, 2H), 7.66 (s, 3H), 8.02 (d, J=7.33 Hz, 2H), 9.96 (s, 1H), 11.98 (s, 1H). ESI-MS: m/z 359.3 (M+H)+.
  • Example 66 N-(2-aminophenyl)-4-((6-methoxy-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00124
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.41-2.46 (m, 3H), 3.78-3.84 (m, 3H), 4.88 (s, 2H), 5.60 (s, 2H), 6.54-6.62 (m, 1H), 6.72-6.79 (m, 2H), 6.92-6.99 (m, 1H), 7.11-7.18 (m, 2H), 7.32 (d, J=8.34 Hz, 2H), 7.58 (d, J=8.59 Hz, 1H), 7.90 (d, J=8.34 Hz, 2H), 9.58 (s, 1H). ESI-MS: m/z 387.2 (M+H)+.
  • Example 67 N-(2-aminophenyl)-4-((6-methoxy-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00125
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.55 (s, 3H), 3.79 (s, 3H), 4.88 (s, 2H), 5.63 (s, 2H), 6.55-6.62 (m, 1H), 6.62-6.68 (m, 1H), 6.77 (dd, J=7.83, 1.26 Hz, 1H), 6.86 (d, J=2.02 Hz, 1H), 6.93-7.00 (m, 1H), 7.14 (d, J=7.33 Hz, 1H), 7.27 (d, J=8.34 Hz, 2H), 7.55 (d, J=9.09 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 9.62 (s, 1H). ESI-MS: m/z 387.2 (M+H)+.
  • Example 68 N-(2-aminophenyl)-4-((6-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00126
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.47-2.49 (m, 3H), 4.87 (s, 2H), 5.61 (s, 2H), 6.55-6.61 (m, 1H), 6.76 (dd, J=8.08, 1.52 Hz, 1H), 6.93-7.02 (m, 2H), 7.14 (d, J=7.83 Hz, 1H), 7.34 (d, J=8.34 Hz, 2H), 7.54-7.61 (m, 1H), 7.77 (dd, J=8.84, 5.31 Hz, 1H), 7.90 (d, J=8.34 Hz, 2H), 9.57 (s, 1H). ESI-MS: m/z 375.3 (M+H)+.
  • Example 69 N-(2-aminophenyl)-4-((6-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00127
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.59-2.63 (m, 3H), 4.88 (s, 2H), 5.69 (s, 2H), 6.53-6.62 (m, 1H), 6.78 (dd, J=7.75, 1.52 Hz, 1H), 6.87-6.99 (m, 2H), 7.15 (d, J=7.33 Hz, 1H), 7.24-7.35 (m, 3H), 7.77 (dd, J=9.09, 5.56 Hz, 1H), 7.94 (d, J=8.08 Hz, 2H), 9.62 (s, 1H). ESI-MS: m/z 375.3 (M+H)+.
  • Example 70 N-(2-aminophenyl)-4-((5-fluoro-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00128
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.46-2.49 (m, 3H), 4.87 (s, 2H), 5.66 (s, 2H), 6.53-6.62 (m, 1H), 6.77 (d, J=1.26 Hz, 1H), 6.91-7.01 (m, 1H), 7.14 (d, J=7.07 Hz, 1H), 7.25-7.35 (m, 3H), 7.55 (d, J=2.27 Hz, 1H), 7.70 (dd, J=9.09, 4.29 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 9.57 (s, 1H). ESI-MS: m/z 375.3 (M+H)+.
  • Example 71 N-(2-aminophenyl)-4-((5-fluoro-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00129
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.58 (s, 3H), 4.88 (s, 2H), 5.72 (s, 2H), 6.55-6.62 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.93-7.00 (m, 1H), 7.14 (td, J=9.35, 2.53 Hz, 2H), 7.29 (d, J=8.34 Hz, 2H), 7.46 (dd, J=9.60, 2.02 Hz, 1H), 7.62 (dd, J=9.35, 4.55 Hz, 1H), 7.94 (d, J=8.34 Hz, 2H), 9.62 (s, 1H). ESI-MS: m/z 375.4 (M+H)+.
  • Example 72 N-(2-aminophenyl)-4-((3-ethyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00130
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.29-1.36 (m, 3H), 2.95 (q, J=7.58 Hz, 2H), 4.87 (s, 2H), 5.67 (s, 2H), 6.53-6.62 (m, 1H), 6.72-6.81 (m, 1H), 6.91-7.00 (m, 1H), 7.08-7.18 (m, 2H), 7.27-7.39 (m, 3H), 7.63 (d, J=8.34 Hz, 1H), 7.77 (d, J=8.08 Hz, 1H), 7.89 (d, J=8.08 Hz, 1H), 9.57 (s, 1H). ESI-MS: m/z 371.4 (M+H)+.
  • Example 73 N-(2-aminophenyl)-4-((3-ethyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00131
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.09-1.20 (m, 3H), 3.09 (q, J=7.58 Hz, 2H), 4.89 (s, 2H), 5.75 (s, 2H), 6.55-6.62 (m, 1H), 6.74-6.80 (m, 1H), 6.93-7.03 (m, 2H), 7.14 (d, J=7.58 Hz, 1H), 7.21-7.31 (m, 3H), 7.56 (d, J=8.84 Hz, 1H), 7.72 (d, J=8.59 Hz, 1H), 7.93 (d, J=8.34 Hz, 2H), 9.63 (s, 1H). ESI-MS: m/z 371.4 (M+H)+.
  • Example 74 N-(2-aminophenyl)-4-((6-hydroxy-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00132
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.42 (s, 3H), 5.52 (s, 2H), 6.59-6.69 (m, 1H), 6.74 (d, J=1.77 Hz, 1H), 6.94 (s, 1H), 7.04 (s, 1H), 7.08-7.20 (m, 2H), 7.23-7.34 (m, 3 H), 7.50 (d, J=8.84 Hz, 1H), 7.92 (d, J=8.34 Hz, 2H), 9.95 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 75 N-(2-aminophenyl)-4-((6-hydroxy-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00133
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 2.54 (s, 3H), 3.97 (s, 1H), 5.62 (s, 2H), 6.56-6.65 (m, 1H), 6.69 (d, J=1.52 Hz, 1H), 7.01 (s, 1H), 7.10 (d, J=7.83 Hz, 1H), 7.14-7.23 (m, 1H), 7.30 (d, J=8.08 Hz, 3H), 7.52 (d, J=8.84 Hz, 1H), 7.96 (d, J=8.34 Hz, 2H), 10.07 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 76 N-(2-aminophenyl)-4-((3-phenyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00134
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.86 (s, 2H), 5.78-5.87 (m, 2H), 6.53-6.61 (m, 1H), 6.75 (dd, J=8.08, 1.26 Hz, 1H), 6.91-6.99 (m, 1H), 7.13 (d, J=7.83 Hz, 1H), 7.22-7.30 (m, 1H), 7.38-7.49 (m, 3H), 7.50-7.57 (m, 2H), 7.80 (d, J=8.34 Hz, 1H), 7.91 (d, J=8.34 Hz, 2H), 8.00 (d, J=7.07 Hz, 2H), 8.11 (d, J=8.08 Hz, 1H), 9.57 (s, 1H). ESI-MS: m/z 419.4 (M+H)+.
  • Example 77 N-(2-aminophenyl)-4-((3-phenyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00135
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.77 (s, 2H), 6.54-6.61 (m, 1H), 6.76 (dd, J=8.08, 1.26 Hz, 1H), 6.92-6.99 (m, 1H), 7.09-7.17 (m, 3H), 7.30-7.37 (m, 1H), 7.52-7.63 (m, 3H), 7.69 (d, J=8.84 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 9.60 (s, 1H). ESI-MS: m/z 419.4 (M+H)+.
  • Example 78 N-(2-aminophenyl)-4-((5-methoxy-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00136
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: 6 ppm 3.78 (s, 3H), 5.71 (s, 2H), 6.53-6.64 (m, 1H), 6.78 (s, 1H), 6.91-7.00 (m, 1H), 7.01-7.08 (m, 1H), 7.13 (s, 1H), 7.20 (d, J=2.27 Hz, 1H), 7.30 (d, J=8.08 Hz, 2H), 7.61 (d, J=9.09 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 8.01 (s, 1H), 9.59 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 79 N-(2-aminophenyl)-4-((5-methoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00137
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.77 (s, 3H), 5.69 (s, 2H), 6.84-6.95 (m, 2H), 6.95-7.05 (m, 2H), 7.07-7.15 (m, 1H), 7.26 (d, J=7.58 Hz, 1H), 7.42 (d, J=8.34 Hz, 2H), 7.51 (d, J=9.09 Hz, 1H), 7.96 (d, J=8.34 Hz, 2H), 8.35 (s, 1H), 9.91 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 80 N-(2-aminophenyl)-4-((3,5-dimethyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00138
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.40 (s, 3H), 2.44-2.48 (m, 3H), 4.87 (s, 2H), 5.62 (s, 2H), 6.54-6.61 (m, 1H), 6.76 (dd, J=8.08, 1.26 Hz, 1H), 6.92-6.99 (m, 1H), 7.13 (d, J=7.07 Hz, 1H), 7.20 (dd, J=8.59, 1.26 Hz, 1H), 7.28 (d, J=8.08 Hz, 2H), 7.49 (s, 1H), 7.52 (d, J=8.59 Hz, 1H), 7.88 (d, J=8.08 Hz, 2H), 9.57 (s, 1H). ESI-MS: m/z 371.4 (M+H)+.
  • Example 81 N-(2-aminophenyl)-4-((3,5-dimethyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00139
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.36 (s, 3H), 2.56 (s, 3H), 4.88 (s, 2H), 5.69 (s, 2H), 6.54-6.62 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.93-7.00 (m, 1H), 7.07 (dd, J=8.84, 1.52 Hz, 1H), 7.14 (d, J=7.07 Hz, 1H), 7.26 (d, J=8.34 Hz, 2H), 7.41 (s, 1H), 7.45 (d, J=8.84 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 9.62 (s, 1H). ESI-MS: m/z 371.4 (M+H)+.
  • Example 82 N-(2-aminophenyl)-4-((7-methoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00140
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.88 (s, 3H), 4.89 (s, 2H), 5.70 (s, 2H), 6.54-6.65 (m, 2H), 6.76 (d, J=7.58 Hz, 1H), 6.90-7.01 (m, 2H), 7.15 (s, 1H), 7.24 (d, J=8.34 Hz, 1H), 7.41 (d, J=7.58 Hz, 2H), 7.95 (d, J=8.08 Hz, 2H), 8.47 (s, 1H), 9.62 (s, 1H). ESI-MS: m/z 373.1 (M+H)+.
  • Example 83 N-(2-aminophenyl)-4-((4-methoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00141
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.89 (s, 3H), 4.89 (s, 2H), 5.69 (s, 2H), 6.36-6.46 (m, 1H), 6.53-6.64 (m, 1H), 6.76 (d, J=8.08 Hz, 1H), 6.91-7.02 (m, 1H), 7.11-7.20 (m, 3H), 7.42 (d, J=7.83 Hz, 2H), 7.94 (d, J=7.83 Hz, 2H), 8.55 (s, 1H), 9.62 (s, 1H). ESI-MS: m/z 373.2 (M+H)+.
  • Example 84 N-(2-aminophenyl)-4-((7-methoxy-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00142
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.92 (s, 3H), 4.87 (s, 2H), 5.85 (s, 2H), 6.54-6.61 (m, 1H), 6.76 (d, J=7.83 Hz, 1H), 6.87 (d, J=7.58 Hz, 1H), 6.92-6.99 (m, 1H), 7.03-7.10 (m, 1H), 7.13 (d, J=7.83 Hz, 1H), 7.24 (d, J=7.83 Hz, 2H), 7.32 (d, J=8.08 Hz, 1H), 7.88 (d, J=7.58 Hz, 2H), 8.09 (s, 1H), 9.56 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 85 N-(2-aminophenyl)-4-((4-methoxy-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00143
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.93 (s, 3H), 4.87 (s, 2H), 5.72 (s, 2H), 6.54-6.63 (m, 2H), 6.76 (d, J=7.83 Hz, 1H), 6.90-7.00 (m, 1H), 7.13 (d, J=7.83 Hz, 1H), 7.23-7.33 (m, 4H), 7.89 (d, J=7.83 Hz, 2H), 8.10 (s, 1H), 9.58 (s, 1H). ESI-MS: m/z 373.3 (M+H)+.
  • Example 86 4-((6-acetamido-3-methyl-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00144
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.06 (s, 3H), 2.53-2.57 (m, 2H), 4.88 (s, 2H), 5.66 (s, 2H), 6.53-6.63 (m, 1H), 6.76 (dd, J=8.08, 1.26 Hz, 1H), 6.93-7.04 (m, 2H), 7.14 (d, J=7.07 Hz, 1H), 7.27 (d, J=8.08 Hz, 2H), 7.59 (d, J=9.09 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 7.99 (s, 1H), 9.62 (s, 1H), 9.91 (s, 1H). ESI-MS: m/z 414.3 (M+H)+.
  • Example 87 4-((6-acetamido-3-methyl-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00145
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.06 (s, 3H), 2.44-2.48 (m, 3H), 4.88 (s, 2H), 5.56 (s, 2H), 6.53-6.63 (m, 1H), 6.73-6.81 (m, 1H), 6.91-7.01 (m, 1H), 7.08-7.19 (m, 2H), 7.24 (d, J=8.08 Hz, 2H), 7.63 (d, J=8.59 Hz, 1H), 7.89 (d, J=8.08 Hz, 2H), 8.01 (s, 1H), 9.58 (s, 1H), 10.09 (s, 1H). ESI-MS: m/z 414.3 (M+H)+.
  • Example 88 4-((3-amino-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00146
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.88 (s, 2H), 5.46 (s, 2H), 6.27 (s, 2H), 6.58 (t, J=7.45 Hz, 1H), 6.65 (t, J=7.33 Hz, 1H), 6.76 (d, J=8.08 Hz, 1H), 6.96 (t, J=7.58 Hz, 1H), 7.05 (t, J=7.58 Hz, 1H), 7.12-7.20 (m, 2H), 7.29 (d, J=7.83 Hz, 2H), 7.63 (d, J=8.34 Hz, 1H), 7.91 (d, J=7.83 Hz, 2H), 9.6 (s, 1H). ESI-MS: m/z 358.2 (M+H)+.
  • Example 89 4-((6-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00147
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.93-2.13 (m, 3H), 4.87 (s, 2H), 5.66 (s, 2H), 6.53-6.61 (m, 1H), 6.75 (dd, J=7.96, 1.39 Hz, 1H), 6.91-6.98 (m, 1H), 7.13 (dd, J=8.72, 1.64 Hz, 2H), 7.24 (d, J=8.34 Hz, 2H), 7.69 (d, J=8.59 Hz, 1H), 7.90 (d, J=8.08 Hz, 2H), 8.05 (s, 1H), 8.08 (s, 1H), 9.59 (s, 1H), 10.11 (s, 1H). ESI-MS: m/z 400.2 (M+H)+.
  • Example 90 4-((6-acetamido-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00148
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.06 (s, 3H), 4.88 (s, 2H), 5.67 (s, 2H), 6.51-6.65 (m, 1H), 6.76 (dd, J=8.08, 1.26 Hz, 1H), 6.89-7.00 (m, 1H), 7.06 (dd, J=8.84, 1.77 Hz, 1H), 7.14 (d, J=7.33 Hz, 1H), 7.40 (d, J=8.08 Hz, 2H), 7.63 (d, J=8.34 Hz, 1H), 7.94 (d, J=8.08 Hz, 2H), 8.04 (s, 1H), 8.42 (s, 1H), 9.62 (s, 1H), 9.93 (s, 1H). ESI-MS: m/z 400.2 (M+H)+.
  • Example 91 N-(2-aminophenyl)-4-((3-methyl-7-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00149
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.66 (s, 3H), 4.89 (s, 2H), 5.82 (s, 2H), 6.58 (t, J=7.45 Hz, 1H), 6.76 (d, J=7.83 Hz, 1H), 6.96 (t, J=7.58 Hz, 1H), 7.14 (ddd, J=7.26, 3.92, 3.73 Hz, 2H), 7.28 (d, J=8.08 Hz, 2H), 7.66 (d, J=6.82 Hz, 1H), 7.94 (d, J=8.08 Hz, 2H), 8.05 (d, J=8.34 Hz, 1H), 9.62 (s, 1H). ESI-MS: m/z 425.2 (M+H)+.
  • Example 92 N-(2-aminophenyl)-4-((3-methyl-6-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00150
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.67 (s, 3H), 4.88 (s, 2H), 5.80 (s, 2H), 6.58 (t, J=7.58 Hz, 1H), 6.76 (d, J=7.83 Hz, 1H), 6.96 (t, J=7.71 Hz, 1H), 7.14 (d, J=7.83 Hz, 1H), 7.22 (d, J=8.84 Hz, 1H), 7.30 (d, J=8.08 Hz, 2H), 7.92-8.02 (m, 4H), 9.63 (s, 1H). ESI-MS: m/z 425.2 (M+H)+.
  • Example 93 N-(2-aminophenyl)-4-((3-methyl-6-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00151
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.55 (s, 3H), 4.87 (s, 2H), 5.79 (s, 2H), 6.57 (t, J=7.33 Hz, 1H), 6.75 (d, J=8.08 Hz, 1H), 6.95 (t, J=7.58 Hz, 1H), 7.13 (d, J=7.58 Hz, 1H), 7.33 (d, J=8.08 Hz, 2H), 7.41 (d, J=8.59 Hz, 1H), 7.90 (d, J=7.83 Hz, 2H), 7.98 (d, J=8.34 Hz, 1H), 8.24 (s, 1H), 9.58 (s, 1H). ESI-MS: m/z 425.2 (M+H)+.
  • Example 94 N-(2-aminophenyl)-4-((3-methyl-5-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00152
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.57 (s, 3H), 4.87 (s, 2H), 5.73 (s, 2H), 6.57 (t, J=7.45 Hz, 1H), 6.75 (d, J=7.83 Hz, 1H), 6.95 (t, J=7.45 Hz, 1H), 7.13 (d, J=7.58 Hz, 1H), 7.33 (d, J=7.83 Hz, 2H), 7.67 (d, J=8.84 Hz, 1H), 7.90 (d, J=8.34 Hz, 3H), 8.22 (s, 1H), 9.58 (s, 1H). ESI-MS: m/z 425.2 (M+H)+.
  • Example 95 N-(2-aminophenyl)-4-((3-methyl-5-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00153
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.70 (s, 3H), 4.88 (s, 2H), 5.78 (s, 2H), 6.58 (t, J=7.45 Hz, 1H), 6.76 (d, J=8.08 Hz, 1H), 6.93-6.99 (m, 1H), 7.14 (d, J=7.33 Hz, 1H), 7.31 (d, J=8.08 Hz, 2H), 7.44 (d, J=8.84 Hz, 1H), 7.75 (d, J=9.09 Hz, 1H), 7.94 (d, J=8.08 Hz, 2H), 8.25 (s, 1H), 9.63 (s, 1H). ESI-MS: m/z 425.2 (M+H)+.
  • Example 96 4-((3-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00154
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.87 (s, 2H), 5.39 (s, 2H), 5.52 (s, 2H), 6.57 (t, J=7.45 Hz, 1H), 6.75 (d, J=7.83 Hz, 1H), 6.94 (q, J=7.75 Hz, 2H), 7.13 (d, J=7.58 Hz, 1H), 7.27 (t, J=8.21 Hz, 3H), 7.44 (d, J=8.34 Hz, 1H), 7.70 (d, J=7.83 Hz, 1H), 7.86 (d, J=7.83 Hz, 2H), 9.55 (s, 1H). ESI-MS: m/z 358.4 (M+H)+.
  • Example 97 N-(2-aminophenyl)-4-((5-hydroxy-6-methoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00155
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.81 (s, 2H) 4.89 (s, 2H) 5.58 (s, 2H) 6.59 (t, J=7.58 Hz, 1H) 6.77 (dd, J=7.96, 1.39 Hz, 1H) 6.85 (s, 1H) 6.91 (s, 1H) 6.96 (dd, J=15.16, 1.52 Hz, 1H) 7.15 (d, J=7.33 Hz, 1H) 7.36 (d, J=8.08 Hz, 2H) 7.93 (d, J=8.08 Hz, 2H) 8.11 (s, 1H) 8.82 (s, 1H) 9.62 (s, 1H). ESI-MS: m/z 389.4 (M+H)+.
  • Example 98 N-(2-aminophenyl)-4-((5,6-dimethoxy-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00156
  • 5,6-dimethoxy-1H-indazole was prepared according to the procedure described in Dennler et al., “Synthesis of indazoles using polyphosphoric acid-1” Tetrahedron, 22(9): 3131-3141 (1966), which is hereby incorporated by reference in its entirety. The title compound was then prepared using a procedure analogous to that described in Scheme 1. 1H NMR (400 MHz, DMSO-d6) δ ppm 3.78 (s, 3H) 3.83 (s, 3H) 4.88 (br. s., 2H) 5.69 (s, 2H) 6.58 (t, J=7.45 Hz, 1H) 6.77 (d, J=1.01 Hz, 1H) 6.96 (dd, J=15.16, 1.26 Hz, 1H) 7.17 (s, 1H) 7.13 (d, J=7.58 Hz, 1H) 7.27 (s, 1H) 7.30 (d, J=8.34 Hz, 2H) 7.83-7.93 (m, 3H) 9.59 (s, 1H). ESI-MS: m/z 403.4 (M+H)+.
  • Example 99 N-(2-aminophenyl)-4-((5,6-dimethoxy-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00157
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.78 (s, 3H) 3.83 (s, 3H) 4.88 (br. s., 2H) 5.69 (s, 2H) 6.58 (s, 1H) 6.77 (d, J=1.01 Hz, 1H) 6.95 (d, J=7.33 Hz, 1H) 7.17 (s, 1H) 7.13 (d, J=7.58 Hz, 1H) 7.27 (s, 1H) 7.30 (d, J=8.34 Hz, 2H) 7.82-7.94 (m, 3H) 9.59 (s, 1H). ESI-MS: m/z 403.4 (M+H)+.
  • Example 100 4-((1H-benzo[d][1,2,3]triazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00158
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.88 (s, 2H), 6.08 (s, 2H), 6.57 (t, J=7.58 Hz, 1H), 6.74-6.78 (m, 1H), 6.93-6.98 (m, 1H), 7.13 (d, J=7.58 Hz, 1H), 7.39-7.46 (m, 3H), 7.55 (t, J=7.71 Hz, 1H), 7.86 (d, J=8.34 Hz, 1H), 7.94 (d, J=8.08 Hz, 2H), 8.07 (d, J=8.34 Hz, 1H), 9.60 (s, 1H). ESI-MS: m/z 344.4 (M+H)+.
  • Example 101 4-((2H-benzo[d][1,2,3]triazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00159
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.88 (s, 2H), 6.07 (s, 2H), 6.55-6.61 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.93-6.99 (m, 1H), 7.15 (d, J=7.58 Hz, 1H), 7.43-7.48 (m, 2H), 7.50 (d, J=8.34 Hz, 2H), 7.91-7.99 (m, 4H), 9.63 (s, 1H). ESI-MS: m/z 344.4 (M+H)+.
  • Example 102 4-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00160
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.05 (s, 3H), 5.70 (s, 2H), 6.95-7.03 (m, 4H), 7.06-7.13 (m, 1H), 7.30-7.40 (m, 5H), 7.64 (d, J=9.09 Hz, 1H), 7.75 (d, J=8.84 Hz, 2H), 7.86 (d, J=8.08 Hz, 2H), 8.11 (d, J=17.68 Hz, 2H), 9.96 (s, 1H), 10.21 (s, 1H). ESI-MS: m/z 477.3 (M+H)+.
  • Example 103 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(piperazin-1-yl)ethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00161
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 3.06 (s, 2H), 3.22 (s, 4H), 3.30 (s, 4H), 3.44-3.52 (m, 2H), 5.46 (s, 2H), 6.79 (t, J=7.33 Hz, 1H), 6.93 (d, J=7.07 Hz, 1H), 7.07 (td, J=7.64, 1.39 Hz, 1H), 7.22 (d, J=8.59 Hz, 1H), 7.38 (d, J=8.34 Hz, 2H), 7.90 (d, J=0.51 Hz, 1H), 7.97 (d, J=8.08 Hz, 2H), 8.29-8.32 (m, 1H), 8.32-8.35 (m, 1H), 9.85 (s, 1H). ESI-MS: m/z 448.4 (M+H)+.
  • Example 104 4-((2H-indazol-2-yl)methyl)-N-(4-aminopyrimidin-5-yl)benzamide
  • Figure US20080114037A1-20080515-C00162
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 5.74 (s, 2H), 6.82 (s, 2H), 7.00-7.08 (m, 1H), 7.23 (dd, J=8.72, 6.69 Hz, 1H), 7.43 (d, J=8.34 Hz, 2H), 7.59 (d, J=8.84 Hz, 1H), 7.72 (d, J=8.34 Hz, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.15 (s, 1H), 8.26 (s, 1H), 8.53 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 345.3 (M+H)+.
  • Example 105 4-((5-acetamido-3-amino-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00163
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.03 (s, 3H), 4.86 (s, 2H), 5.35 (s, 2H), 5.43 (s, 2H), 6.57 (t, J=7.45 Hz, 1H), 6.75 (d, J=7.07 Hz, 1H), 6.92-6.98 (m, 1H), 7.13 (d, J=7.33 Hz, 1H), 7.24-7.31 (m, 3H), 7.35-7.41 (m, 1H), 7.86 (d, J=7.83 Hz, 2H), 7.96 (s, 1H), 9.55 (s, 1H), 9.81 (s, 1H). ESI-MS: m/z 415.3 (M+H)+.
  • Example 106 4-((5-acetamido-3-methyl-2H-indazol-2-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00164
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.05 (s, 3H), 2.53 (s, 3H), 4.88 (s, 2H), 5.68 (s, 2H), 6.58 (t, J=7.45 Hz, 1H), 6.76 (d, J=7.83 Hz, 1H), 6.93-6.98 (m, 1H), 7.14 (d, J=7.83 Hz, 1H), 7.20 (dd, J=9.22, 1.64 Hz, 1H), 7.27 (d, J=8.34 Hz, 2H), 7.49 (d, J=9.09 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 8.05 (s, 1H), 9.62 (s, 1H), 9.85 (s, 1H). ESI-MS: m/z 414.3 (M+H)+.
  • Example 107 4-((5-acetamido-3-methyl-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00165
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.04 (s, 3H), 2.45 (s, 3H), 4.87 (s, 2H), 5.61 (s, 2H), 6.54-6.60 (m, 1H), 6.75 (dd, J=7.96, 1.39 Hz, 1H), 6.92-6.98 (m, 1H), 7.13 (d, J=7.33 Hz, 1H), 7.30 (d, J=8.34 Hz, 2H), 7.38 (dd, J=9.09, 1.77 Hz, 1H), 7.57 (d, J=8.84 Hz, 1H), 7.88 (d, J=8.34 Hz, 2H), 8.04 (d, J=1.26 Hz, 1H), 9.57 (s, 1H), 9.94 (s, 1H). ESI-MS: m/z 414.3 (M+H)+.
  • Example 108 4-(1-(1H-indazol-1-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00166
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.97 (d, J=7.07 Hz, 3H), 4.86 (s, 2H), 6.16 (q, J=6.99 Hz, 1H), 6.57 (t, J=7.58 Hz, 1H), 6.75 (d, J=6.82 Hz, 1H), 6.91-6.98 (m, 1H), 7.13 (t, J=7.20 Hz, 2H), 7.34 (t, J=7.58 Hz, 1H), 7.39 (d, J=8.34 Hz, 2H), 7.66 (d, J=8.34 Hz, 1H), 7.77 (d, J=8.08 Hz, 1H), 7.87 (d, J=8.08 Hz, 2H), 8.17 (s, 1H), 9.55 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 109 4-(1-(2H-indazol-2-yl)ethyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00167
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.99 (d, J=7.07 Hz, 3H), 4.88 (s, 2H), 6.04 (q, J=6.99 Hz, 1H), 6.57 (t, J=7.58 Hz, 1H), 6.76 (d, J=8.08 Hz, 1H), 6.91-6.99 (m, 1H), 7.01-7.06 (m, 1H), 7.14 (d, J=7.58 Hz, 1H), 7.20-7.26 (m, 1H), 7.44 (d, J=8.34 Hz, 2H), 7.61 (d, J=8.84 Hz, 1H), 7.71 (d, J=8.34 Hz, 1H), 7.92 (d, J=8.34 Hz, 2H), 8.56 (s, 1H), 9.60 (s, 1H). ESI-MS: m/z 357.4 (M+H)+.
  • Example 110 5-((5-acetamido-1H-indazol-1-yl)methyl)-N-(2-aminophenyl)thiophene-2-carboxamide
  • Figure US20080114037A1-20080515-C00168
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.05 (s, 3H), 4.86 (s, 2H), 5.85 (s, 2H), 6.52-6.58 (m, 1H), 6.74 (d, J=6.82 Hz, 1H), 6.92-6.97 (m, 1H), 7.02-7.07 (m, 1H), 7.16 (d, J=3.54 Hz, 1H), 7.44 (dd, J=9.09, 1.77 Hz, 1H), 7.71 (d, J=9.09 Hz, 1H), 7.78 (d, J=3.03 Hz, 1H), 8.08 (s, 1H), 8.13 (d, J=1.26 Hz, 1H), 9.62 (s, 1H), 9.97 (s, 1H). ESI-MS: m/z 406.3 (M+H)+.
  • Example 111 4-((2H-indazol-2-yl)methyl)-N-(2-amino-5-(thiophen-2-yl)phenyl)benzamide
  • Figure US20080114037A1-20080515-C00169
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.75 (s, 2H), 6.98-7.09 (m, 3H), 7.24 (ddd, J=8.46, 6.95, 1.26 Hz, 1H), 7.31-7.35 (m, 1H), 7.39-7.47 (m, 4H), 7.54 (d, J=2.02 Hz, 1H), 7.60 (dd, J=8.84, 1.01 Hz, 1H), 7.73 (d, J=8.34 Hz, 1H), 7.98 (d, J=8.34 Hz, 2H), 8.54 (d, J=1.01 Hz, 1H), 9.96 (s, 1H). ESI-MS: m/z 425.5 (M+H)+.
  • Example 112 N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4,6-difluoro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00170
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.76 (s, 2H), 6.93 (s, 1H), 7.07 (d, J=8.59 Hz, 2H), 7.30 (d, J=3.28 Hz, 1H), 7.35-7.42 (m, 4H), 7.50 (d, J=1.77 Hz, 1H), 7.62 (d, J=9.35 Hz, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.30 (s, 1H), 9.86 (s, 1H). ESI-MS: m/z 461.3 (M+H)+.
  • Example 113 N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4,6-difluoro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00171
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.74 (s, 2H), 6.85-6.97 (m, 2H), 7.04-7.07 (m, 1H), 7.24-7.29 (m, 2H), 7.34 (dd, J=8.34, 1.77 Hz, 1H), 7.38 (d, J=5.05 Hz, 1H), 7.45-7.50 (m, 3H), 7.98 (d, J=8.08 Hz, 2H), 8.84 (s, 1H), 9.81 (s, 1H). ESI-MS: m/z 461.3 (M+H)+.
  • Example 114 N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((6-methoxy-3-methyl-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00172
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 2.54 (s, 3H), 3.78 (s, 3H), 5.64 (s, 2H), 6.65 (d, J=9.09 Hz, 1H), 6.86 (s, 2H), 7.05 (s, 1H), 7.27 (s, 4H), 7.37 (s, 1H), 7.48 (s, 1H), 7.56 (s, 1H), 7.96 (d, J=7.83 Hz, 2H), 9.78 (s, 1H). ESI-MS: m/z 469.3 (M+H)+.
  • Example 115 N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((4-methyl-1H-pyrazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00173
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.02 (s, 3H), 5.15 (s, 2H), 5.34 (s, 2H), 6.80 (d, J=8.34 Hz, 1H), 7.03-7.07 (m, 1H), 7.24 (d, J=3.28 Hz, 1H), 7.27-7.33 (m, 4H), 7.35 (d, J=5.05 Hz, 1H), 7.46 (s, 1H), 7.60 (s, 1H), 7.95 (d, J=7.83 Hz, 2H), 9.71 (s, 1H). ESI-MS: m/z 389.3(M+H)+.
  • Example 116 N-(2-amino-5-(thiophen-2-yl)phenyl)-4-((6-methoxy-3-methyl-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00174
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 2.43 (s, 3H), 3.81 (s, 3H), 5.61 (s, 2H), 6.74 (dd, J=8.84, 2.02 Hz, 1H), 7.00 (d, J=8.34 Hz, 1H), 7.08 (dd, J=5.05, 3.79 Hz, 1H), 7.15 (d, J=2.02 Hz, 1H), 7.35 (d, J=8.08 Hz, 3H), 7.40-7.45 (m, 2H), 7.52-7.60 (m, 2H), 7.93 (d, J=8.08 Hz, 2H), 9.94 (s, 1H). ESI-MS: m/z 469.3 (M+H)+.
  • Example 117 N-(2-aminophenyl)-4-((4-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00175
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.88 (s, 2H), 6.95 (t, J=7.33 Hz, 1H), 7.05 (d, J=7.33 Hz, 1H), 7.11-7.18 (m, 1H), 7.27 (d, J=7.33 Hz, 1H), 7.39 (d, J=8.34 Hz, 2H), 7.59 (s, 1H), 7.60 (d, J=2.27 Hz, 1H), 7.93 (d, J=8.08 Hz, 2H), 8.10-8.17 (m, 1H), 8.27 (s, 1H), 9.97 (s, 1H). ESI-MS: m/z 411.3 (M+H)+.
  • Example 118 N-(2-aminophenyl)-4-((4-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00176
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.83 (s, 2H), 7.11 (t, J=7.33 Hz, 1H), 7.16-7.25 (m, 2H), 7.34 (d, J=7.33 Hz, 1H), 7.38-7.44 (m, 1H), 7.52 (d, J=8.08 Hz, 3H), 7.95 (d, J=8.59 Hz, 1H), 7.99 (d, J=8.08 Hz, 2H), 8.80 (s, 1H), 10.16 (s, 1H). ESI-MS: m/z 411.3 (M+H)+.
  • Example 119 N-(2-aminophenyl)-4-((5-(trifluoromethyl)-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00177
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.85 (s, 2H), 6.93 (t, J=7.20 Hz, 1H), 7.03 (d, J=7.83 Hz, 1H), 7.13 (t, J=7.07 Hz, 1H), 7.25 (d, J=7.33 Hz, 1H), 7.36 (d, J=8.08 Hz, 2H), 7.70 (dd, J=8.84, 1.52 Hz, 1H), 7.91-7.99 (m, 3H), 8.28 (s, 1H), 8.34 (s, 1H), 9.95 (s, 1H). ESI-MS: m/z 411.3 (M+H)+.
  • Example 120 N-(2-aminophenyl)-4-((5-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00178
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.83 (s, 2H), 6.98 (t, J=7.45 Hz, 1H), 7.07 (d, J=7.83 Hz, 1H), 7.16 (t, J=7.20 Hz, 1H), 7.29 (d, J=7.58 Hz, 1H), 7.44-7.50 (m, 3H), 7.81 (d, J=9.09 Hz, 1H), 7.98 (d, J=8.08 Hz, 2H), 8.28 (s, 1H), 8.80 (s, 1H), 10.03 (s, 1H). ESI-MS: m/z 411.3 (M+H)+.
  • Example 121 N-(2-aminophenyl)-4-((6-(trifluoromethyl)-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00179
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.90 (s, 2H), 5.82 (s, 2H), 6.59 (t, J=7.45 Hz, 1H), 6.77 (d, J=7.07 Hz, 1H), 6.94-6.99 (m, 1H), 7.16 (d, J=7.58 Hz, 1H), 7.45 (d, J=8.59 Hz, 3H), 7.97 (d, J=8.34 Hz, 2H), 8.27 (s, 1H), 8.74 (s, 1H), 8.79 (s, 1H), 9.65 (s, 1H). ESI-MS: m/z 411.3 (M+H)+.
  • Example 122 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxylic acid
  • Figure US20080114037A1-20080515-C00180
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 5.47 (s, 2H), 6.93-6.99 (m, 1H), 7.05 (d, J=7.33 Hz, 1H), 7.15 (t, J=8.34 Hz, 1H), 7.28 (d, J=7.83 Hz, 1H), 7.41 (d, J=8.34 Hz, 2H), 7.85 (s, 1H), 7.97 (d, J=8.34 Hz, 2H), 8.44 (s, 1H), 10.01 (s, 1H). ESI-MS: m/z 337.3 (M+H)+.
  • Example 123 N-(2-aminophenyl)-4-((3-methyl-1H-pyrazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00181
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 2.15 (s, 3H), 5.33 (s, 2H), 6.07 (d, J=2.27 Hz, 1H), 7.11-7.16 (m, 1H), 7.18-7.26 (m, 2H), 7.34 (d, J=8.34 Hz, 2H), 7.37 (dd, J=7.96, 0.88 Hz, 1H), 7.74 (d, J=2.02 Hz, 1H), 7.98 (d, J=8.34 Hz, 2H), 10.18 (s, 1H). ESI-MS: m/z 307.4 (M+H)+.
  • Example 124 N-(2-aminophenyl)-4-((3,5-dimethyl-1H-pyrazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00182
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 2.12 (s, 3H), 2.17 (s, 3H), 5.30 (s, 2H), 5.90 (s, 1H), 7.10-7.15 (m, 1H), 7.16-7.20 (m, 1H), 7.21-7.25 (m, 3H), 7.35 (dd, J=7.83, 1.26 Hz, 1H), 7.96 (d, J=8.34 Hz, 2H), 10.15 (s, 1H). ESI-MS: m/z 321.4 (M+H)+.
  • Example 125 Ethyl 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxylate
  • Figure US20080114037A1-20080515-C00183
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 1.27 (t, J=7.07 Hz, 3H), 4.22 (q, J=7.07 Hz, 2H), 5.48 (s, 2H), 6.99 (t, J=7.33 Hz, 1H), 7.05-7.11 (m, 1H), 7.14-7.20 (m, 1H), 7.30 (d, J=7.83 Hz, 1H), 7.41 (d, J=8.34 Hz, 2H), 7.90 (s, 1H), 7.97 (d, J=8.08 Hz, 2H), 8.53 (s, 1H), 10.04 (s, 1H). ESI-MS: m/z 365.4 (M+H)+.
  • Example 126 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-2-ylmethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00184
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.49 (d, J=6.06 Hz, 2H), 4.90 (s, 2H), 5.45 (s, 2H), 6.59 (td, J=7.45, 1.26 Hz, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.16 (d, J=7.07 Hz, 1H), 7.25 (ddd, J=7.58, 4.80, 1.01 Hz, 1H), 7.30 (d, J=7.83 Hz, 1H), 7.39 (d, J=8.08 Hz, 2H), 7.75 (td, J=7.64, 1.89 Hz, 1H), 7.94-7.98 (m, 3H), 8.34 (s, 1H), 8.50 (ddd, J=4.93, 1.89, 1.01 Hz, 1H), 8.75 (t, J=5.94 Hz, 1H), 9.65 (s, 1H). ESI-MS: m/z 427.4 (M+H)+.
  • Example 127 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-phenyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00185
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.90 (s, 2H), 5.49 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.97 (td, J=7.71, 1.52 Hz, 1H), 7.04-7.09 (m, 1H), 7.16 (d, J=7.33 Hz, 1H), 7.30-7.35 (m, 2H), 7.40 (d, J=8.34 Hz, 2H), 7.70 (dt, J=8.65, 1.61 Hz, 2H), 7.97 (d, J=8.08 Hz, 2H), 8.09 (d, J=0.76 Hz, 1H), 8.49 (s, 1H), 9.66 (s, 1H), 9.86 (s, 1H). ESI-MS: m/z 412.4 (M+H)+.
  • Example 128 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-benzyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00186
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.41 (d, J=6.06 Hz, 2H), 4.90 (s, 2H), 5.44 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.07 Hz, 1H), 7.21-7.25 (m, 1H), 7.27-7.34 (m, 4H), 7.38 (d, J=8.34 Hz, 2H), 7.95 (s, 2H), 7.97 (s, 1H), 8.32 (s, 1H), 8.65 (t, J=6.06 Hz, 1H), 9.65 (s, 1H). ESI-MS: m/z 426.4 (M+H)+.
  • Example 129 N-(2-aminophenyl)-4-((4,5,6,7-tetrahydro-2H-indazol-2-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00187
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 1.63-1.74 (m, 4H), 2.45-2.49 (m, 4H), 5.27 (s, 2H), 6.90-6.98 (m, 1H), 7.04 (d, J=8.59 Hz, 1H), 7.14 (t, J=7.83 Hz, 1H), 7.28 (d, J=7.58 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.50 (s, 1H), 7.94 (d, J=8.34 Hz, 2H), 9.96 (s, 1H). ESI-MS: m/z 347.4 (M+H)+.
  • Example 130 N-(2-aminophenyl)-4-((4,5,6,7-tetrahydro-1H-indazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00188
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 1.59-1.66 (m, 2H), 1.68-1.76 (m, 2H), 2.44 (t, J=5.94 Hz, 2H), 2.47-2.50 (m, 2H), 5.31 (s, 2H), 7.04 (t, J=7.33 Hz, 1H), 7.10-7.13 (m, 1H), 7.16-7.21 (m, 1H), 7.23-7.27 (m, 3H), 7.31 (dd, J=7.96, 1.14 Hz, 1H), 7.95 (d, J=8.34 Hz, 2H), 10.07 (s, 1H). ESI-MS: m/z 347.4 (M+H)+.
  • Example 131 4-((1H-pyrazol-3-ylamino)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00189
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 4.42 (s, 2H), 5.74 (d, J=2.53 Hz, 1H), 6.85 (t, J=7.45 Hz, 1H), 6.96-7.00 (m, 1H), 7.10 (td, J=7.64, 1.39 Hz, 1H), 7.23-7.27 (m, 1H), 7.48 (d, J=8.34 Hz, 2H), 7.80 (d, J=2.78 Hz, 1H), 7.97 (d, J=8.34 Hz, 2H), 9.89 (s, 1H). ESI-MS: m/z 308.4 (M+H)+.
  • Example 132 4-((5-amino-1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00190
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.89 (s, 2H), 5.19 (s, 2H), 5.29 (s, 2H), 5.31 (d, J=2.02 Hz, 1H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.52 Hz, 1H), 6.96 (td, J=7.58, 1.52 Hz, 1H), 7.10 (d, J=2.02 Hz, 1H), 7.15 (d, J=6.82 Hz, 1H), 7.22 (d, J=8.34 Hz, 2H), 7.90 (d, J=8.08 Hz, 2H), 9.60 (s, 1H). ESI-MS: m/z 308.4 (M+H)+.
  • Example 133 4-((3-amino-1H-pyrazol-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00191
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.59 (s, 2H), 4.89 (s, 2H), 5.11 (s, 2H), 5.44 (d, J=2.27 Hz, 1H), 6.59 (td, J=7.58, 1.26 Hz, 1H), 6.77 (dd, J=8.08, 1.52 Hz, 1H), 6.96 (td, J=7.58, 1.52 Hz, 1H), 7.16 (d, J=7.83 Hz, 1H), 7.28 (d, J=8.08 Hz, 2H), 7.48 (d, J=2.02 Hz, 1H), 7.91 (d, J=8.08 Hz, 2H), 9.61 (s, 1H). ESI-MS: m/z 308.4 (M+H)+.
  • Example 134 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-methyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00192
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.70 (d, J=4.55 Hz, 3H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.52, 1.14 Hz, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.35 (d, J=8.34 Hz, 2H), 7.86 (s, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.05 (q, J=4.55 Hz, 1H), 8.25 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 350.3 (M+H)+.
  • Example 135 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-ethyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00193
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.07 (t, J=7.20 Hz, 3H), 3.16-3.24 (m, 2H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.58, 1.26 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.96 (td, J=7.71, 1.52 Hz, 1H), 7.15 (d, J=8.34 Hz, 1H), 7.35 (d, J=8.34 Hz, 2H), 7.87 (d, J=0.76 Hz, 1H), 7.95 (d, J=8.34 Hz, 2H), 8.07 (t, J=5.43 Hz, 1H), 8.25 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 364.4 (M+H)+.
  • Example 136 1-(4-(2-aminophenylcarbamoyl)benzyl)-N,N-dimethyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00194
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.97 (m, 3H), 3.14 (m, 3H), 4.90 (s, 2H), 5.44 (s, 2H), 6.59 (t, J=7.71 Hz, 1H), 6.77 (dd, J=7.83, 1.01 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=7.83 Hz, 1H), 7.37 (d, J=8.34 Hz, 2H), 7.77 (s, 1H), 7.95 (d, J=8.34 Hz, 2H), 8.31 (s, 1H), 9.63 (s, 1H). ESI-MS: m/z 364.3 (M+H)+.
  • Example 137 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-isopropyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00195
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.11 (d, J=6.82 Hz, 6H), 4.02 (dq, J=13.80, 6.77 Hz, 1H), 4.89 (s, 2H), 5.43 (s, 2H), 6.56-6.61 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.97 (td, J=7.58, 1.26 Hz, 1H), 7.15 (d, J=7.58 Hz, 1H), 7.36 (d, J=8.08 Hz, 2H), 7.83 (d, J=7.83 Hz, 1H), 7.89 (s, 1H), 7.96 (d, J=8.08 Hz, 2H), 8.26 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 378.4 (M+H)+.
  • Example 138 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-cyclopropyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00196
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 0.48 (m, 2H), 0.65 (m, 2H), 2.70-2.77 (m, J=7.31, 7.31, 3.92, 3.74, 3.74 Hz, 1H), 4.90 (s, 2H), 5.42 (s, 2H), 6.56-6.62 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.35 (d, J=8.08 Hz, 2H), 7.87 (s, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.08 (d, J=3.79 Hz, 1H), 8.25 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 376.3 (M+H)+.
  • Example 139 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(cyclopropylmethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00197
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 0.17-0.21 (m, 2H), 0.39-0.44 (m, 2H), 0.91-1.01 (m, 1H), 3.07 (t, J=6.32 Hz, 2H), 4.90 (s, 2H), 5.44 (s, 2H), 6.59 (t, J=7.45 Hz, 1H), 6.77 (dd, J=8.08, 1.01 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=7.58 Hz, 1H), 7.37 (d, J=8.08 Hz, 2H), 7.90 (s, 1H), 7.96 (d, J=8.08 Hz, 2H), 8.17 (t, J=5.68 Hz, 1H), 8.28 (s, 1H), 9.65 (s, 1H). ESI-MS: m/z 390.4 (M+H)+.
  • Example 140 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(dimethylamino)ethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00198
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 2.52-2.55 (m, 2H), 2.83 (d, J=4.80 Hz, 6H), 3.17-3.23 (m, 2H), 5.46 (s, 2H), 6.61-6.68 (m, 1H), 6.82 (d, J=7.33 Hz, 1H), 6.97-7.03 (m, 1H), 7.16 (d, J=8.59 Hz, 1H), 7.38 (d, J=7.83 Hz, 2H), 7.90 (s, 1H), 7.96 (d, J=8.34 Hz, 2H), 8.30 (s, 1H), 8.34 (t, J=6.06 Hz, 1H), 9.70 (s, 1H). ESI-MS: m/z 407.4 (M+H)+.
  • Example 141 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(dimethylamino)ethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00199
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 2.23 (s, 6H), 2.47-2.49 (m, 2H), 3.27-3.33 (m, 2H), 4.89 (s, 2H), 5.44 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.52 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=8.34 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.88 (s, 1H), 7.95 (d, J=8.34 Hz, 2H), 8.04 (t, J=5.81 Hz, 1H), 8.27 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 407.4 (M+H)+.
  • Example 142 1-(4-(2-aminophenylcarbamoyl)benzyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00200
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.90 (s, 2H), 5.43 (s, 2H), 6.56-6.62 (m, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.94-6.99 (m, 1H), 7.03 (s, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.36 (d, J=8.08 Hz, 2H), 7.58 (s, 1H), 7.88 (s, 1H), 7.96 (d, J=8.34 Hz, 2H), 8.26 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 336.3 (M+H)+.
  • Example 143 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-hydroxyethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00201
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.24 (q, J=5.98 Hz, 2H), 3.45 (q, J=6.15 Hz, 2H), 4.69-4.72 (m, 1H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.52 Hz, 1H), 6.97 (td, J=7.71, 1.52 Hz, 1H), 7.15 (d, J=7.58 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.90 (d, J=0.51 Hz, 1H), 7.95 (d, J=8.34 Hz, 2H), 8.09 (t, J=5.68 Hz, 1H), 8.28 (d, J=0.76 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 380.3 (M+H)+.
  • Example 144 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-(dimethylamino)propyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00202
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.59 (qd, J=7.07, 6.82 Hz, 2H), 2.12 (s, 6H), 2.23 (t, J=7.20 Hz, 2H), 3.16-3.22 (m, 2H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.52 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.36 (d, J=8.08 Hz, 2H), 7.87 (d, J=0.76 Hz, 1H), 7.95 (d, J=8.34 Hz, 2H), 8.10 (t, J=5.68 Hz, 1H), 8.25 (d, J=0.51 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 421.4 (M+H)+.
  • Example 145 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-3-ylmethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00203
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.42 (d, J=5.81 Hz, 2H), 4.89 (s, 2H), 5.44 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.33-7.36 (m, 1H), 7.36-7.39 (m, 2H), 7.69 (dt, J=7.83, 2.02 Hz, 1H), 7.93 (d, J=0.51 Hz, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.32 (d, J=0.51 Hz, 1H), 8.45 (dd, J=4.80, 1.77 Hz, 1H), 8.52 (d, J=1.52 Hz, 1H), 8.70 (t, J=5.94 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 427.3 (M+H)+.
  • Example 146 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(pyridin-4-ylmethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00204
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 4.43 (d, J=6.06 Hz, 2H), 4.90 (s, 2H), 5.46 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.94-6.99 (m, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.26-7.29 (m, 2H), 7.39 (d, J=8.34 Hz, 2H), 7.93-7.98 (m, 3H), 8.34 (s, 1H), 8.48-8.50 (m, 2H), 8.75 (t, J=6.06 Hz, 1H), 9.65 (s, 1H). ESI-MS: m/z 427.3 (M+H)+.
  • Example 147 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-morpholinoethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00205
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 3.07-3.19 (m, 2H), 3.26 (t, J=6.19 Hz, 2H), 3.49-3.59 (m, 4H), 3.59-3.71 (m, 2H), 4.00 (d, J=13.64 Hz, 2H), 5.46 (s, 2H), 6.79 (t, J=7.20 Hz, 1H), 6.93 (d, J=7.07 Hz, 1H), 7.05-7.10 (m, 1H), 7.22 (d, J=7.07 Hz, 1H), 7.39 (d, J=8.34 Hz, 2H), 7.91 (d, J=0.51 Hz, 1H), 7.97 (d, J=8.34 Hz, 2H), 8.32 (d, J=0.51 Hz, 1H), 8.42 (t, J=5.68 Hz, 1H), 9.85 (s, 1H). ESI-MS: m/z 449.4 (M+H)+.
  • Example 148 N-(2-aminophenyl)-4-((5-methyl-1H-pyrazol-1-yl)methyl)benzamide
  • Figure US20080114037A1-20080515-C00206
  • 1H NMR (400 MHz, DMSO-d6) of the trifluoroacetic acid salt: δ ppm 2.22 (s, 3H), 5.40 (s, 2H), 6.11 (dd, J=2.15, 1.14 Hz, 1H), 7.05 (t, J=7.58 Hz, 1H), 7.11-7.15 (m, 1H), 7.19 (d, J=7.33 Hz, 1H), 7.22 (d, J=8.08 Hz, 2H), 7.32 (d, J=7.83 Hz, 1H), 7.39 (d, J=1.77 Hz, 1H), 7.96 (d, J=8.08 Hz, 2H), 10.09 (s, 1H). ESI-MS: m/z 307.3 (M+H)+.
  • Example 149 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-propyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00207
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 0.86 (t, J=7.33 Hz, 3H), 1.43-1.52 (m, J=7.23, 7.23, 7.23, 7.23, 7.23 Hz, 2H), 3.11-3.17 (m, 2H), 4.90 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.45, 1.26 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.97 (td, J=7.71, 1.52 Hz, 1H), 7.15 (d, J=7.83 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.89 (d, J=0.51 Hz, 1H), 7.96 (d, J=8.08 Hz, 2H), 8.06 (t, J=5.68 Hz, 1H), 8.26 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 378.3 (M+H)+.
  • Example 150 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-isobutyl-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00208
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 0.86 (d, J=6.57 Hz, 6H), 1.76 (tt, J=13.52, 6.82 Hz, 1H), 2.98-3.02 (m, 2H), 4.90 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.58, 1.26 Hz, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.58 Hz, 1H), 7.37 (d, J=8.34 Hz, 2H), 7.91 (d, J=0.51 Hz, 1H), 7.96 (d, J=8.08 Hz, 2H), 8.06 (t, J=5.94 Hz, 1H), 8.27 (s, 1H), 9.64 (s, 1H). ESI-MS: m/z 392.4 (M+H)+.
  • Example 151 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-methoxyethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00209
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 3.25 (s, 3H), 3.33-3.36 (m, 2H), 3.38-3.42 (m, 2H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.97 (td, J=7.64, 1.39 Hz, 1H), 7.15 (d, J=7.07 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.90 (d, J=0.51 Hz, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.16 (t, J=5.43 Hz, 1H), 8.28 (d, J=0.51 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 394.3 (M+H)+.
  • Example 152 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-methoxypropyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00210
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.66-1.73 (m, 2H), 3.18-3.22 (m, 1H), 3.22 (s, 4H), 3.33 (t, J=3.16 Hz, 2H), 4.89 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.58, 1.26 Hz, 1H), 6.77 (dd, J=7.96, 1.39 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.33 Hz, 1H), 7.36 (d, J=8.34 Hz, 2H), 7.88 (d, J=0.76 Hz, 1H), 7.95 (d, J=8.08 Hz, 2H), 8.08 (t, J=5.68 Hz, 1H), 8.26 (d, J=0.76 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 408.4 (M+H)+.
  • Example 153 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-hydroxypropyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00211
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 1.62 (qd, J=6.74, 6.57 Hz, 2H), 3.23 (q, J=6.65 Hz, 2H), 3.40-3.46 (m, 2H), 4.46 (t, J=5.31 Hz, 1H), 4.90 (s, 2H), 5.43 (s, 2H), 6.59 (td, J=7.52, 1.39 Hz, 1H), 6.77 (dd, J=8.08, 1.26 Hz, 1H), 6.97 (td, J=7.58, 1.52 Hz, 1H), 7.15 (d, J=7.07 Hz, 1H), 7.36 (d, J=8.08 Hz, 2H), 7.89 (d, J=0.51 Hz, 1H), 7.96 (d, J=8.08 Hz, 2H), 8.07 (t, J=5.68 Hz, 1H), 8.26 (d, J=0.51 Hz, 1H), 9.64 (s, 1H). ESI-MS: m/z 394.3 (M+H)+.
  • Example 154 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-morpholinopropyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00212
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 1.83-1.91 (m, 2H), 3.01-3.10 (m, 2H), 3.10-3.17 (m, 2H), 3.26 (q, J=6.32 Hz, 2H), 3.43 (d, J=11.87 Hz, 2H), 3.64 (t, J=11.87 Hz, 2H), 3.97 (d, J=11.87 Hz, 2H), 5.46 (s, 2H), 6.93 (t, J=7.07 Hz, 1H), 7.04 (dd, J=8.08, 1.26 Hz, 1H), 7.11-7.17 (m, 1H), 7.28 (d, J=8.84 Hz, 1H), 7.39 (d, J=8.34 Hz, 2H), 7.91 (s, 1H), 7.98 (d, J=8.08 Hz, 2H), 8.29-8.35 (m, 2H), 10.00 (s, 1H). ESI-MS: m/z 463.4 (M+H)+.
  • Example 155 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(2-(piperidin-1-yl)ethyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00213
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 1.32-1.43 (m, 1H), 1.60-1.71 (m, 3H), 1.81 (d, J=14.40 Hz, 2H), 2.87-2.97 (m, 2H), 3.18 (d, J=5.31 Hz, 2H), 3.49-3.59 (m, 4H), 5.47 (s, 2H), 6.94 (t, J=6.95 Hz, 1H), 7.05-7.08 (m, 1H), 7.15 (td, J=7.64, 1.39 Hz, 1H), 7.30 (d, J=6.82 Hz, 1H), 7.40 (d, J=8.34 Hz, 2H), 7.92 (d, J=0.76 Hz, 1H), 7.99 (d, J=8.08 Hz, 2H), 10.02 (s, 1H). ESI-MS: m/z 447.4 (M+H)+.
  • Example 156 1-(4-(2-aminophenylcarbamoyl)benzyl)-N-(3-(4-methylpiperazin-1-yl)propyl)-1H-pyrazole-4-carboxamide
  • Figure US20080114037A1-20080515-C00214
  • 1H NMR (400 MHz, DMSO-d6) of the bis-trifluoroacetic acid salt: δ ppm 1.80-1.88 (m, 2H), 2.84 (s, 3H), 3.06-3.13 (m, 2H), 3.26-3.60 (m, 10H), 5.45 (s, 2H), 6.91 (t, J=6.95 Hz, 1H), 7.03 (dd, J=8.08, 1.26 Hz, 1H), 7.13 (td, J=7.58, 1.52 Hz, 1H), 7.27 (d, J=8.84 Hz, 1H), 7.39 (d, J=8.34 Hz, 2H), 7.91 (d, J=0.76 Hz, 1H), 7.98 (d, J=8.34 Hz, 2H), 8.30 (d, J=0.76 Hz, 1H), 9.98 (s, 1H). ESI-MS: m/z 476.4 (M+H)+.
  • Example 157 4-((1H-Pyrrolo[2,3-b]pyridin-1-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00215
  • 1H NMR (400 MHz, DMSO-D6) of the trifluoroacetic acid salt: δ ppm 6.03 (s, 2H) 6.67 (s, 1H) 6.83 (s, 1H) 6.91-7.02 (m, 2H) 7.12 (d, J=7.33 Hz, 1H) 7.31-7.40 (m, 2H) 7.45 (d, J=8.34 Hz, 1H) 7.58-7.70 (m, 1H) 7.87-7.99 (m, 2H) 8.65-8.80 (m, 2H) 9.76 (s, 1H). ESI-MS: m/z 343.4 (M+H)+.
  • Example 158 4-((1H-Indol-3-yl)methyl)-N-(2-aminophenyl)benzamide
  • Figure US20080114037A1-20080515-C00216
  • 1H NMR (400 MHz, DMSO-D6) δ ppm 4.10 (s, 2H) 4.84 (s, 2H) 6.53-6.60 (m, 1H) 6.74 (d, J=7.83 Hz, 1H) 6.88-6.97 (m, 2H) 7.00-7.07 (m, 2H) 7.11 (s, 1H) 7.19 (d, J=2.27 Hz, 1H) 7.32 (d, J=8.08 Hz, 1H) 7.36-7.42 (m, 2H) 7.85 (d, J=8.08 Hz, 2H) 9.54 (s, 1H) 10.87 (s, 1H). ESI-MS: m/z 342.4 (M+H)+.
  • In addition to the examples described above, the following non-limiting group of compounds can be prepared utilizing the above reaction schemes, and variations thereof, with the appropriate selection of substituents:
  • Figure US20080114037A1-20080515-C00217
    Figure US20080114037A1-20080515-C00218
    Figure US20080114037A1-20080515-C00219
    Figure US20080114037A1-20080515-C00220
    Figure US20080114037A1-20080515-C00221
    Figure US20080114037A1-20080515-C00222
    Figure US20080114037A1-20080515-C00223
  • Biological Testing
  • The activity of compounds as HDAC inhibitors may be assayed in vitro, in vivo or in a cell line. Further, compounds according to the present invention may be screened for activity against one or more HDACs. Provided below are assays for activity against HDAC1, HDAC2, HDAC6 and HDAC8.
  • Purified HDAC1, HDAC2, HDAC6, and HDAC8 may be obtained as follows.
  • For HDAC1, DNA encoding residues 1-482 of the full-length sequence of the human enzyme may be amplified by PCR and cloned into the BamHI/XbaI sites of pFastbac (Invitrogen), which incorporates a Flag tag at both the N- and C-terminus. SEQ ID NO: 1 corresponds to residues 1-482 with the N- and C-terminal Flag tag and SEQ ID NO: 2 is the DNA sequence that was used to encode SEQ ID NO: 1.
  • 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/SmaI 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.
  • For HDAC6, DNA encoding residues 73-845 of the human enzyme may be amplified by PCR and cloned into the SmaI site of pFastbac (Invitrogen), which incorporates a 6× Histidine tag at the C-terminus. SEQ ID 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.
  • For HDAC8, DNA encoding residues 1-377 corresponding to the entire sequence of the human enzyme may be amplified by PCR and cloned into the BamHI/SmaI 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 baculovirus 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 10 L Wave Bioreactors (Wave Biotech).
  • Recombinant protein may be isolated from cellular extracts by passage over ProBond resin (Invitrogen), or Anti-Flag M2 Affinity Gel (Sigma) for HDAC1. Partially purified HDAC1 may then be further purified by high pressure liquid chromatography over a Mono Q column. Partially purified extracts of HDACs other than HDAC1 and HDAC6 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 0.6 mg/mL for HDAC1, 10 mg/mL for HDAC2, 0.3 mg/mL for HDAC6, and 3 mg/mL for HDAC8. The proteins may be either stored at −78° C. in a buffer containing 25 mM TRIS-HCl pH 7.6, 150 mM NaCl, 0.1 mM EDTA and 0.25 mM TCEP or at −20° C. in the presence of glycerol (final concentration of glycerol at 50%). Alternatively, HDAC6 protein can be stored at −78° C. in a buffer containing 25 mM TRIS-HCl pH 7.2, 250 mM NaCl, and 5% glycerol.
  • It should be noted that a variety of other expression systems and hosts are also suitable for the expression of HDAC, as would be readily appreciated by one of skill in the art.
  • The inhibitory properties of compounds relative to HDAC1, 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 NaCl, 50 mM KCl, 0.1 mM EDTA, 0.01% Brij35, 0.1 mM TCEP. 50 μM 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 μL of 150 μM tBoc-Lys(Ac)AMC was added to each well of the plate, followed by the addition of 5 μL of inhibitor (2 fold serial dilutions for 11 data points for each inhibitor) containing 6% DMSO. 5 μL of either HDAC1, HDAC2, HDAC6 or HDAC8 solution may be added to initiate the reaction (final enzyme concentrations were 2.5 nM for HDAC1, 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 minutes, and quenched and developed by addition of 5 μL 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. As a reference point for this assay, suberanilohydroxamic acid (SAHA) showed an IC50 of 63 nM for HDAC 1, 69 nM for HDAC2, 108 nM for HDAC6 and 242 nM for HDAC8. IC50 values for selected compounds of the present invention are given in Table 4.
  • TABLE 4
    IC50 of SELECTED EXEMPLIFIED
    COMPOUNDS AGAINST HDAC2
    EXAMPLE IC50 (nM)
    1. ≦50
    2. 50-100
    3. ≦50
    4. ≦50
    5. ≦50
    6. ≦50
    7. ≦50
    8. ≦50
    9. 50-100
    10. ≦50
    11. ≦50
    12. ≦50
    13. ≦50
    14. ≦50
    15. ≦50
    16. ≦50
    17. ≦50
    18. ≦50
    19. 100-500 
    20. ≦50
    21. ≦50
    22. ≦50
    23. ≦50
    24. 100-500 
    25. 100-500 
    26. ≦50
    27. ≦50
    28. 50-100
    29. 50-100
    30. 50-100
    31. ≧500 
    32. ≦50
    33. ≧500 
    34. 100-500 
    35. 50-100
    36. 50-100
    37. 50-100
    38. ≦50
    39. ≧500 
    40. 100-500 
    41. 50-100
    42. ≦50
    43. ≦50
    44. 50-100
    45. 50-100
    46. 50-100
    47. 50-100
    48. 50-100
    49. ≦50
    50. 50-100
    51. 50-100
    52. 50-100
    53. 50-100
    54. 100-500 
    55. 50-100
    56. 50-100
    57. 50-100
    58. 50-100
    59. ≦50
    60. 50-100
    61. ≦50
    62. ≦50
    63. 50-100
    64. 50-100
    65. 100-500 
    66. ≦50
    67. ≦50
    68. 50-100
    69. 50-100
    70. 100-500 
    71. 50-100
    72. 50-100
    73. 100-500 
    74. ≦50
    75. 100-500 
    76. 50-100
    77. 50-100
    78. ≦50
    79. ≦50
    80. 50-100
    81. 50-100
    82. 50-100
    83. ≦50
    84. 100-500 
    85. ≦50
    86. ≦50
    87. 50-100
    88. 50-100
    89. ≦50
    90. ≦50
    91. 50-100
    92. 50-100
    93. 50-100
    94. 50-100
    95. ≦50
    96. 50-100
    97. 50-100
    98. ≦50
    99. ≦50
    100. 50-100
    101. 50-100
    102. ≧500 
    103. ≦50
    104. ≧500 
    105. 50-100
    106. ≦50
    107. 50-100
    108. 50-100
    109. 50-100
    110. 50-100
    111. ≦50
    112. 100-500 
    113. 100-500 
    114. 50-100
    115. ≦50
    116. 50-100
    117. 50-100
    118. 50-100
    119. 50-100
    120. 50-100
    121. ≦50
    122. ≧500 
    123. 100-500 
    124. 100-500 
    125. 50-100
    126. ≦50
    127. ≦50
    128. ≦50
    129. 50-100
    130. 100-500 
    131. ≧500 
    132. ≧500 
    133. 100-500 
    134. 50-100
    135. 50-100
    136. 100-500 
    137. ≦50
    138. 50-100
    139. 50-100
    140. 50-100
    141. 50-100
    142. 50-100
    143. 50-100
    144. ≦50
    145. ≦50
    146. ≦50
    147. 50-100
    148. 100-500 
    149. ≦50
    150. ≦50
    151. 50-100
    152. ≦50
    153. ≦50
    154. ≦50
    155. ≦50
    156. 50-100
    157. 100-500 
    158. 50-100
  • It will be apparent to those skilled in the art that various modifications and variations can be made in 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 (1)

1. A compound comprising:
Figure US20080114037A1-20080515-C00224
wherein:
n is selected from the group consisting of 0, 1, 2, 3 and 4;
A1 is selected from the group consisting of (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl, and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
L is a linker providing a 0-6 atom separation between the two ring atoms to which L is attached;
J1 is selected from the group consisting of —CR7R7′—and —NR19—;
J2 is selected from the group consisting of —CR2OR20′ and —NR10—;
R1 and R2 are each independently selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R1 and R2 may be taken together to form a substituted or unsubstituted ring;
R3 is selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
each R4 is selected from the group consisting of hydrogen, halo, nitro, cyano, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted;
R5, R5′, R6, R6′, R7, R7′, R20 and R20′ are each independently selected from the group consisting of hydrogen, halo, nitro, cyano, oxo, thio, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, amido, carboxamido, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or any two of R5, R5′, R6, R6′, R7, R7′, R20 and R20′ may be taken together to form a substituted or unsubstituted ring, provided that R5, R6, R7 and R20, are each independently absent when the C to which they are bound is bound to L, and R5′, R6′, R7′ and R20′, are each independently absent when the C to which they are bound form part of a double bond; and
R8, R10 and R19 are each individually selected from the group consisting of hydrogen, hydroxy, alkoxy, aryloxy, heteroaryloxy, carbonyl, amino, (C1-10)alkylamino, sulfonamido, imino, sulfonyl, sulfinyl, (C1-10)alkyl, halo(C1-10)alkyl, carbonyl(C1-3)alkyl, thiocarbonyl(C1-3)alkyl, sulfonyl(C1-3)alkyl, sulfinyl(C1-3)alkyl, amino(C1-10)alkyl, imino(C1-3)alkyl, (C3-12)cycloalkyl(C1-5)alkyl, hetero(C3-12)cycloalkyl(C1-5)alkyl, aryl(C1-10)alkyl, heteroaryl(C1-5)alkyl, (C9-12)bicycloaryl(C1-5)alkyl, hetero(C8-12)bicycloaryl(C1-5)alkyl, (C3-12)cycloalkyl, hetero(C3-12)cycloalkyl, (C9-12)bicycloalkyl, hetero(C3-12)bicycloalkyl, aryl, heteroaryl, (C9-12)bicycloaryl and hetero(C4-12)bicycloaryl, each substituted or unsubstituted, or R5 and any one of R5, R5′, R6, R6′, R7, R7′, R20, R20′, R10 and R19 may be taken together to form a substituted or unsubstituted ring, or R10 and any one of R5, R5′, R6, R6′, R7, R7′, R20, R20′, and R19, may be taken together to form a substituted or unsubstituted ring, or R19 and any one of R5, R5′, R6, R6′, R20, and R20′, may be taken together to form a substituted or unsubstituted ring, provided that R8, R10 and R19 are each independently absent when the N to which they are bound is bound to L, and R8, R10 and R19 are each independently absent when the N to which they are bound form part of a double bond.
US11/932,727 2005-07-14 2007-10-31 Histone deacetylase inhibitors Abandoned US20080114037A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/932,727 US20080114037A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US69913905P 2005-07-14 2005-07-14
US11/457,260 US7732475B2 (en) 2005-07-14 2006-07-13 Histone deacetylase inhibitors
US11/932,727 US20080114037A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/457,260 Continuation US7732475B2 (en) 2005-07-14 2006-07-13 Histone deacetylase inhibitors

Publications (1)

Publication Number Publication Date
US20080114037A1 true US20080114037A1 (en) 2008-05-15

Family

ID=37075806

Family Applications (7)

Application Number Title Priority Date Filing Date
US11/457,260 Expired - Fee Related US7732475B2 (en) 2005-07-14 2006-07-13 Histone deacetylase inhibitors
US11/933,036 Abandoned US20080119658A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,995 Abandoned US20080119648A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,860 Expired - Fee Related US7741494B2 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,308 Abandoned US20090105485A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,636 Abandoned US20080108829A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,727 Abandoned US20080114037A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US11/457,260 Expired - Fee Related US7732475B2 (en) 2005-07-14 2006-07-13 Histone deacetylase inhibitors
US11/933,036 Abandoned US20080119658A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,995 Abandoned US20080119648A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,860 Expired - Fee Related US7741494B2 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,308 Abandoned US20090105485A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors
US11/932,636 Abandoned US20080108829A1 (en) 2005-07-14 2007-10-31 Histone deacetylase inhibitors

Country Status (18)

Country Link
US (7) US7732475B2 (en)
EP (1) EP1904452A2 (en)
JP (1) JP2009501236A (en)
KR (1) KR20080032188A (en)
CN (1) CN101263121A (en)
AR (1) AR054866A1 (en)
AU (1) AU2006270322A1 (en)
BR (1) BRPI0613429A2 (en)
CA (1) CA2615105A1 (en)
CR (1) CR9664A (en)
EA (1) EA200800321A1 (en)
IL (1) IL188692A0 (en)
MA (1) MA29629B1 (en)
NO (1) NO20080784L (en)
PE (1) PE20070195A1 (en)
TW (1) TW200740761A (en)
WO (1) WO2007011626A2 (en)
ZA (1) ZA200800901B (en)

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6897220B2 (en) 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
US7868204B2 (en) 2001-09-14 2011-01-11 Methylgene Inc. Inhibitors of histone deacetylase
US7253204B2 (en) 2004-03-26 2007-08-07 Methylgene Inc. Inhibitors of histone deacetylase
US8088951B2 (en) * 2006-11-30 2012-01-03 Massachusetts Institute Of Technology Epigenetic mechanisms re-establish access to long-term memory after neuronal loss
WO2008089436A2 (en) * 2007-01-18 2008-07-24 Takeda San Diego, Inc. Crystalline forms of histone deacetylase inhibitors
EP2134683A2 (en) * 2007-02-12 2009-12-23 Intermune, Inc. Novel inhibitors hepatitis c virus replication
US8030344B2 (en) 2007-03-13 2011-10-04 Methylgene Inc. Inhibitors of histone deacetylase
CN103087043A (en) * 2007-03-16 2013-05-08 中国人民解放军军事医学科学院放射与辐射医学研究所 Benzamide derivative resisting proliferative activity and pharmaceutical preparation thereof
CA2690191C (en) * 2007-06-27 2015-07-28 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
EP2197552B1 (en) * 2007-09-19 2012-11-21 4Sc Ag Novel tetrahydrofusedpyridines as histone deacetylase inhibitors
BRPI0821247A2 (en) * 2007-12-14 2015-06-16 Univ Georgetown Histone Deacetylase Inhibitors
WO2009089277A2 (en) * 2008-01-08 2009-07-16 The Trustees Of The University Of Pennsylvania Rel inhibitors and methods of use thereof
WO2010028192A1 (en) 2008-09-03 2010-03-11 Repligen Corporation Compositions including 6-aminohexanoic acid derivatives as hdac inhibitors
AU2010313255B2 (en) 2009-10-30 2015-04-30 Massachusetts Institute Of Technology The use of CI-994 and dinaline for the treatment of memory/cognition and anxiety disorders
TWI429628B (en) 2010-03-29 2014-03-11 Univ Taipei Medical Indolyl or indolinyl hydroxamate compounds
MX2012013197A (en) 2010-05-12 2013-04-03 Abbvie Inc Indazole inhibitors of kinase.
US10059723B2 (en) 2011-02-28 2018-08-28 Biomarin Pharmaceutical Inc. Histone deacetylase inhibitors
US8957066B2 (en) 2011-02-28 2015-02-17 Biomarin Pharmaceutical Inc. Histone deacetylase inhibitors
EP2680694B1 (en) 2011-02-28 2019-01-02 BioMarin Pharmaceutical Inc. Histone deacetylase inhibitors
JP5989805B2 (en) 2012-02-10 2016-09-07 コンステレーション・ファーマシューティカルズ・インコーポレイテッドConstellation Pharmaceuticals,Inc. Methyl group-modifying enzyme regulator, composition and use thereof
JP5881460B2 (en) * 2012-02-23 2016-03-09 公立大学法人名古屋市立大学 Novel amide compounds and uses thereof
EP2844247A4 (en) * 2012-04-20 2015-11-25 Anderson Gaweco Ror modulators and their uses
EP2970305B1 (en) 2013-03-15 2017-02-22 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
BR112015023399A8 (en) 2013-03-15 2019-12-03 Biomarin Pharm Inc hdac inhibitors, their use and pharmaceutical composition
TWI636047B (en) 2013-08-14 2018-09-21 英商卡爾維斯塔製藥有限公司 Heterocyclic derivatives
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
TWI667233B (en) 2013-12-19 2019-08-01 德商拜耳製藥公司 Novel indazolecarboxamides, processes for their preparation, pharmaceutical preparations comprising them and their use for producing medicaments
CN103739550B (en) * 2014-01-02 2016-06-01 中国药科大学 2,3-dimethyl-6-urea-2H-indazole compounds and its preparation method and application
WO2015143652A1 (en) * 2014-03-26 2015-10-01 Merck Sharp & Dohme Corp. TrkA KINASE INHIBITORS,COMPOSITIONS AND METHODS THEREOF
JP6726658B2 (en) * 2014-04-11 2020-07-22 イェン、ユンYEN,Yun Histone deacetylase inhibitor
WO2015190700A1 (en) * 2014-06-11 2015-12-17 성균관대학교산학협력단 Pharmaceutical composition for preventing or treating sepsis
WO2016086060A1 (en) 2014-11-26 2016-06-02 The J. David Gladstone Institutes Methods for treating a cytomegalovirus infection
GB201421085D0 (en) * 2014-11-27 2015-01-14 Kalvista Pharmaceuticals Ltd New enzyme inhibitors
GB201421083D0 (en) 2014-11-27 2015-01-14 Kalvista Pharmaceuticals Ltd Enzyme inhibitors
WO2016161268A1 (en) 2015-04-01 2016-10-06 Enanta Pharmaceuticals, Inc. Hepatitis b antviral agents
US10738035B2 (en) 2015-05-13 2020-08-11 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
US10179131B2 (en) 2015-07-13 2019-01-15 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
US10301255B2 (en) * 2015-07-22 2019-05-28 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
WO2017015451A1 (en) * 2015-07-22 2017-01-26 Enanta Pharmaceuticals, Inc. Hepatitis b antiviral agents
KR101799007B1 (en) * 2015-07-27 2017-11-17 주식회사 종근당 1,3,4-Oxadiazole Sulfonamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
CA2993918C (en) 2015-07-27 2020-09-29 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole amide derivative compound as histone deacetylase 6 inhibitor, and pharmaceutical composition containing same
MY194018A (en) 2015-07-27 2022-11-08 Chong Kun Dang Pharmaceutical Corp 1,3,4-oxadiazole sulfamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
WO2017023133A2 (en) 2015-08-04 2017-02-09 Chong Kun Dang Pharmaceutical Corp. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
WO2017040190A1 (en) 2015-08-28 2017-03-09 Constellation Pharmaceuticals, Inc. Crystalline forms of (r)-n-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1h-indole-3-carboxamide
PL3362445T3 (en) 2015-10-12 2023-08-07 Chong Kun Dang Pharmaceutical Corp. Oxadiazole amine derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US10280175B2 (en) 2016-02-02 2019-05-07 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
PL3426245T3 (en) 2016-03-07 2023-05-22 Enanta Pharmaceuticals, Inc. Hepatitis b antiviral agents
US9728231B1 (en) * 2016-05-03 2017-08-08 Taiwan Semiconductor Manufacturing Co., Ltd. Device and method for data-writing
CN113387933A (en) 2016-05-31 2021-09-14 卡尔维斯塔制药有限公司 Pyrazole derivatives as plasma kallikrein inhibitors
GB201609603D0 (en) 2016-06-01 2016-07-13 Kalvista Pharmaceuticals Ltd Polymorphs of N-[(6-cyano-2-fluoro-3-methoxyphenyl)Methyl]-3-(methoxymethyl)-1-({4-[(2-ox opyridin-1-YL)Methyl]phenyl}methyl)pyrazole-4-carboxamide
GB201609607D0 (en) 2016-06-01 2016-07-13 Kalvista Pharmaceuticals Ltd Polymorphs of N-(3-Fluoro-4-methoxypyridin-2-yl)methyl)-3-(methoxymethyl)-1-({4-((2-oxopy ridin-1-yl)methyl)phenyl}methyl)pyrazole-4-carboxamide and salts
CA3028228A1 (en) 2016-06-10 2017-12-14 Enanta Pharmaceuticals, Inc. Hepatitis b antiviral agents
EP3529242A1 (en) 2016-10-19 2019-08-28 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of ezh2
TW201829381A (en) * 2016-12-22 2018-08-16 美商拜奧馬林製藥公司 Histone Deacetylase Inhibitors
WO2018165520A1 (en) 2017-03-10 2018-09-13 Vps-3, Inc. Metalloenzyme inhibitor compounds
CN109232358A (en) * 2017-07-10 2019-01-18 复旦大学 Indole derivatives or its salt and its preparation method and application
CN111263586B (en) 2017-08-28 2021-12-03 英安塔制药有限公司 Hepatitis B antiviral agent
GB201719881D0 (en) 2017-11-29 2018-01-10 Kalvista Pharmaceuticals Ltd Solid forms of plasma kallikrein inhibitor and salts thereof
US11234939B2 (en) 2017-11-29 2022-02-01 Kalvista Pharmaceuticals Limited Dosage forms comprising a plasma kallikrein inhibitor
TW201936192A (en) 2017-12-06 2019-09-16 美商因那塔製藥公司 Hepatitis B antiviral agents
TW201927789A (en) 2017-12-06 2019-07-16 美商因那塔製藥公司 Hepatitis B antiviral agents
US11058678B2 (en) 2018-01-22 2021-07-13 Enanta Pharmaceuticals, Inc. Substituted heterocycles as antiviral agents
US10729688B2 (en) 2018-03-29 2020-08-04 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
KR20210065965A (en) 2018-09-21 2021-06-04 이난타 파마슈티칼스, 인코포레이티드 Heterocycles functionalized as antiviral agents
WO2020106816A1 (en) 2018-11-21 2020-05-28 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
JP2020081996A (en) * 2018-11-28 2020-06-04 株式会社日本フォトサイエンス Ultraviolet treatment method and system
US20220154282A1 (en) 2019-03-12 2022-05-19 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells
WO2020247444A1 (en) 2019-06-03 2020-12-10 Enanta Pharmaceuticals, Inc, Hepatitis b antiviral agents
US11472808B2 (en) 2019-06-04 2022-10-18 Enanta Pharmaceuticals, Inc. Substituted pyrrolo[1,2-c]pyrimidines as hepatitis B antiviral agents
US11760755B2 (en) 2019-06-04 2023-09-19 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
WO2021007488A1 (en) 2019-07-11 2021-01-14 Enanta Pharmaceuticals, Inc. Substituted heterocycles as antiviral agents
CN114206852A (en) 2019-08-09 2022-03-18 卡尔维斯塔制药有限公司 Plasma kallikrein inhibitors
WO2021055425A2 (en) 2019-09-17 2021-03-25 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11802125B2 (en) 2020-03-16 2023-10-31 Enanta Pharmaceuticals, Inc. Functionalized heterocyclic compounds as antiviral agents

Citations (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US5858365A (en) * 1993-10-29 1999-01-12 Trustees Of Boston University Methods for the treatment of wounds using butyric acid salts and derivatives
US6011000A (en) * 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
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
US6043389A (en) * 1997-03-11 2000-03-28 Mor Research Applications, Ltd. Hydroxy and ether-containing oxyalkylene esters and uses thereof
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
US6197743B1 (en) * 1996-07-26 2001-03-06 The Trustees Of Boston University Compositions and methods for the treatment of viral disorders
US6231880B1 (en) * 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
US6235474B1 (en) * 1996-12-30 2001-05-22 The Johns Hopkins University Methods and kits for diagnosing and determination of the predisposition for diseases
US6335170B1 (en) * 1999-02-22 2002-01-01 Torben F. Orntoft Gene expression in bladder tumors
US20020002183A1 (en) * 2000-02-29 2002-01-03 Bing-Yan Zhu Benzamides and related inhibitors of factor Xa
US6372957B1 (en) * 1998-11-10 2002-04-16 Board Of Regents, The University Of Texas System Transgenic mouse comprising a MEF2 binding site operatively linked to an indicator gene and methods of use
US6376508B1 (en) * 2000-12-13 2002-04-23 Academia Sinica Treatments for spinal muscular atrophy
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
US20020061860A1 (en) * 2000-03-24 2002-05-23 Zuomei Li Antisense oligonucleotide inhibition of specific histone deacetylase isoforms
US20020065282A1 (en) * 2000-07-12 2002-05-30 Guy Georges Tetralone derivatives
US6506574B1 (en) * 2000-04-27 2003-01-14 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
US20030013757A1 (en) * 2001-06-15 2003-01-16 Ulrike Leser-Reiff Aromatic dicarboxylic acid derivatives
US20030013176A1 (en) * 1999-09-08 2003-01-16 Nikola Pavletich Crystal structure of a deacetylase and inhibitors thereof
US20030018062A1 (en) * 2000-09-01 2003-01-23 Remiszewski Stacy W. Deacetylase inhibitors
US20030017454A1 (en) * 2001-01-26 2003-01-23 Saraswati Sukumar Aberrantly methylated genes as markers of breast malignancy
US6512123B2 (en) * 2001-04-23 2003-01-28 Hoffmann-La Roche Inc. Tricyclic alkylhydroxamate derivatives
US6511990B1 (en) * 1999-09-08 2003-01-28 Sloan-Kettering Institute For Cancer Research Class of cytodifferentiating agents and histone deacetylase inhibitors, and methods of use thereof
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
US6538030B2 (en) * 2000-09-20 2003-03-25 Yih-Lin Chung Treating radiation fibrosis
US20030059812A1 (en) * 2001-06-14 2003-03-27 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
US6541661B1 (en) * 1999-11-23 2003-04-01 Methylgene, Inc. Inhibitors of histone deacetylase
US6544957B2 (en) * 2000-01-04 2003-04-08 The Johns Hopkins University Methods and reagents for facilitating transcription
US6548479B1 (en) * 1999-12-08 2003-04-15 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
US20030078216A1 (en) * 1999-05-03 2003-04-24 Macleod A. Robert Inhibition of histone deactylase
US20030078369A1 (en) * 1999-07-23 2003-04-24 Meinke Peter T. Apicidin-derived cyclic tetrapeptides
US20030082668A1 (en) * 1999-11-29 2003-05-01 Cyclex Co., Ltd Method for measuring the activity of deacetylase and method of screening for inhibitors and accelerators of the enzyme
US20030083521A1 (en) * 2001-03-27 2003-05-01 Circagen Pharmaceutical, A Maryland Corporation Histone deacetylase inhibitors
US20030082666A1 (en) * 2000-11-21 2003-05-01 Kammer Gary M. Method of treating autoimmune diseases
US6562995B1 (en) * 2000-12-21 2003-05-13 Beacon Laboratories, Inc. Delta dicarbonyl compounds and methods for using the same
US20040002447A1 (en) * 2002-06-04 2004-01-01 Regents Of The University Of California Induction of insulin expression
US6673587B1 (en) * 2000-08-11 2004-01-06 The Salk Institute For Biological Studies Histone deacetylase, and uses therefor
US20040005574A1 (en) * 2002-07-08 2004-01-08 Leonard Guarente SIR2 activity
US20040014647A1 (en) * 2000-12-22 2004-01-22 Lee Hyang Woo Apicidin-derivatives, their synthetic methods and anti-tumor compositions containing them
US20040018522A1 (en) * 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
US20040018968A1 (en) * 2002-04-15 2004-01-29 George Sgouros Use of histone deacetylase inhibitors in combination with radiation for the treatment of cancer
US20040023944A1 (en) * 2002-05-22 2004-02-05 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on alpha-chalcogenmethylcarbonyl compounds
US6689558B2 (en) * 2000-02-08 2004-02-10 Sangamo Biosciences, Inc. Cells for drug discovery
US20040029922A1 (en) * 2002-05-22 2004-02-12 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
US20040028607A1 (en) * 2002-05-20 2004-02-12 Verdin Eric M. Methods of modulating tubulin deacetylase activity
US20040029903A1 (en) * 2002-05-22 2004-02-12 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on trihalomethylcarbonyl compounds
US6699902B2 (en) * 2000-12-21 2004-03-02 Beacon Laboratories, Inc. Acetyloxymethyl esters and methods for using the same
US20040043470A1 (en) * 2000-10-31 2004-03-04 Yonghong Xiao Regulation of human histone deacetylase
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
US20040053960A1 (en) * 2000-12-23 2004-03-18 Guy Georges Tetrahydropyridine derivatives, their preparation and their use as cell proliferation inhibitors
US20040053820A1 (en) * 2000-07-17 2004-03-18 Hidenori Nakajima Reduced fk228 and use thereof
US20040058868A1 (en) * 2002-07-09 2004-03-25 Stephen James Methods for identification of compounds modulating insulin resistance
US20040072849A1 (en) * 2001-05-09 2004-04-15 Schreiber Stuart L. Dioxanes and uses thereof
US20040072770A1 (en) * 2002-07-03 2004-04-15 Besterman Jeffrey M. Methods for specifically inhibiting histone deacetylase-7 and 8
US20040072735A1 (en) * 2002-03-04 2004-04-15 Richon Victoria M. Methods of inducing terminal differentiation
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
US20040077698A1 (en) * 2001-01-27 2004-04-22 Guy Georges Tricyclic lactam and sultam derivatives and their use as histone deacetylase inhibitors
US20040077046A1 (en) * 2000-12-20 2004-04-22 Dalia Cohen Histone deacetylase-related gene and protein
US20040077726A1 (en) * 2000-09-29 2004-04-22 Clare Watkins Carbamic acid compounds comprising a sulfonamide linkage as hdac inhibitors
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
US20040077578A1 (en) * 2002-06-14 2004-04-22 Monia Brett P. Antisense modulation of histone deacetylase 2 expression
US20040081976A1 (en) * 2002-03-07 2004-04-29 David Sidransky Genomic screen for epigenetically silenced tumor suppressor genes
US20040087652A1 (en) * 2000-07-07 2004-05-06 Goettlicher Martin Valproic acid and derivatives thereof as histone deacetylase inhibitors
US20040087631A1 (en) * 2002-03-04 2004-05-06 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
US20040087657A1 (en) * 2001-10-16 2004-05-06 Richon Victoria M. Treatment of neurodegenerative diseases and cancer of the brain using histone deacetylase inhibitors
US20040091951A1 (en) * 2002-02-07 2004-05-13 Axys Pharmaceuticals, Inc. Assay for measuring acetylation or deacetylation activity of an enzyme
US20040091953A1 (en) * 2002-05-23 2004-05-13 Verdin Eric M. Methods of modulating mitochondrial NAD-dependent deacetylase
US20050003031A1 (en) * 1997-08-19 2005-01-06 Aylward James Harrison Anti-cancer compounds
US6841565B1 (en) * 2002-03-29 2005-01-11 The Ohio State University Treatment of patients with chronic lymphocytic leukemia
US20050009030A1 (en) * 2002-03-26 2005-01-13 Fabien Schweighoffer Histone deacetylase: novel molecular target of neurotoxicity
US20050020557A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors
US20050026907A1 (en) * 2003-06-10 2005-02-03 Kalypsys, Inc. Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
US20050032794A1 (en) * 2003-08-05 2005-02-10 Padia Janak K. Diamine derivatives of quinone and uses thereof
US20050032899A1 (en) * 2001-11-06 2005-02-10 Chen Ying-Nan Pan Cyclooxygenase-2 inhibitor/histone deacetylase inhibitor combination
US20050032831A1 (en) * 2003-07-07 2005-02-10 Kozikowski Alan P. Histone deacetylase inhibitors and methods of use thereof
US20050038113A1 (en) * 2001-09-18 2005-02-17 G2M Cancer Drugs Ag Valproic acid and derivatives for the combinatorial therapeutic treatment of human cancers and for the treatment of tumor metastasis and minimal residual disease
US20050037992A1 (en) * 2003-07-22 2005-02-17 John Lyons Composition and method for treating neurological disorders
US20050059682A1 (en) * 2003-09-12 2005-03-17 Supergen, Inc., A Delaware Corporation Compositions and methods for treatment of cancer
US6869953B2 (en) * 2001-08-07 2005-03-22 Hoffman-La Roche Inc. N-monoacylated derivatives of o-phenylenediamines, their analogs and their use as pharmaceutical agents
US20050065596A1 (en) * 2002-07-24 2005-03-24 Xufan Tseng Stents capable of controllably releasing histone deacetylase inhibitors
US20050070467A1 (en) * 2003-09-25 2005-03-31 Fujisawa Pharmaceutical Co., Ltd. Antitumor agent
US6875598B1 (en) * 1999-12-08 2005-04-05 Applera Corporation Histone deacetylase-8 proteins, nuclei acids, and methods for use
US20050079995A1 (en) * 2001-11-27 2005-04-14 Antonio Bedaloy Methods for inhibiting deacetylase activity
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
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

Family Cites Families (233)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US794392A (en) * 1904-01-29 1905-07-11 Nat Tube Co Bending-roll.
US4482571A (en) 1982-06-21 1984-11-13 University Of Pittsburgh Sickle cell anemia treatment and compound
GB8524157D0 (en) 1984-10-19 1985-11-06 Ici America Inc Heterocyclic amides
GB8607294D0 (en) 1985-04-17 1986-04-30 Ici America Inc Heterocyclic amide derivatives
IE862737L (en) 1985-10-17 1987-04-17 Staehler Theo Heterocyclic compounds
US4964895A (en) 1987-06-08 1990-10-23 Monsanto Company Substituted 4-(4-nitrophenoxy) pyrazoles and their use as herbicides
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
WO1995013062A1 (en) 1993-11-10 1995-05-18 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
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
US5789434A (en) 1994-11-15 1998-08-04 Bayer Corporation Derivatives of substituted 4-biarylbutyric acid as matrix metalloprotease inhibitors
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
US6110697A (en) 1995-09-20 2000-08-29 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents
US5922837A (en) 1995-09-20 1999-07-13 Merck & Co., Inc. Antiprotozoal cyclic tetrapeptides
JPH11514857A (en) 1995-09-20 1999-12-21 メルク エンド カンパニー インコーポレーテッド Histone deacetylase as a target for antiprotozoal agents
US20030219832A1 (en) 1996-03-11 2003-11-27 Klein Elliott S. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US6777217B1 (en) 1996-03-26 2004-08-17 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
WO1997047307A1 (en) 1996-06-14 1997-12-18 The Uab Research Foundation Use of histone deacetylase inhibitors to activate transgene expression
EP0927033A1 (en) 1996-07-02 1999-07-07 Bar-Ilan University Retinoyloxy(substituted)alkylene butyrates useful for the treatment of cancer and other proliferative diseases
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
US6794392B1 (en) 1996-09-30 2004-09-21 Schering Aktiengesellschaft Cell differentiation inducer
ES2331860T3 (en) 1996-12-23 2010-01-18 Bristol-Myers Squibb Pharma Company HETEROAROMATIC COMPOUNDS CONTAINING NITROGEN AS INHIBITORS OF FACTOR XA.
US6130248A (en) 1996-12-30 2000-10-10 Bar-Ilan University Tricarboxylic acid-containing oxyalkyl esters and uses thereof
AU5617398A (en) 1996-12-30 1998-07-31 Beacon Laboratories L.L.C. Tricarboxylic acid-containing oxyalkyl esters and uses thereof
US7062219B2 (en) 1997-01-31 2006-06-13 Odyssey Thera Inc. Protein fragment complementation assays for high-throughput and high-content screening
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
US5939455A (en) 1997-03-11 1999-08-17 Beacon Laboratories, Inc. Therapeutic augmentation of oxyalkylene diesters and butyric acid derivatives
US6110955A (en) 1997-03-11 2000-08-29 Beacon Laboratories, Inc. Metabolically stabilized oxyalkylene esters and uses thereof
AUPO721997A0 (en) 1997-06-06 1997-07-03 Queensland Institute Of Medical Research, The Anticancer compounds
JP3494624B2 (en) 1997-09-02 2004-02-09 住友製薬株式会社 Novel cyclic tetrapeptide derivatives and their pharmaceutical uses
WO1999023885A1 (en) 1997-11-10 1999-05-20 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
US6262116B1 (en) 1998-01-23 2001-07-17 Sloan-Kettering Institute For Cancer Research Transcription therapy for cancers
WO1999040883A2 (en) 1998-02-11 1999-08-19 Faller Douglas V 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
US6288063B1 (en) 1998-05-27 2001-09-11 Bayer Corporation Substituted 4-biarylbutyric and 5-biarylpentanoic acid derivatives as matrix metalloprotease inhibitors
AUPP505798A0 (en) 1998-08-04 1998-08-27 Fujisawa Pharmaceutical Co., Ltd. Novel compound fr225497 substance
EP1105143A4 (en) 1998-08-21 2002-07-17 Smithkline Beecham Corp Human histone deacetylase gene hd4
DE19842506A1 (en) 1998-09-17 2000-03-23 Volkswagen Ag Fault and signal plausibility monitoring method for data processing network uses monitoring processor associated with each electronic main processor for handling same signals and data
IL142531A0 (en) 1998-10-13 2002-03-10 Fujisawa Pharmaceutical Co Cyclic peptide compound, processes for the preparation thereof and pharmaceutical compositions containing the same
AU6339399A (en) 1998-10-16 2000-05-08 Novartis Ag Promotion of self-renewal and improved gene transduction of hematopoietic stem cells by histone deacetylase inhibitors
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
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
ATE329058T1 (en) 1999-03-23 2006-06-15 Chugai Pharmaceutical Co Ltd METHOD FOR SCREENING FOR ANTI-CANCER DRUGS
ES2187473T3 (en) 1999-04-09 2003-06-16 Smithkline Beecham Corp TRIARYLIMIDAZOLES.
JP2003500052A (en) 1999-05-03 2003-01-07 メチルジーン インコーポレイテッド Inhibition of histone deacetylase
WO2001007042A1 (en) 1999-07-23 2001-02-01 Merck & Co., Inc. Apicidin-derived cyclic tetrapeptides
CA2382045A1 (en) 1999-08-20 2001-03-01 Board Of Regents, The University Of Texas System Hdac4 and hdac5 in the 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
AU7108400A (en) 1999-09-03 2001-04-10 Salk Institute For Biological Studies, The Modulation of gene expression by modulating histone acetylation
WO2001027314A1 (en) 1999-10-08 2001-04-19 Merck & Co., Inc. Antiprotozoal histone acetyl transferase inhibitors
CA2291367A1 (en) 1999-12-06 2001-06-06 Isabelle Henry Genetic constructions having a reduced or an increased number of epigenetic control regions and methods of use thereof
JP2003516144A (en) 1999-12-08 2003-05-13 アクシス・ファーマシューティカルズ・インコーポレイテッド Histone deacetylase-8 proteins, nucleic acids and methods of using the same
US6828302B1 (en) 1999-12-08 2004-12-07 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
US7452664B2 (en) 1999-12-15 2008-11-18 Massachusetts Institute Of Technology Methods for identifying agents which alter histone protein acetylation
US20030129724A1 (en) 2000-03-03 2003-07-10 Grozinger Christina M. Class II human histone deacetylases, and uses related thereto
GB0005199D0 (en) 2000-03-04 2000-04-26 Imp College Innovations Ltd Modulation of histone deacetylase
US7288567B2 (en) 2000-03-24 2007-10-30 Methylgene Inc. Inhibitors of histone deacetylase
GB0007217D0 (en) 2000-03-24 2000-05-17 Europ I Of Oncology Materials and methods relating to the treatment of leukaemias
EP1268534A2 (en) 2000-03-24 2003-01-02 Filgen Biosciences, Inc. Peptides binding to non-acetylated h3 and h4 histones for cancer therapy
US20020137162A1 (en) 2000-03-24 2002-09-26 Zuomei Li Antisense oligonucleotide inhibition of specific histone deacetylase isoforms
GB0007405D0 (en) 2000-03-27 2000-05-17 Smithkline Beecham Corp Compounds
AU2001277105A1 (en) 2000-07-21 2002-02-05 Millennium Pharmaceuticals, Inc. 47508, a novel human histone deacetylase family member and uses thereof
AU2001275794A1 (en) 2000-07-28 2002-02-13 Sumitomo Pharmaceuticals Co. Ltd. Pyrrole derivatives
EP1182263A1 (en) 2000-08-11 2002-02-27 Evotec OAI AG Process for detecting threonine/serine kinase activity
WO2002015921A2 (en) 2000-08-18 2002-02-28 The Governement Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Methods of treating cutaneous and peripheral t-cell lymphoma by a histone deacetylase inhibitor
WO2002022133A1 (en) 2000-09-12 2002-03-21 Virginia Commonwealth University Promotion of adoptosis in cancer cells by co-administration of cyclin dependent kinase inhibitors and cellular differentiation agents
GB0023983D0 (en) 2000-09-29 2000-11-15 Prolifix Ltd Therapeutic compounds
ES2257441T3 (en) 2000-09-29 2006-08-01 Topotarget Uk Limited CARBON ACID COMPOUNDS THAT INCLUDE AN AMINO LINK AS HDAC INHIBITORS.
WO2002030970A2 (en) 2000-10-13 2002-04-18 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
JP2005519848A (en) 2000-11-02 2005-07-07 スローン−ケッタリング・インスティテュート・フォー・キャンサー・リサーチ Small molecule composition for binding to HSP90
DE10061162A1 (en) 2000-11-30 2002-07-11 Schering Ag Aryl-substituted indirubin derivatives, their preparation and use
US20030154032A1 (en) 2000-12-15 2003-08-14 Pittman Debra D. Methods and compositions for diagnosing and treating rheumatoid arthritis
US20040204373A1 (en) 2000-12-19 2004-10-14 Isis Pharmaceuticals Inc. Antisense modulation of histone deacetylase I expression
US20050171042A1 (en) 2000-12-19 2005-08-04 Monia Brett P. Compositions and their uses directed to binding proteins
US6693132B2 (en) 2000-12-21 2004-02-17 Beacon Laboratories, Inc. Methods for using alkanoyloxymethyl esters
WO2002055688A2 (en) 2001-01-10 2002-07-18 Us Gov Health & Human Serv Histone deacetylase inhibitors in diagnosis and treatment of thyroid neoplasms
CA2434601A1 (en) 2001-01-12 2002-09-12 Methylgene, Inc. Methods for specifically inhibiting histone deacetylase-4
US20030148970A1 (en) 2001-01-12 2003-08-07 Besterman Jeffrey M. Methods for specifically inhibiting histone deacetylase-4
US20030152557A1 (en) 2001-01-12 2003-08-14 Besterman Jeffrey M. Methods for inhibiting histone deacetylase-4
WO2002060430A1 (en) 2001-02-01 2002-08-08 Cornell Research Foundation, Inc. Use of retinoids plus histone deacetylase inhibitors to inhibit the growth of solid tumors
AU2002233314A1 (en) 2001-02-05 2002-09-19 Bayer Aktiengesellschaft Regulation of human histone acetyltransferase
US20020177594A1 (en) 2001-03-14 2002-11-28 Curtin Michael L. Inhibitors of histone deacetylase
US8026280B2 (en) 2001-03-27 2011-09-27 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors
US7312247B2 (en) 2001-03-27 2007-12-25 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors
EP1408946A4 (en) 2001-03-27 2004-06-09 Circagen Pharmaceutical Histone deacetylase inhibitors
ATE305297T1 (en) 2001-04-10 2005-10-15 Deutsches Krebsforsch USE OF HISTONEDEACETYLASE INHIBITORS FOR THE TREATMENT OF PAPILLOMAVIRUS-ASSOCIATED DISEASES
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
WO2002090534A1 (en) 2001-05-02 2002-11-14 The Regents Of The University Of California Method for treating neurodegenerative, psychiatric and other disorders with deacetylase inhibitors
AU2002305523A1 (en) 2001-05-09 2002-11-18 President And Fellows Of Harvard College Dioxanes and uses thereof
AU2002318364A1 (en) 2001-06-14 2003-01-02 Bristol-Myers Squibb Company Novel human histone deacetylases
AU2002315166A1 (en) 2001-06-15 2003-01-02 The Trustees Of Columbia University In The City Of New York Sir2alpha-based therapeutic and prophylactic methods
WO2002102316A2 (en) 2001-06-19 2002-12-27 The Regents Of The University Of California Histone deacetylase and methods of use thereof
CA2451654A1 (en) 2001-06-22 2003-01-03 Ceres, Inc. Chimeric histone acetyltransferase polypeptides
US20040186274A1 (en) 2001-07-03 2004-09-23 Allis C. David Methylation of histone h4 at arginine 3
AU2002333315A1 (en) 2001-08-03 2003-02-24 Novartis Ag Human histone deacetylase-related gene and protein hdac10
US20040266718A1 (en) 2001-08-06 2004-12-30 Zuomei Li Inhibition of specific histone deacetylase isoforms
ITMI20011733A1 (en) 2001-08-07 2003-02-07 Italfarmaco Spa HYDROXAMIC ACID DERIVATIVES HISTONE DEHYACETYLASE ENZYM INHIBITORS, WHICH NEW ANTI-INFLAMMATORY DRUGS INHIBIT CITOC SYNTHESIS
JP4238728B2 (en) 2001-08-21 2009-03-18 アステラス製薬株式会社 Medicinal uses of histone deacetylase inhibitors and methods for evaluating their antitumor effects
US20040259772A1 (en) 2001-08-24 2004-12-23 Antonio Fojo Use of a histone deacetylase inhibitor to increase the entry of an adenoviral agent into a cell
EP1291015A1 (en) 2001-09-10 2003-03-12 Lunamed AG Dosage forms having prolonged active ingredient release
US7595343B2 (en) 2001-09-14 2009-09-29 Methylgene, Inc. Inhibitors of histone deacetylase
US7868204B2 (en) 2001-09-14 2011-01-11 Methylgene Inc. Inhibitors of histone deacetylase
US6897220B2 (en) 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
DE10148618B4 (en) 2001-09-25 2007-05-03 Schering Ag Substituted N- (1,4,5,6-tetrahydro-cyclopentapyrazol-3-yl) derivatives, their preparation and use as medicaments
GB0123664D0 (en) 2001-10-02 2001-11-21 Inst Of Cancer Res The Histone deacetylase 9
AU2002348474A1 (en) 2001-10-18 2003-04-28 The Salk Institute For Biological Studies Methods of using deacetylase inhibitors to promote cell differentiation and regeneration
CN1310937C (en) 2001-11-21 2007-04-18 犹太大学阿尔伯特爱因斯坦医学院 Sir2 products and activities
AU2002361022A1 (en) 2001-12-07 2003-06-17 Novartis Ag Use of alpha-tubulin acetylation levels as a biomarker for protein deacetylase inhibitors
AU2002351898A1 (en) 2001-12-21 2003-07-09 Universite Libre De Bruxelles Method for obtaining the elimination of integrated and functional viruses from infected mammal cells
WO2003057722A2 (en) 2001-12-28 2003-07-17 Fujisawa Pharmaceutical Co., Ltd. Cyclic tetrapeptide compound and use thereof
ITRM20020016A1 (en) 2002-01-15 2003-07-15 Sigma Tau Ind Farmaceuti FENYL ACID DERIVATIVES (ALCHYL) CARBOXYL AND DYNIC PHENYLALKYL THEROCYCLIC DERIVATIVES, THEIR USE AS MEDICATIONS WITH HYPOGLYCEMIC ACTIVITY
WO2003064698A1 (en) 2002-01-25 2003-08-07 Board Of Regents, The University Of Texas System Electrophoretic assay to predict risk of cancer and the efficacy and toxicity of cancer therapy
US20050118596A1 (en) 2002-02-08 2005-06-02 Asselbergs Fredericus Alphonsus M. Method for screening for compounds having hdac inhibitory activity
CA2476434A1 (en) 2002-02-15 2003-08-28 Sloan-Kettering Institute For Cancer Research Method of treating trx mediated diseases
US7488712B2 (en) 2002-02-20 2009-02-10 Kyushu Institute Of Technology Histone deacetylase inhibitors and methods for producing the same
AU2003211362A1 (en) 2002-02-21 2003-09-09 Osaka Industrial Promotion Organization N-hydroxycarboxamide derivative
US7456219B2 (en) 2002-03-04 2008-11-25 Merck Hdac Research, Llc Polymorphs of suberoylanilide hydroxamic acid
US20040132825A1 (en) 2002-03-04 2004-07-08 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
WO2003076593A2 (en) 2002-03-07 2003-09-18 The Johns Hopkins University School Of Medicine Genomic screen for epigenetically silenced genes associated with cancer
CA2476583C (en) 2002-03-13 2011-07-05 Kristof Van Emelen Aminocarbonyl-derivatives as novel inhibitors of histone deacetylase
MXPA04008795A (en) 2002-03-13 2004-11-26 Janssen Pharmaceutica Nv Piperazinyl-, piperidinyl- and morpholinyl-derivatives as novel inhibitors of histone deacetylase.
DE60321324D1 (en) 2002-03-13 2008-07-10 Janssen Pharmaceutica Nv SULFONYLAMINODERIVATES AS NEW INHIBITORS OF HISTONDEACETYLASE
CN100445276C (en) 2002-03-13 2008-12-24 詹森药业有限公司 Sulfonylamino-derivatives as novel inhibitors of histone deacetylase
KR20040090981A (en) 2002-03-13 2004-10-27 얀센 파마슈티카 엔.브이. Carbonylamino-derivatives as novel inhibitors of histone deacetylase
BR0308908A (en) 2002-04-03 2005-01-04 Topotarget Uk Ltd Compound, composition, use of a compound, and methods for inhibiting hdac in a cell for the treatment of an hdac-mediated condition, a proliferative condition, cancer, and psoriasis
TWI319387B (en) 2002-04-05 2010-01-11 Astrazeneca Ab Benzamide derivatives
US8673888B2 (en) 2002-04-05 2014-03-18 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Depsipeptide for therapy of kidney cancer
WO2003087066A1 (en) 2002-04-11 2003-10-23 Sk Chemicals, Co., Ltd. α,β-UNSATURATED HYDROXAMIC ACID DERIVATIVES AND THEIR USE AS HISTONE DEACETYLASE INHIBITORS
MY134200A (en) 2002-04-12 2007-11-30 Kowa Co Method for treating cancer
US6620838B1 (en) 2002-04-19 2003-09-16 Signal Pharmaceuticals, Inc. Benzopyrazone compounds, compositions thereof, and methods of treatment therewith
US20030206946A1 (en) 2002-04-26 2003-11-06 Yih-Lin Chung Methods for therapy of connective tissue disease
US6809118B2 (en) 2002-07-25 2004-10-26 Yih-Lin Chung Methods for therapy of radiation cutaneous syndrome
GB0209715D0 (en) 2002-04-27 2002-06-05 Astrazeneca Ab Chemical compounds
NO20032183L (en) 2002-05-15 2003-11-17 Schering Ag Histone deacetylase inhibitor and its use
US20030224473A1 (en) 2002-05-23 2003-12-04 Mccafferty Dewey G. Methods for screening for histone deacetylase activity and for identifying histone deacetylase inhibitors
EP1515750B1 (en) 2002-06-10 2010-08-11 Novartis AG Combinations comprising epothilones and pharmaceutical uses thereof
AU2002349786A1 (en) 2002-06-19 2004-01-06 Japan As Represented By The President Of The University Of Tokyo Method for diagnosis of colorectal tumors
US20040142993A1 (en) 2002-07-01 2004-07-22 Carlo Battistini Inhibitors of HCV NS5B polymerase
GB0215844D0 (en) 2002-07-09 2002-08-14 Novartis Ag Organic compounds
AU2003253872A1 (en) 2002-07-17 2004-02-09 Chemgenex Pharmaceuticals, Inc. Formulations and methods of administration of cephalotaxines, including homoharringtonine
KR20050040907A (en) 2002-07-17 2005-05-03 타이탄 파머슈티컬즈 인코퍼레이티드 Combination of chemotherapeutic drugs for increasing antitumor activity
DE10233412A1 (en) 2002-07-23 2004-02-12 4Sc Ag New compounds as histone deacetylase inhibitors
US20050176686A1 (en) 2002-07-23 2005-08-11 4Sc Ag Novel compounds as histone deacetylase inhibitors
US20080145928A1 (en) 2002-07-25 2008-06-19 Shuki Mizutani Factor Taking Part in Transcription Control
BR0313371A (en) 2002-08-02 2005-07-05 Argenta Discovery Ltd Compound, use of a compound and disease treatment method
ATE465757T1 (en) 2002-08-20 2010-05-15 Astellas Pharma Inc INHIBITOR OF THE DEGRADATION OF THE EXTRACELLULAR MATRIX OF ARTHRODIA CARTILAGE
WO2004020460A1 (en) 2002-08-30 2004-03-11 Yamanouchi Pharmaceutical Co., Ltd. Novel depsipeptide compound
EP1553948B1 (en) 2002-09-13 2011-08-24 Virginia Commonwealth University Combination of imatnib and a histone deacetylase inhibitor for the treatment of leukemia
EP1400806A1 (en) 2002-09-18 2004-03-24 G2M Cancer Drugs AG The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
CA2499189A1 (en) 2002-09-19 2004-04-01 University Of South Florida Method of treating leukemia with a combination of suberoylanilide hydromaxic acid and imatinib mesylate
EP1403639A1 (en) 2002-09-30 2004-03-31 G2M Cancer Drugs AG Antibody tools for the diagnostic use in the medical therapy with inhibitors of histone deacetylases
WO2004035525A1 (en) 2002-10-17 2004-04-29 Methylgene Inc. Inhibitors of histone deacetylase
CA2504226A1 (en) 2002-11-12 2004-05-27 Alcon, Inc. Histone deacetylase inhibitors for treating degenerative diseases of the eye
BR0316206A (en) 2002-11-12 2005-09-27 Alcon Inc Use of histone deacetylase inhibitors for the treatment of neovascular or edematous eye disorders
AU2003295444A1 (en) 2002-11-15 2004-06-15 The Board Of Trustees Of The University Of Illinois Methods for in vitro expansion of hematopoietic stem cells
GB0226855D0 (en) 2002-11-18 2002-12-24 Queen Mary & Westfield College Histone deacetylase inhibitors
WO2004046104A2 (en) 2002-11-20 2004-06-03 Errant Gene Therapeutics, Llc Treatment of lung cells with histone deacetylase inhibitors
AU2003296310A1 (en) 2002-12-06 2004-06-30 University Of South Florida Histone deacetylase inhibitor enhancement of trail-induced apoptosis
TW200426138A (en) 2002-12-10 2004-12-01 Hoffmann La Roche Novel arylene-carboxylic acid (2-amino-phenyl)-amide derivatives, their manufacture and use as pharmaceutical agents
EP1590318A2 (en) 2002-12-12 2005-11-02 Jawaharlal Nehru Centre For Advanced Scientific Research Modulators (inhibitors/activators) of histone acetyltransferases
US7098241B2 (en) 2002-12-16 2006-08-29 Hoffmann-La Roche Inc. Thiophene hydroxamic acid derivatives
ITMI20022745A1 (en) 2002-12-23 2004-06-24 Coimex Scrl United Companies MIXED ESTERS OF HYALURONIC ACID FOR CYTOSTATIC AND MANUFACTURING ACTIVITIES AND PROCEDURE FOR THEIR PRODUCTION.
AU2003292888A1 (en) 2002-12-27 2004-07-22 Schering Aktiengesellschaft Pharmaceutical combinations of phthalazine vegf inhibitors and benzamide hdac inhibitors
US20040197888A1 (en) 2002-12-31 2004-10-07 Armour Christopher D. Alternatively spliced isoforms of histone deacetylase 3 (HDAC3)
ITMI20030025A1 (en) 2003-01-10 2004-07-11 Italfarmaco Spa HYDROXAMIC ACID DERIVATIVES FOR ANTI-INFLAMMATORY ACTIVITY.
TW200418806A (en) 2003-01-13 2004-10-01 Fujisawa Pharmaceutical Co HDAC inhibitor
US20050222013A1 (en) 2003-01-16 2005-10-06 Georgetown University Methods for the use of inhibitors of histone deacetylase as synergistic agents in cancer therapy
EP1583736A1 (en) 2003-01-17 2005-10-12 TopoTarget UK Limited Carbamic acid compounds comprising an ester or ketone linkage as hdac inhibitors
WO2004067480A2 (en) 2003-01-25 2004-08-12 Oxford Glycosciences (Uk) Ltd Substituted phenylurea derivatives as hdac inhibitors
TW200424174A (en) 2003-02-06 2004-11-16 Hoffmann La Roche New TP diamide
US7208491B2 (en) * 2003-02-07 2007-04-24 Hoffmann-La Roche Inc. N-monoacylated o-phenylenediamines
AU2003900608A0 (en) 2003-02-11 2003-02-27 Fujisawa Pharmaceutical Co., Ltd. Hdac inhibitor
AU2003900587A0 (en) 2003-02-11 2003-02-27 Fujisawa Pharmaceutical Co., Ltd. Hdac inhibitor
EP1592811A2 (en) 2003-02-11 2005-11-09 Wyeth Methods for monitoring drug activities in vivo
WO2004071443A2 (en) 2003-02-12 2004-08-26 Irm Llc Methods and compositions for modulating stem cells
WO2004071464A2 (en) 2003-02-12 2004-08-26 Johns Hopkins University School Of Medicine Diagnostic application of differentially-expressed genes in lympho-hematopoietic stem cells
US7244751B2 (en) 2003-02-14 2007-07-17 Shenzhen Chipscreen Biosciences Ltd. Histone deacetylase inhibitors of novel benzamide derivatives with potent differentiation and anti-proliferation activity
CN1777675A (en) 2003-02-19 2006-05-24 安斯泰来制药有限公司 Method of estimating antitumor effect of histone deacetylase inhibitor.
AU2004215624B2 (en) 2003-02-25 2011-06-02 Topotarget Uk Limited Hydroxamic acid compounds comprising a bicyclic heteroaryl group as HDAC inhibitors
JP2006520796A (en) 2003-03-17 2006-09-14 タケダ サン ディエゴ インコーポレイテッド Histone deacetylase inhibitor
AU2004228011A1 (en) 2003-04-01 2004-10-21 Memorial Sloan-Kettering Cancer Center Hydroxamic acid compounds and methods of use thereof
TW200424187A (en) 2003-04-04 2004-11-16 Hoffmann La Roche New oxime derivatives and their use as pharmaceutically active agents
NZ542445A (en) 2003-04-07 2008-03-28 Pharmacyclics Inc Hydroxamates as therapeutic agents
US20040213826A1 (en) 2003-04-28 2004-10-28 Marx Steven O. Medical devices and methods for inhibiting proliferation of smooth muscle cells
CN1812776A (en) 2003-05-21 2006-08-02 得克萨斯州大学系统董事会 Inhibition of protein kinase c-mu (pkd) as a treatment for cardiac hypertrophy and heart failure
CN1791396A (en) 2003-05-21 2006-06-21 诺瓦提斯公司 Combination of histone deacetylase inhibitors with chemotherapeutic agents
PE20050206A1 (en) 2003-05-26 2005-03-26 Schering Ag PHARMACEUTICAL COMPOSITION CONTAINING AN INHIBITOR OF HISTONE DEACETILASE
AU2004247136A1 (en) 2003-06-10 2004-12-23 Kalypsys, Inc. Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
EP1633751A1 (en) 2003-06-16 2006-03-15 Chroma Therapeutics Limited Carboline and betacarboline derivatives for use as hdac enzyme inhibitors
EP1491188A1 (en) 2003-06-25 2004-12-29 G2M Cancer Drugs AG Topical use of valproic acid for the prevention or treatment of skin disorders
DK2238982T3 (en) 2003-06-27 2013-01-28 Astellas Pharma Inc Therapeutic agent for soft tissue sarcoma
AU2004253579B2 (en) 2003-07-01 2010-12-23 Biomol International L.P. Sirt1 modulators for manipulating cells/organism lifespan/stress response
WO2005016342A1 (en) 2003-07-02 2005-02-24 Albert Einstein College Of Medicine Of Yeshiva University Sir2 regulation
EP1644323B1 (en) 2003-07-07 2015-03-18 Georgetown University Histone deacetylase inhibitors and methods of use thereof
WO2005004861A1 (en) 2003-07-15 2005-01-20 Korea Research Institute Of Bioscience And Biotechnology A use of novel 2-oxo-heterocyclic compounds and the pharmaceutical compositions comprising the same
GB0316889D0 (en) 2003-07-18 2003-08-20 Univ London Pharmaceutical agents
WO2005011598A2 (en) 2003-07-31 2005-02-10 University Of South Florida Leukemia treatment method and composition
WO2005014588A1 (en) 2003-08-01 2005-02-17 Argenta Discovery Limited Substituted thienyl-hydroxamic acids having histone deacetylase activity
WO2005013958A1 (en) 2003-08-07 2005-02-17 Novartis Ag Histone deacetylase inhibitors as immunosuppressants
JP2007501774A (en) 2003-08-08 2007-02-01 ノバルティス アクチエンゲゼルシャフト Combinations containing staurosporine
EP1656348B1 (en) 2003-08-20 2007-02-07 Axys Pharmaceuticals, Inc. Acetylene derivatives as inhibitors of histone deacetylase
JP4338734B2 (en) 2003-08-26 2009-10-07 メルク エイチディーエーシー リサーチ エルエルシー Cancer treatment with HDAC inhibitors
EP1667680A4 (en) 2003-08-29 2008-10-08 Aton Pharma Inc Combination methods of treating cancer
US8404458B2 (en) 2003-09-16 2013-03-26 The Rockefeller University Histone modifications as binary switches controlling gene expression
BRPI0414506A (en) 2003-09-18 2006-11-07 Novartis Ag combination of a histone deacetylase inhibitor with a death receptor ligand
WO2005028447A1 (en) 2003-09-22 2005-03-31 S*Bio Pte Ltd Benzimidazole derivates: preparation and pharmaceutical applications
WO2005030705A1 (en) 2003-09-24 2005-04-07 Methylgene, Inc. Inhibitors of histone deacetylase
JP2007508318A (en) 2003-10-09 2007-04-05 エートン ファーマ インコーポレーティッド Thiophene and benzothiophene hydroxamic acid derivatives
US20050124625A1 (en) 2003-10-21 2005-06-09 Salvati Mark E. Piperazine derivatives and their use as modulators of nuclear hormone receptor function
WO2005040161A1 (en) 2003-10-27 2005-05-06 S*Bio Pte Ltd Biaryl linked hydroxamates: preparation and pharmaceutical applications
MXPA06004735A (en) 2003-10-27 2006-12-14 S Bio Pte Ltd Acylurea connected and sulfonylurea connected hydroxamates.
IN2003CH00929A (en) 2003-11-13 2008-10-06
WO2005053610A2 (en) 2003-11-26 2005-06-16 Aton Pharma, Inc. Diamine and iminodiacetic acid hydroxamic acid derivatives
WO2005053609A2 (en) 2003-11-26 2005-06-16 Guilford Pharmaceuticals Inc. Methods of nad+-dependent deacetylase inhibitors
EP1694640A4 (en) 2003-11-28 2007-01-10 Univ Queensland Anti-cancer agents
EP1696898B1 (en) 2003-12-02 2015-11-18 The Ohio State University Research Foundation Zn 2+ -chelating motif-tethered short-chain fatty acids as a novel class of histone deacetylase inhibitors
EP1713460A2 (en) 2003-12-10 2006-10-25 Wisconsin Alumni Research Foundation Fk228 analogs and their use as hdac-inhibitors
EP1541549A1 (en) 2003-12-12 2005-06-15 Exonhit Therapeutics S.A. Tricyclic hydroxamate and benzaminde derivatives, compositions and methods
US8652502B2 (en) 2003-12-19 2014-02-18 Cordis Corporation Local vascular delivery of trichostatin A alone or in combination with sirolimus to prevent restenosis following vascular injury
WO2005066151A2 (en) 2003-12-19 2005-07-21 Takeda San Diego, Inc. Histone deacetylase inhibitors
US20050159470A1 (en) 2003-12-19 2005-07-21 Syrrx, Inc. Histone deacetylase inhibitors
CA2548671C (en) 2003-12-29 2015-02-24 President And Fellows Of Harvard College Compositions for treating or preventing obesity and insulin resistance disorders
WO2005071079A1 (en) 2004-01-21 2005-08-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Inhibitors of mammalian hdac 11 useful for treating hdac 11 mediated disorders
CA2559733C (en) 2004-03-26 2014-05-13 Methylgene Inc. Inhibitors of histone deacetylase
US7253204B2 (en) * 2004-03-26 2007-08-07 Methylgene Inc. Inhibitors of histone deacetylase

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US5858365A (en) * 1993-10-29 1999-01-12 Trustees Of Boston University Methods for the treatment of wounds using butyric acid salts and derivatives
US6043277A (en) * 1994-09-16 2000-03-28 Bar-Ilan University Retinoyloxy (alkyl-substituted) methyl butyrates useful for the treatment of gastrointestinal disorders, cutaneous ulcers and wounds
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
US6011000A (en) * 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US6197743B1 (en) * 1996-07-26 2001-03-06 The Trustees Of Boston University Compositions and methods for the treatment of viral disorders
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
US6043389A (en) * 1997-03-11 2000-03-28 Mor Research Applications, Ltd. Hydroxy and ether-containing oxyalkylene esters and uses thereof
US6239176B1 (en) * 1997-03-11 2001-05-29 Beacon Laboratories, Inc. Uses of hydroxy and ether-containing oxyalkylene esters for treating metabolic conditions
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds 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
US6706762B1 (en) * 1997-05-01 2004-03-16 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
US6231880B1 (en) * 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
US20050003031A1 (en) * 1997-08-19 2005-01-06 Aylward James Harrison Anti-cancer compounds
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
US6372957B1 (en) * 1998-11-10 2002-04-16 Board Of Regents, The University Of Texas System Transgenic mouse comprising a MEF2 binding site operatively linked to an indicator gene and methods of use
US6335170B1 (en) * 1999-02-22 2002-01-01 Torben F. Orntoft Gene expression in bladder tumors
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
US20030078369A1 (en) * 1999-07-23 2003-04-24 Meinke Peter T. Apicidin-derived cyclic tetrapeptides
US6511990B1 (en) * 1999-09-08 2003-01-28 Sloan-Kettering Institute For Cancer Research Class of cytodifferentiating agents and histone deacetylase inhibitors, and methods of use thereof
US20030013176A1 (en) * 1999-09-08 2003-01-16 Nikola Pavletich Crystal structure of a deacetylase and inhibitors thereof
US20040002506A1 (en) * 1999-09-08 2004-01-01 Sloan Kettering Institute For Cancer Research Novel class of cytodifferentiating agents and histone deacetylase inhibitors, and methods of use thereof
US6541661B1 (en) * 1999-11-23 2003-04-01 Methylgene, Inc. Inhibitors of histone deacetylase
US20030082668A1 (en) * 1999-11-29 2003-05-01 Cyclex Co., Ltd Method for measuring the activity of deacetylase and method of screening for inhibitors and accelerators of the enzyme
US20050080249A1 (en) * 1999-12-08 2005-04-14 Applera Corporation Histone deacetylase-8 proteins, nucleic acids, and methods for use
US6548479B1 (en) * 1999-12-08 2003-04-15 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
US6875598B1 (en) * 1999-12-08 2005-04-05 Applera Corporation Histone deacetylase-8 proteins, nuclei acids, and methods for use
US6544957B2 (en) * 2000-01-04 2003-04-08 The Johns Hopkins University Methods and reagents for facilitating transcription
US6689558B2 (en) * 2000-02-08 2004-02-10 Sangamo Biosciences, Inc. Cells for drug discovery
US20020002183A1 (en) * 2000-02-29 2002-01-03 Bing-Yan Zhu Benzamides and related inhibitors of factor Xa
US20020061860A1 (en) * 2000-03-24 2002-05-23 Zuomei Li Antisense oligonucleotide inhibition of specific histone deacetylase isoforms
US6506574B1 (en) * 2000-04-27 2003-01-14 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
US20040087652A1 (en) * 2000-07-07 2004-05-06 Goettlicher Martin Valproic acid and derivatives thereof as histone deacetylase inhibitors
US20020065282A1 (en) * 2000-07-12 2002-05-30 Guy Georges Tetralone derivatives
US20040053820A1 (en) * 2000-07-17 2004-03-18 Hidenori Nakajima Reduced fk228 and use thereof
US6673587B1 (en) * 2000-08-11 2004-01-06 The Salk Institute For Biological Studies Histone deacetylase, and uses therefor
US20050085507A1 (en) * 2000-09-01 2005-04-21 Remiszewski Stacy W. Deacetylase inhibitors
US20040024067A1 (en) * 2000-09-01 2004-02-05 Remiszewski Stacy William Deacetylase inhibitors
US20030018062A1 (en) * 2000-09-01 2003-01-23 Remiszewski Stacy W. Deacetylase inhibitors
US6552065B2 (en) * 2000-09-01 2003-04-22 Novartis Ag Deacetylase inhibitors
US6538030B2 (en) * 2000-09-20 2003-03-25 Yih-Lin Chung Treating radiation fibrosis
US20040077726A1 (en) * 2000-09-29 2004-04-22 Clare Watkins Carbamic acid compounds comprising a sulfonamide linkage as hdac inhibitors
US20050085515A1 (en) * 2000-09-29 2005-04-21 Topo Target Uk Limited, Carbamic acid compounds comprising a sulfonamide linkage as HDAC inhibitors
US20040043470A1 (en) * 2000-10-31 2004-03-04 Yonghong Xiao Regulation of human histone deacetylase
US20030082666A1 (en) * 2000-11-21 2003-05-01 Kammer Gary M. Method of treating autoimmune diseases
US6531472B2 (en) * 2000-12-07 2003-03-11 Hoffman-La Roche Inc. Tetralone derivatives
US6376508B1 (en) * 2000-12-13 2002-04-23 Academia Sinica Treatments for spinal muscular atrophy
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
US6699902B2 (en) * 2000-12-21 2004-03-02 Beacon Laboratories, Inc. Acetyloxymethyl esters and methods for using the same
US6562995B1 (en) * 2000-12-21 2003-05-13 Beacon Laboratories, Inc. Delta dicarbonyl compounds and methods for using the same
US20040014647A1 (en) * 2000-12-22 2004-01-22 Lee Hyang Woo Apicidin-derivatives, their synthetic methods and anti-tumor compositions containing them
US20040053960A1 (en) * 2000-12-23 2004-03-18 Guy Georges Tetrahydropyridine derivatives, their preparation and their use as cell proliferation inhibitors
US20030017454A1 (en) * 2001-01-26 2003-01-23 Saraswati Sukumar Aberrantly methylated genes as markers of breast malignancy
US20040077698A1 (en) * 2001-01-27 2004-04-22 Guy Georges Tricyclic lactam and sultam derivatives and their use as histone deacetylase inhibitors
US20030083521A1 (en) * 2001-03-27 2003-05-01 Circagen Pharmaceutical, A Maryland Corporation Histone deacetylase inhibitors
US6512123B2 (en) * 2001-04-23 2003-01-28 Hoffmann-La Roche Inc. Tricyclic alkylhydroxamate derivatives
US20040072849A1 (en) * 2001-05-09 2004-04-15 Schreiber Stuart L. Dioxanes and uses thereof
US20030059812A1 (en) * 2001-06-14 2003-03-27 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
US20030013757A1 (en) * 2001-06-15 2003-01-16 Ulrike Leser-Reiff Aromatic dicarboxylic acid derivatives
US6869953B2 (en) * 2001-08-07 2005-03-22 Hoffman-La Roche Inc. N-monoacylated derivatives of o-phenylenediamines, their analogs and their use as pharmaceutical agents
US20050038113A1 (en) * 2001-09-18 2005-02-17 G2M Cancer Drugs Ag Valproic acid and derivatives for the combinatorial therapeutic treatment of human cancers and for the treatment of tumor 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
US20040087657A1 (en) * 2001-10-16 2004-05-06 Richon Victoria M. Treatment of neurodegenerative diseases and cancer of the brain using histone deacetylase inhibitors
US20050032899A1 (en) * 2001-11-06 2005-02-10 Chen Ying-Nan Pan Cyclooxygenase-2 inhibitor/histone deacetylase inhibitor combination
US20050079995A1 (en) * 2001-11-27 2005-04-14 Antonio Bedaloy Methods for inhibiting deacetylase activity
US20040091951A1 (en) * 2002-02-07 2004-05-13 Axys Pharmaceuticals, Inc. Assay for measuring acetylation or deacetylation activity of an enzyme
US20040072735A1 (en) * 2002-03-04 2004-04-15 Richon Victoria M. Methods of inducing terminal differentiation
US20040087631A1 (en) * 2002-03-04 2004-05-06 Bacopoulos Nicholas G. Methods of treating cancer with HDAC inhibitors
US20040081976A1 (en) * 2002-03-07 2004-04-29 David Sidransky Genomic screen for epigenetically silenced tumor suppressor genes
US20050009030A1 (en) * 2002-03-26 2005-01-13 Fabien Schweighoffer 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
US20040018968A1 (en) * 2002-04-15 2004-01-29 George Sgouros Use of histone deacetylase inhibitors in combination with radiation for the treatment of cancer
US20040018522A1 (en) * 2002-05-09 2004-01-29 Brigham And Women's Hospital, Inc. Identification of dysregulated genes in patients with multiple sclerosis
US20040028607A1 (en) * 2002-05-20 2004-02-12 Verdin Eric M. Methods of modulating tubulin deacetylase activity
US20040023944A1 (en) * 2002-05-22 2004-02-05 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on alpha-chalcogenmethylcarbonyl compounds
US20040029922A1 (en) * 2002-05-22 2004-02-12 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
US20040029903A1 (en) * 2002-05-22 2004-02-12 Beacon Laboratories, Inc. Histone deacetylase inhibitors based on trihalomethylcarbonyl compounds
US20040091953A1 (en) * 2002-05-23 2004-05-13 Verdin Eric M. 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
US20040077578A1 (en) * 2002-06-14 2004-04-22 Monia Brett P. Antisense modulation of histone deacetylase 2 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
US20040005574A1 (en) * 2002-07-08 2004-01-08 Leonard Guarente SIR2 activity
US20040058868A1 (en) * 2002-07-09 2004-03-25 Stephen James Methods for identification of compounds modulating insulin resistance
US20050065596A1 (en) * 2002-07-24 2005-03-24 Xufan Tseng Stents capable of controllably releasing histone deacetylase inhibitors
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
US20050020557A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors
US20050026907A1 (en) * 2003-06-10 2005-02-03 Kalypsys, Inc. Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
US20050032831A1 (en) * 2003-07-07 2005-02-10 Kozikowski Alan P. 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
US20050070467A1 (en) * 2003-09-25 2005-03-31 Fujisawa Pharmaceutical Co., Ltd. Antitumor agent

Also Published As

Publication number Publication date
PE20070195A1 (en) 2007-05-06
US20080108829A1 (en) 2008-05-08
US20080119648A1 (en) 2008-05-22
US7732475B2 (en) 2010-06-08
AU2006270322A1 (en) 2007-01-25
NO20080784L (en) 2008-03-31
WO2007011626A3 (en) 2007-05-03
US20090105485A1 (en) 2009-04-23
TW200740761A (en) 2007-11-01
EP1904452A2 (en) 2008-04-02
JP2009501236A (en) 2009-01-15
CN101263121A (en) 2008-09-10
CA2615105A1 (en) 2007-01-25
EA200800321A1 (en) 2008-06-30
US20070015809A1 (en) 2007-01-18
KR20080032188A (en) 2008-04-14
ZA200800901B (en) 2010-05-26
MA29629B1 (en) 2008-07-01
US7741494B2 (en) 2010-06-22
BRPI0613429A2 (en) 2009-02-10
US20090111996A1 (en) 2009-04-30
WO2007011626A2 (en) 2007-01-25
IL188692A0 (en) 2008-08-07
AR054866A1 (en) 2007-07-25
CR9664A (en) 2009-07-07
US20080119658A1 (en) 2008-05-22

Similar Documents

Publication Publication Date Title
US7741494B2 (en) Histone deacetylase inhibitors
US7642253B2 (en) Histone deacetylase inhibitors
US7642275B2 (en) Histone deacetylase inhibitors
US20070173527A1 (en) Histone deacetylase inhibitors
US7169801B2 (en) Histone deacetylase inhibitors
US7399884B2 (en) Histone deacetylase inhibitors
US7572914B2 (en) Kinase inhibitors
US20110070297A1 (en) Glucokinase activators
US20050159470A1 (en) Histone deacetylase inhibitors
US20050137234A1 (en) Histone deacetylase inhibitors
US20100093767A1 (en) Mitotic Kinase Inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: TAKEDA SAN DIEGO, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRESSI, JEROME C;GANGLOFF, ANTHONY R;KWOK, LILY;REEL/FRAME:033305/0894

Effective date: 20060728