CA2567835A1 - Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease - Google Patents

Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease Download PDF

Info

Publication number
CA2567835A1
CA2567835A1 CA002567835A CA2567835A CA2567835A1 CA 2567835 A1 CA2567835 A1 CA 2567835A1 CA 002567835 A CA002567835 A CA 002567835A CA 2567835 A CA2567835 A CA 2567835A CA 2567835 A1 CA2567835 A1 CA 2567835A1
Authority
CA
Canada
Prior art keywords
compound
optionally substituted
group
hydrogen
lower alkyl
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
CA002567835A
Other languages
French (fr)
Inventor
James Malecha
Stewart Noble
Christian Hassig
Paul Wash
Brandon Wiley
Charles Lawrence
Timothy Hoffman
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.)
Kalypsys Inc
Original Assignee
Individual
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
Priority claimed from PCT/US2004/018502 external-priority patent/WO2004110418A2/en
Priority claimed from US10/865,743 external-priority patent/US7271195B2/en
Application filed by Individual filed Critical Individual
Publication of CA2567835A1 publication Critical patent/CA2567835A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/20Esters of monothiocarboxylic acids
    • C07C327/30Esters of monothiocarboxylic acids having sulfur atoms of esterified thiocarboxyl groups bound to carbon atoms of hydrocarbon radicals substituted by nitrogen atoms, not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C329/00Thiocarbonic acids; Halides, esters or anhydrides thereof
    • C07C329/02Monothiocarbonic acids; Derivatives thereof
    • C07C329/04Esters of monothiocarbonic acids
    • C07C329/06Esters of monothiocarbonic acids having sulfur atoms of thiocarbonic groups bound to acyclic carbon atoms
    • 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
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/38One sulfur atom
    • C07D239/40One sulfur atom as doubly bound sulfur atom or as unsubstituted mercapto radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D251/00Heterocyclic compounds containing 1,3,5-triazine rings
    • C07D251/02Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings
    • C07D251/12Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D251/14Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom
    • C07D251/22Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom to two ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more 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
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole 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
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • C07D263/50Benzene-sulfonamido oxazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • C07D271/071,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom 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
    • C07D307/64Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom 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
    • C07D333/30Hetero atoms other than halogen
    • C07D333/34Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom 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
    • C07D333/30Hetero atoms other than halogen
    • C07D333/36Nitrogen 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen 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
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/16Esters of thiophosphoric acids or thiophosphorous acids
    • C07F9/165Esters of thiophosphoric acids
    • C07F9/1653Esters of thiophosphoric acids with arylalkanols

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Molecular Biology (AREA)
  • Cardiology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biochemistry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pyridine Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Indole Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Furan Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Disclosed herein are carbonyl compounds of having the structural formula (I) or a pharmaceutically acceptable salt, amide, ester, or prodrug thereof, Methods and compositions are disclosed for treating disease states including, but not limited to cancers, autoimmune diseases, tissue damage, central nervous system disorders, neurodegenerative disorders, fibrosis, bone disorders, polyglutamine-repeat disorders, anemias, thalassemias, inflammatory conditions, cardiovascular conditions, and disorders in which angiogenesis play a role in pathogenesis, using the compounds of the invention. In addition, methods of modulating the activity of histone deacetylase (HDAC) are also disclosed.

Description

NOVEL SULFONAMIDES AS INHIBITORS OF HISTONE DEACETYLASE FOR
THE TREATMENT OF DISEASE

RELATED DOCUMENTS
The present application claims priority to the following applications: U.S.
Patent Application No.: 60/635,019 filed December 9, 2004; U.S. Patent Application No.:
10/865,743 filed June 10, 2004 and PCT/US2004/018502 filed June 10, 2004 which are incorporated herein by reference in their entirety.

FIELD OF THE INVENTION
[0001] The present invention is directed to carbonyl compounds as inhibitors of histone deacetylase (HDAC). These compounds are useful in treating disease states including cancers, autoiininune diseases, tissue damage, central nervous system disorders, neurodegenerative disorders, fibrosis, bone disorders, polyglutamine-repeat disorders, anemias, thalassemias, inflammatory conditions, cardiovascular conditions, and disorders in which angiogenesis plays a role in pathogenesis.
BACKGROUND OF THE INVENTION
[0002] Histone proteins organize DNA into nucleosomes, which are regular repeating structures of chromatin. The acetylation status of histones alters chromatin structure, which, in turn, is involved in gene expression. Two classes of enzymes can affect the acetylation of histones - histone acetyltransferases (HATs) and histone deacetylases (HDACs). A number of HDAC inlzibitors have been characterized. However, to date no effective candidate for cancer therapy has been identified. Therefore, there is a need in the art to discover HDAC inhibitors that have effective anti-tumor activity.
SUMMARY OF THE INVENTION
[00031 Disclosed herein are carbonyl compounds having structural formula (I) or related formulate as described herein:

__~G4 G3-G2-G s Rl R2 (I) related fonnulae as described herein, including their pharmaceutically acceptable salts, esters, and prodrugs thereof, wherein GI is an optionally substituted phenyl or optionally substituted or 6 meinbered heteroaryl, G2 is an N-sulfonainide moiety having structure (II) or an S-sulfonamide moiety having structure (III);

\ j S
N
K3 (II) R4 (III);
G3 is optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl, or optionally substituted alkyl, wherein Rl and R2 are each independently selected from the group consisting of hydrogen, lower alkyl, halogen. and perhaloalkyl, or R1 and R2 taken together fonn optionally substituted cycloalkyl, wherein R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted lower alkyl, optionally substituted aryl, and optionally substituted alkaryl; or a structural element known to confer aqueous solubility, e.g. N-piperazinylethyl, N-morpholinylethyl, 1,3-dihydroxy-2N -propanoyl etc., G4 is chosen from the group consisting of optionally substituted acyl, wherein G4 taken in combination with sulfur forms a thioester, optionally substituted thiol, wherein G4 taken in combination wit11 sulfur forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in cornbination with sulfur, forms a phosphorothioate diester or phosphorothioate; and eacll R5 is independently selected from the group consisting of hydrogen, alkyl, aryl, and arylalkyl.

[0004] The invention provides phannaceutical coinpositions comprising a compound having structural formula (I) or a related formula, which are capable of inhibiting the catalytic activity of histone deacetylase (HDAC).
[0005] The invention also provides methods and compositinos for treating diseases in mainmals using coinpounds of the invention, including but not limited to, treating cancers, autoimmune diseases, tissue dainage, central nervous systein disorders, neurodegenerative disorders, fibrosis, bone disorders, polyglutamine-repeat disorders, anemias, thalasseinias, inflainmatory conditions, cardiovascular conditions, and disorders in which angiogenesis plays a role in pathogenesis.
[0006] The invention further provides imthods of inhibiting the catalytic activity and cellular function of histone deacetylase (HDAC).
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
Definition of Terms [0007] The term "pharmaceutically acceptable salt" refers to a fonnulation of a compound that does not cause significant irritation to an organisin to which it is administered and does not abrogate the biological activity and properties of the compound.
Pharmaceutical salts can be obtained by reacting a coinpound of the invention with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
Pharmaceutical salts can also be obtained by reacting a compound of the invention with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like.
[0008] The terms "physiologically acceptable" and "physiologically compatible"
refers to excipients, products, or hydrolysis products of disclosed molecular embodiments of the invention. By way of example, protected thiol prodrug embodiments may release acids upoii hydrolysis of the protected thiol. Physiologically acceptable excipients and acids are those that do not abrogate the biological activity or properties of the compound, and are nontoxic. "Physiologically acceptable" and "pharmaceutically acceptable" may be coextensive terms.
[0009] The tenn "ester" refers to a chemical moiety with formula -(R)õCOOR', where R and R' are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1. Any amine, hydroxy, or carboxyl side chain on the compounds of the present invention can be esterified. The procedures and specific groups to be used to achieve makes such esters are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3'd Ed., John Wiley & Sons, New York, NY, 1999, which is incorporated herein by reference in its entirety.
[0010] An "amide" is a chemical moiety with formula -(R)õC(O)NHR' or -(R),1 NHC(O)R', where R and R' are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded througll a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1. An amide may be an ainino acid or a peptide molecule attached to a molecule of the present invention, thereby forming a prodrug.
[0011] Any amine, hydroxy, or carboxyl side chain on the coinpounds of the present invention can be esterified or anlidified. The procedures and specific groups to be used to achieve this end is known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, which is incorporated herein in its entirety.
[0012] A "prodrug" refers to an ageint that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility over the parent drug. An example, without limitation, of a prodrug would be a coinpound of the present invention which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety. Yet another example of a prodrug are protected thiol compounds.
Thiols bearing hydrolyzable protecting groups can unmask protected SH groups prior to or siinultaneous to use. As shown below, the moiety -C(O)-RE of a thioester may be hydrolyzed to yield a thiol and a pharmaceutically acceptable acid HO-C(O)-RE.
O O
---- SH HO"'~RE
~S,,-I~ RE + H20 )0 [0013] The term "thiol protecting group" refers to thiols bearing hydrolyzable protecting groups that can unrnask protected SH groups prior to or simultaneous to use.
Preferred thiol protecting groups include but are not limited to thiol esters which release pharmaceutically acceptable acids along with an active thiol moiety. Such phaimaceutically acceptable acids are generally nontoxic and do not abbrogate the biological activity of the active thiol moiety. Exainples of pharmaceutically acceptable acids include, but are not limited to: N,N-diethylglycine; 4-ethylpiperazino acetic acid; ethyl 2-methoxy-2-phenylacetic acid; N,N-dimethylglycine; (nitrophenoxysulfonyl)benzoic acid; acetic acid;
maleic acid;
fumaric acid; benzoic acid; tartraric acid; natural amino acids (like glutamate, aspartate, cyclic ainino acids such proline); D-amino acids; butyric acid; fatty acids like palmitic acid, stearic acid, oleate; pipecolic acid; phosphonic acid; phosphoric acid;
pivalate (trimethylacetic acid); succinic acid; cinnamic acid; anthranilic acid;
salicylic acid; lactic acid; and pyruvic acids.
[0014] As used herein, the term "alkyl" refers to an aliphatic hydrocarbon group.
The alkyl moiety may be a "saturated alkyl" group, which means that it does not contain any alkene or alkyne moieties. The alkyl moiety may also be an "unsaturated alkyl"
moiety, which means that it contains at least one alkene or alkyne moiety. An "alkene"
moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an "alkyne" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
[0015] The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range sucll as "1 to 20" refers to each integer in the given range; e.g., "1 to 20 carbon atoms" means that the alkyl group may consist of I carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no nuinerical range is designated). The alkyl group may also be a mediuin size alkyl having 1 to 10 carbon atoms.
The alkyl group could also be a lower alkyl having 1 to 4 carbon atoms. The alkyl group of the compounds of the invention may be designated as "C1-CS alkyl" or similar designations.
By way of exalnple only, "C1-C4 alkyl" indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
[0016] The alkyl group may be substituted or unsubstituted. When substituted, any group(s) besides hydrogen can be the substitutent group(s). When substituted, the substituent group(s) is(are) one or more group(s) individually and independently selected from the following non-limiting illustrative list: alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, hydroxy, alkoxy, aryloxy, 0, S, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, 0-carbamyl, N-carbamyl, 0-thiocarbamyl, N-thiocarbamyl, C-ainido, N-ainido, S-sulfonainido, N-sulfonainido, C-carboxy, 0-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, and ainino, including mono- and di-substituted amino groups, and the protected derivatives thereof. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. Each substituent group may be further substituted.
[0017] Unless otlierwise indicated, when a group is described as "optionally substituted," it is meant that the group may be substituted with one or more substituents selected from the following non-limiting illustrative list: hydroxy, alkyl, alkoxy, aryloxy, cycloalkyl, aryl, carbocyclic cycloalkyl, carbocyclic aryl, heteroaryl, heterocycloalkyl, 0, S, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, 0-carbamyl, N-carbamyl, 0-thiocarbamyl, N-thiocarbainyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, 0-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, trihalomethanesulfonyl, and amino, including mono- and di-substituted amino groups, and protected derivatives thereof. Protecting groups that may form the protective derivatives of the substituents recited above are known to those of skill in the art and may be found in references such as Greene and Wuts, above. Each optional substituent may be further optionally substituted. Optionally substituted groups may be unsubstituted.
[0018] The term "halo" or, alternatively, "halogen" means fluoro, chloro, bromo or iodo. Preferred halo groups are fluoro, chloro and bromo. The terms "haloalkyl,"
"haloalkenyl," "haloalkynyl" and "haloalkoxy" include alkyl, alkenyl, alkynyl and alkoxy structures, that are substituted with one or more halo groups or with combinations thereof.
The terms "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
[0019] The terin "hetero" in such terms as "heteroalkyl," "heteroalkenyl,"
"heteroalkynyl," "heterocycloalkyl," and "heteroaryl" refers to groups in which one or more of the backbone atoms is selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
[0020] Cyclic alkyl moeities contain one or more covalently closed ring structures. Cyclic alkyl moeities can have a single ring (inonocyclic) or two or more rings (polycyclic or multicyclic). Polycyclic groups include fused polycyclic groups wherein rings share adjacent pairs of backbone atoms, and linked cyclic groups wherein the rings are separate but linked. In fused polycyclic groups, rings may share adjacent carbon atoms, or may share non-carbon atoms such as N. Linked polycyclic groups may be coiuiected by a bond or a linker. Polycyclic groups can be linked by an optionally substituted alkyl moeity including but not limited to saturated alkyl linkers, or unsaturated alkyl linlcers such as alkylene (e.g., methylene, ethylene, or propylene) or alkynylene linlcers.
[0021] The term "carbocyclic" refers to a compound which contains one or more covalently closed ring structures, wherein the atoms fonning the backbone of the ring are all carbon atoms.
[0022] The tenn "heterocyclic" refers to a compound with contains one or more covalently closed ring structures, wherein at least one ring backbone contains at least one atom which is different from carbon. Generally, heterocyclic groups can contain one to four heteroatoms, each selected from 0, S and N, wherein each ring has from 4 to 10 atoms in the ring. Geiierally, heterocyclic rings do not contain two adjacent 0 or S atoms.
An exalnple of a 4-membered heterocyclic group is azetidinyl (derived from azetidine). An example of a 5-membered heterocyclic group is thiazolyl. An example of a 6-membered heterocyclic group is pyridyl, and an example of a 10-membered heterocyclic group is quinolinyl.
[0023] The term "cycloalkyl" refers to an aliphatic cyclic alkyl moeity wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring do not give rise to aromaticity. The term "cycloalkyl" may refer to a monocyclic or polycyclic group.
Cycloalkyl groups may be fused or linked to other cyclic alkyl moeities. A
cycloalkyl group may be optionally substituted. Preferred cycloalkyl groups include groups having from tliree to twelve ring atoms, more preferably from 5 to 10 ring atoms. The term "carbocyclic cycloalkyl" refers to a monocyclic or polycyclic cycloalkyl group which contains only carbon and hydrogen. The term "heterocycloalkyl" refers to ainonocyclic or polycyclic cycloalkyl group wherein at least one ring backbone contains at least one atom which is different from carbon. Illustrative examples of carbocyclic cycloalkyl groups include the following moieties:

A-, E>, >EO, 0, 0, (> o'c,,o co, and the like.
[0024] A heterocycloalkyl group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. Heterocycloalkyl groups may be fused with one or more aryl, heteroaryl, cycloalkyl, or heterocycloalkyl groups.
Heterocycloalkyl groups may be linked with one or more aryl, heteroaryl, cycloalkyl, or heterocycloalkyl groups. Examples of heterocycloalkyl (non-aromatic heterocyclic groups) are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, iinidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl.
Illustrative examples of heterocycloalkyl groups include:

~~5~ N
Cs, NN N O O O
0' CN N O O S N
O
~ N-N

o N I0 ( N/\O
ON N c N
H
O
N-S=O CT
N N N N a O
, N .~' O
N N N N
~~~ N

N N and the like.

[0025] The terms "aryl" or "aromatic" refer to a group which has at least one ring having a conjugated pi electron system, Aryl groups can be carbocylic aryl groups or heteroaryl groups. The term "carbocyclic aryl" refers to a group (e.g., phenyl) in which all ring backbone atoms are carbon. The terms "heteroaryl" or "heteroaromatic"
refer to an aryl (aromatic) group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. Aryl groups may be optionally substituted. Aryl groups may be monocyclic or polycyclic. Polycyclic aryl groups may be fused or linked. Polycyclic aryl groups can be fused or linked to aryl groups or cycloalkyl groups.
[0026] Examples of heteroaryl groups include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl.
Polycyclic heteroaryl groups may be attached througll carbon ring backbone atoms, or may be attached througll ring backbone heteroatoms, especially N, depending on structure of the group. For instance, a group derived from pyrrole may be pyrrol-l-yl (N-attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole may be iinidazol-l-yl or imidazol-3-yl (both N-attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached). Polycyclic heteroaryl groups include benzo-fused ring systeins and ring systems substituted with one or two oxo (=0) moieties such as pyrrolidin-2-one.
[0027] Illustrative examples of heteroaryl groups include the following moieties:
N S N
>, OO( ~N// N

N CN CN
N\ / ~N N) N ~I~ N /N
N~ N
N ~,i S

a:), N' N' and the like.
[0028] The substituent "R" appea.ring by itself and without a number designation refers to a substituent selected from the group consisting of optionally substituted alkyl, including optionally substituted alkenyl or alkynyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl (bonded through a ring carbon) and optionally substituted hetercycloalkyl (bonded through a ring carbon).
[0029] An "O-carboxy" group refers to a RC(=O)O- group, where R is as defined herein.
[0030] A "C-carboxy" group refers to a -C(=O)OR groups where R is as defined herein.
[0031] An "acyl" group refers to a -C(=O)R group.
[0032] An "acetyl" group refers to a-C(=O)CH3, group.
[0033] A"trihalomethanesulfonyl" group refers to a X3CS(=O)2- group where X
is a halogen.
[0034] A "cyano" group refers to a -CN group.
[0035] An "isocyanato" group refers to a -NCO group.
[0036] A "thiocyanato" group refers to a -CNS group.
[0037] An "isothiocyanato" group refers to a -NCS group.
[0038] A "sulfinyl" group refers to a-S(=O)-R group, with R as defined herein.
[0039] A"S-sulfonainido" group refers to a-S(=O)2NR, group, with R as defined herein.
[0040] A "N-sulfonamido" group refers to a RS(=O)2NH- group with R as defined herein.
[0041] A"trihaloinethanesulfonamido" group refers to a X3CS(=0)2NR- group with X and R as defined herein.
[0042] An "O-carbamyl" group refers to a -OC(=O)-NR, group-with R as defined herein.

[0043] An "N-carbamyl" group refers to a ROC(=O)NH- group, with R as defined herein.
[0044] An "O-thiocarbamyl" group refers to a -OC(=S)-NR, group with R as defined herein.
[0045] An "N-thiocarbamyl" group refers to an ROC(-S)NH- group, with R as defined herein.
[0046] A "C-amido" group refers to a -C(=O)-NR2 group with R as defined herein.
[0047] An "N-amido" group refers to a RC(=O)NH- group, with R as defined herein.
[0048] The term partially halogenated alkyl refers to an alkyl group having both hydrogen and halogen substituents, [0049] The term "perhaloalkyl" refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
[0050] The term "lower perfluoroalkoxy" refers to a radical -O-(CX2),iCX3 where X is any halogen, preferable F or Cl, and n is 1-5.
[0051] When two substituents taken together along with the two ring carbons to wl7ich they are attached form a ring, it is meant that the following structure:

is, for example, representative of a structure such as the following:
~
[0052] In the above exatnple, Rl and R2, taken together along with the two ring carbons to which they are attached, form a six-membered aroinatic ring.

Solubility is a thermodynamic parameter and plays an important role in the determination of a drug's bioavailability. Since a drug inust be soluble in the gastrointestinal fluid to be orally active, the rate and extent of dissolution depend critically upon intrinsic water solubility (neutral species solubility) ( Dressman, J.; Amindo, G. L.,;
Reppas, C.; Shah.
V. P. Pharm. Res., 1998, 15, 11.) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development setting have been described (Lipinski C. A. et al. 1997 Adv. Drug Deliv. Rev. 23, 3-25)Adv. Drug Deliv.
Rev. 23, 3-25 ).
Traditional analytical methods define solubility as the concentration of material in solution at equilibrium with its solid fonn. In this method a compound is extensively shaken in the buffer of choice, filtered through a micropore membrane, and the concentration of dissolved compound in the filtrate determined. This approach results in a thermodynamic solubility assessment. For discovery, it is beneficial to measure kinetic solubility in which a compound DMSO solution is added to aqueous buffer. Several high throughput approaches for solubility have been described, e.g. turbidimetric method (Bevan, C. and Lloyd, R. S. Anal.
Chem. 2000 72, 1781-1787), nephelometric method (Avdeef, A. (2001) High througllput measurements of solubility profiles. In Pharmacokinetic Optimization in D ug Research;
Biological, Physicochemical, And Computational Strategies (Testa, B. et al., eds), pp. 305-326, Verlag Helvitica Chimica Actaand). Measurement of solubility at multiple pH levels (pH 1-8), is more useful that a single pH, since many drug candidates contain ionizable groups. A solubility-pH profile provides the pH gradient of the gastrointestinal tract.
Accurate understanding of a compound's solubility is also necessary to not only prepare and dispense formulations, but also to evaluate new chemical series and provide feedback to drive synthetic optimization. Structural series of compounds are synthesized with the aim of iinproving solubility by the addition of various chemical moieties. Sti-uctural elements known to confer aqueous solubility on otherwise insoluble molecular entities include but are not limited to N-piperazinylethyl, N-morpholinylethyl, 1,3-dihydroxy-2N -propanoyl moieties. Common solubilizing groups often incorporated in synthetic approaches to improve solubility of molecules include ainine functionality, such as dimethylainino, diethylamino, piperazinyl, N-methyl-N-isopropylainino, morpholino, pyrrolidino moieties, or groups bearing aliphatic alcohol functionality, such as that found in ethanolamine or glycerol.

In certain einbodiments of the invention, a structural element known to confer aqueous solubility is incorporated in a compound of the invention. Such structural elements are preferably attached to synthetically accessible regions of the coinpound.
In certain einbodiments, such structural elements are attached to or incorporate synthetically available N atoms in amine or amide or sulfonamide moieties of the compound. In certain embodiments a solubilizing group is attached to or incorporates a N atom and is chosen from the group consisting of dimethylamino, diethylamino, piperazinyl, N-methyl-N-isopropylamino, morpholino, pyrrolidino moieties, or groups bearing aliphatic alcohol functionality, such as that found in ethanolamine or glycerol.

Compounds of the invention The invention provides compounds having structural fonnula (I), s G3"G2-G1 \G4 Rl R2 (I) or a pharmaceutically acceptable salt, amide, ester, or prodrug thereof,wherein G1 is optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl, or G1 and R3 taken together form an optionallysubstituted heterocycloalkyl, G2 is an N-sulfonamide moiety having structure (II), an S-sulfonarnide moiety having structure (III), or an amide of the fonn -NR3C(O)- or -C(O)NR3-:

\\S/ S
"-R3 (II) R4 (III);
G3 is optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl, or optionally substituted alkyl, Rl and R2 are each independently selected from the group consisting of hydrogen, lower alkyl, halogen and perhaloalkyl, or Rl and R2 taken together form optionally substituted cycloalkyl, R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted lower alkyl, optionally substituted aryl, and optionally substituted alkaryl, or a structural element known to confer aqueous solubility, including but not limited to N-piperazinylethyl, N-morpholinylethyl, or 1,3-dihydroxy-2N-propanoyl, or R3 and R4 taken togetller form an optionally substituted heterocycloalkyl, and G4 is chosen from the group consisting of optionally substituted acyl, wherein G4 taken in combination with sulfur forins a thioester, optionally substituted thiol, wherein G4 taken in combination with sulfur forms a disulfide, and structure (IV) P\ oR5 yO-R5 IV

wherein G4, taken in combination with sulfur, forms a phosphorothioate diester or phosphorothioate.
In certain embodiments of compounds having Structure (I) as described above, G1 is an optionally substituted phenyl having structure (IV) or (V):

X~ O X~ O
~\ S,G4 S, X2~ ;_ R1 R2 2 ~
G3-G2 (IV) X2 (V) wherein Xl and X2 are eacll independently selected from the group consisting of hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkoxy.
In certain elnbodiments, compounds have the structure of Forinula (1), wherein G1 is an optionally substituted phenyl having structure (IV) or (V) and G2 is N-sulfonainide moiety having structure (VI) or (VII);
X, O
S, X2~ G4 X O
R1 R2 ~~ S'G
\
O O, NR3 OO I/~ 4 G3 N R, R2 (VI) (VII) In certain embodiments, compounds have the structure of Formula (1), wherein Gz is an optionally substituted phenyl having structure (IV) or (V) and G2 is N-sulfonamide moiety having structure (VI) or (VII), and G3 comprises is an optionally substituted phenyl of structure (VIII) or (IX) X, O
X2 P-"' R \G4 O~, NR3 X / i X
3 l\ \ 5 X4 (VIII) X, O
S, SN , .\ R, R2 X4 X5 (IX) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, liydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxanido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, halo alkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylainino, aminothiocarbonylaminoalkyl.
In certain embodiments, compounds have the structure of Formula (1), wherein GI is an optionally substituted phenyl having structure (IV) or (V), G2 is N-sulfonamide moiety having structure (VI) or (VII), G3 comprises is an optionally substituted phenyl of structure (VIII) or (IX), and G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE
is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in combination with sulfur, forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in coinbination with sulfur, forms a phosphorothioate.
diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and Xl and X2 are each independently selected froin hydrogen, halogen, hydroxyl, lower alkyl, lower perizaloalkyl, lower alkoxy and lower perhaloalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. Particular einbodiments include:

S~/
~~~~ ~ ~~/,0 II
S.H / 0 C'N 0 ~cr H

Me0 0S~ S 1 0 1 ~S~ SII

H H
Me0) / CIaCi ~ S\~-op R\ ip I j1 S.N 0 S.N / 0 I/ H I/ H

OO SY OO I~ SY
S. O F3C S' 0 H H

oso SY Oso icr"--- SY
j~ H Me0~~ H0 %\ ~
F

O
\ S'N ID,-~,SyQN
01 /0 Sy ~S~

N
~ / H

OSO HI~ SY OSO H/ Sy C O C
I

R"P SY 0\\,p S-Ir C SN~ 0 Me0 ~ S.N J(D

H I H
Ci \%

O O

OõO I\ SY O0 O sY
I\ S'H / 0 H 0 '\!/
F CI
O O
CI OSO J/ S0 F co OSO I/ S0 H I H

a,, SY S~
SO 0 ~S~
'N ~ O
H I / H

F

O O
\ S \ \ S~
OS~ O OSO I/ O

O S SY
SO 0 OSO I \ / CI 0 SY J&SyNH
S N r 0 OS~ / 0 H
F3CO ~ F3CO

O
R\ iP jqr"" S O Y
~oj:~ \ SN I/ H F O

O
~
pS. O H~ ~ 0 F3CO I)", O O
R'lp ~
S. H/ 0 O O
H~OMe S.H O
I/

O /
S ~~
OO
S.N 0 O~
H
O
1)", In certain embodiments, compounds have the structure of Formula (1), wherein Gt is an optionally substituted phenyl having structure (IV) or (V), G2 is N-sulfonamide moiety having structure (VI) or (VII), and G3 comprises an 5 or, 6 membered heteroaromatic optionally substituted by X3, X4, X5, wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl; N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkeilyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxainido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylainino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aininoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl. In par-ticular embodiments, G3 is selected can be any of the following:
N-~
O~J N
N N H
HN N 1i S~i <\O J ~NN
N O

N~i ~N ~N~i N
N ININ N:NJ
H

N N . N
N N

N N % N-N, ri -N~ N N~ L//N N
N~
I rN N
~ J

In further embodiments, G4 is an optionally substituted acyl of the forinula -C(O)RE , wherein RE is any pharmaceutically acceptable acid; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substiiuted cycloalkyl; and Xl and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural eleinent known to confer aqueous solubility. Particular embodiments include:

O O
OO S,~r OO STr ~ O S SN O
O\N~ H N:C' H

O O
o~H 0 0S0 S0 N ' 0 ~ --~s N, N
H
O O

O \01 I\ I S 101 N \SHN N S' N
~ ,O
O HO O
Jo--~ 5\ S

O HN ~OI O\S/N

Cl- ~O N O
k,N N
O ~Nr O
\ / I S~ ~ S o/

O\ ~N \ O O\ ,N
a~l o I ~O
O HO
O

jo-,~ S1~ ~ ~ ~ S1~(~
OHN 0 O\N\ 0 ~ S
O O

N N

O
S S",/
~ O 101 ~ HN O IHN

\N II s S~ \O
\O
O'N

O p HN S-k" HN
O S,O N

~ ~ INI~N
N

/
~-N
/ O
41irs-k, N . N HN ~O 0 S~ O 0 , N"~ N /

In certain embodiments, compounds have the structure of Forinula (1), wherein G1 is an optionally substituted phenyl having structure (IV) or (V), G2 is N-sulfonamide moiety having structure (VI) or (VII), and G3 is an optionally substituted alkyl. In further embodiments, G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE is any pharmaceutically acceptable acid; R1 and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. Particular embodiments include:
O
sy oSo \

H
O
S
OSO

\ S~
O O
,N
O O
O O

1 S0 ~~ ~
~S. N O N \ O
~' ~O ' ~-O

~ ~ II
HN \ S~ N

In certain einbodiments , compounds have Structure (I) as described above, Gl is an optionally substituted phenyl having structure (IV) or (V),and G2 is S-sulfonamide moiety having structure (X) or (XI) as shown below.
X, X2 X, 0 R4 \~/ R~ R2 Rq ri\ s\G4 G3 N~S~ S'G4 G" N,S~ % R~ R2 O~ ~0 3 /s ~
0 (X) 0 0 X2 (XI) In further embodiments, G3 is an optionally substituted phenyl of structure (XII) or (XIII) X, ~ 2 X, 0 R2 R4 S, R4 ' R, N~S S~G4 \ N~S" R1 R2 X3 00 0 X3 l' O 0 X2 /
X4 X5 (XII) X4 X5 (XIII) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylallcoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxainide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylainidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aininothiocarbonylamino, aminothiocarbonylaminoalkyl. In further embodiments, G4 is an optionally substituted acyl of the forinula -C(O)RE , wherein RE is any pharmaceutically acceptable acid; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and XI and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural eleinent known to confer aqueous solubility. Particular embodiments include:

O
S
N\ Y

~ ~ oso O O
SY SY
OS O OS ~ ~ O
HN ~O I ~HN ~O

/
~
F

N"~s joj ~ ~HN ~p I ~p F ~ H3CO 0 0 S,,r )1tSOS O /s IOI
~ \ ' \O ~ \ NO
HO
OH

O\ p p\ 0 ~O \ HN~SO \ N SO
~O

O 0 Os Os ~ I

HN O O HN ~p O
~
O N
~ p p os ~ I S~ s I~ HN ~O O I~ HN ~O
O
CI ~ ~

Og g'~~ p ~
, ~
~?'**' N O 0 rz,~""THN O O
/

~ P 1 ~ cv-~
N,O O N N,SO O S

r S\ ~ r SY
O ~ O
NISO
HWSO CCF
F

SY Sy O O O~ O
\ HN'S10 \ HN'SO
~ r I .r CI
CI

SY r-Sy ~S O \ ~S \ ~ O
a H N I N
CI CI
CI

0, 0 0 HN
S O O F

p , ~ ~ o~ ~ I 0 NO S~ N.SO

F r r CI

HN'p HNO
O
CI C!

In certain embodiments wherein compounds have Structure (I) as described above, Gt is an optionally substituted phenyl having structure (IV) or (V),and G2 is S-sulfonainide moiety having structure (X) or (XI), G3 can be a 5 or 6 meinbered heteroaromatic optionally substituted by X3, X4, Xs as shown below:
N~
o~J N
N N H

<\O--~j' N -N HN N 5 N O

I N1~ ~N1~ ~N~i N
INI~ N,,~,- N NN
H
N
N ~ LS~ ~/ . / N ~//
N N /

N r N-N, ~ N N N N

wherein X3, X4 and X5 are each indepeildently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxainidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylainino, aminoalkyl, , dialkylaminoalkyl, aininoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylarnino, aminothiocarbonylaininoalkyl.
In certain embodiments of the compounds described in the preceding paragraph, wherein compounds have Structure (I) as described above, Gl is an optionally substituted phenyl having structure (IV) or (V),and G2 is S-sulfonamide moiety having structure (X) or (XI), G3 can be a 5 or 6 meinbered heteroaromatic optionally substituted by X3, X4, X5 as described in the preceding paragraph, G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE is any pharmaceutically acceptable acid; Rl and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected fiom hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. Particular einbodiments include:
O O
O \ 0 O \ S~
N HN' N N~S\

O O
\ ~
HO Sj 0S O }~S \ I TO
N \ NHO N0 ' N N
O O

SY N io Sy ~ O ~O
I \ N \O N
Nr~

O O
HO
O\ SY ~~ )cr&sY0 ~S O
N r NH ~ ' N N~
~N
~ C' O
S'Ir S O HN
N~NH~O c O O
~
-N /
/ O ~ O
9\ ~
' ~ S
NISO S N NH S\
~O
/OD ~ 0 N %~ 0 \N N

/ I O HO a O
,5~ \ S.S~ S~
N~
HNN O O O
N- N(~
In certain embodiments wherein coinpounds have Structure (I) as described above, Gl is an optionally substituted phenyl having structure (IV) or (V),and G2 is S-sulfonamide moiety having structure (X) or (XI),G3 can be an optionally substituted alkyl.
In further embodiments, G4 can be an optionally substituted acyl of the formula -C(O)RE , wherein RE
is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in combination with sulfur, forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in combination with sulfur, foirns a phosphorothioate diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and Xl and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural element kiiown to confer aqueous solubility. Particular embodiments include:

O ~ O
HO
/ SY -'N SY
~S I O z 0S O
N ~p N

O O
S~
0 S 0 O\ 0 N' ~HN'%

O
O O
--,,HN O O ~NO
O
In certain einbodiments of compounds having Structure (I) as described above, Gl is a or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV);
O
S, 5or6 G4 Membered R1 R2 Heteroaromatic ring X, (XIV) wherein Xi and X2 are each independently selected from the group consisting of hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkoxy.
In certain embodiments, coxnpounds have the structure of Fonnula (1) above, Gl is a 5 or 6 meinbered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), and G2 is a sulfonainide having an N-sulfonamide moiety of structure (XV) or an S-sulfonamide moeity of structure (XVI) O O
S, S~
5 or 6 G4 5 or 6 G4 Membered R, R2 Membered R, R2 Heteroaromatic Heteroaromatic 0~ 0 ring R4 ring N, X' (XV) X, (XVI) In certain einbodiments, coinpounds have the structure of Fonnula (1) above, G, is a 5 or 6 membered heteroaromatic optionally substituted by Xi, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV) or S-sulfonainide (XVI), and G3 is an optionally substituted phenyl of structure (XVII):

S, 5 or 6 G4 Membered R, R2 Heteroaromatic X 0 0 ring N
C ~ R3 X2 X, X4 X5 (XVII) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylainino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylainino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyllaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkyltllio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
In certain einbodiments, compounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV) or S-sulfonamide (XVI), and G3 is an optionally substituted phenyl of structure (XVII), and X3, X4 and X5 are selected as described in the preceding paragraph, G4 can be an optionally substituted acyl of the formula -C(O)RE , wherein RE is any phalmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in combination with sulfur, forms a disulfide, and -P(O)(OR5)2 or --P(O)(OH)2, wherein G4 taken in combination with sulfur, forms a phosphorothioate diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and Xl and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. In these embodiments, G1 can be any of:

O1~
N NJ
H
~O J'' ~N N
N O \J ~
~Nl~ IIN~~' NNN N~
~ NJ N:N~

H
N .
N N ~J N
N N

N N N-N, N N N~ N N
Particular embodiments include:

O O
S S~,/ N~
0 HN~\N IOI ~\ HN O
O ~ \ O
~
O
HN~N 0 \ S O S
O~~ HN ~ 0 NI ~
0 0 ~ N O
~
H3CO N~
I

O O

O~ NS ~ O S S
~HN 1 101 O O

CI ~

O O
NS /S S
~\ ~N- ~N ~ O HN"\N K 0 I\ S,.o O I\ o ~
a In certain embodiments, coinpounds have the structure of Fonnula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xi, X2 of structure (XIV), G2 is an N-sulfonainide moiety having structure (XV) or S-sulfonamide (XVI), and G3 can be a 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5 of structure (XVIII) S, or 6 G4 Membered R, R2 Heteroaromatic X3 O~ O ring S~N

X4 X5 (XVIII) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perllaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxainidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylalnino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylainino, aminocarbonylalkoxy, amino carbonylamino, aminothiocarbonylainino, or aminothiocarbonylaminoalkyl.
In certain embodiments, compounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV) or S-sulfonamide (XVI), and G3 is can be a 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5 of structure (XVIII), with X3, X4 and X5 selected as described in the preceding paragraph, G4 can be an optionally substituted acyl of the fonnula -C(O)RE , wherein RE is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in coinbination with sulfur, forins a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in combination with sulfur, forms a phosphorothioate diester or phosphorothioate; R1 and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and Xl and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perlialoalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. In these einbodiments, G3 can be :

/S' 01 N~ 'NJ H

</N N H N N, S~i N O ~
03- N " II N~/ ~N'' N
N NN NN
H
.
N ~ ~S~ N 'N N

N"N/ i N
N~
LN~ N N~ N

Particular embodiments include:

O
O N S"r S
~ HN~ I O ~ HN O
N N~ ~
O I N O

O O
O S~ S S~

QS~O N~N ~ 101 O SHN I , ~N ~ O:1 <\
O'N N
In certain embodiments, compounds have the structure of Formula (1) above, G1 is a 5 or 6 membered heteroaromatic optionally substituted by Xi, X2 of structure (XIV), and G2 is an N-sulfonamide moiety having structure (XV), wherein G3 comprises an optionally substituted alkyl.

S, 5or6 C'4 Membered R, R2 Heteroaromatic 0~ s~
3 ring G ~S1N

X1 (XV) In certain embodiments, compounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV), G3 coinprises an optionally substituted alkyl, and G4 is an optionally substituted acyl of the forinula -C(O)RE , wherein RE
is any pharmaceutically acceptable acid, aii optionally substituted alkylthiol, wherein G4 taken in combination with sulfur, forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in combination witl7 sulfur, fonns a phosphorothioate diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and X, and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R3 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. In certain einbodiments, Gl can be:

O/~'~
N N H
N N
''O J'~ \ N HN'i 5~--' N O

02' N ' I'N~~" IIN\~ NI' N,,,J N~N N, ~
N
H

N S~ N N
N N

LN,,- N N
N~
N N~

Particular embodiments include:

O N\ Sy SY
HN~ :~' ~~ HN 0 O N yg O
+ S S g~
ONN ~ N ~ 101 \O b In certain embodiments, compounds have the structure of Formula (1) above, G, is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV) or S-sulfonainide (XVI), and G3 is an optionally substituted phenyl of structure (XIX):

S, 5 or 6 04 Membered R, R2 Heteroaromatic R4 ring N, S
Xg X2 X4 X5 xI (XIX) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxainido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylainino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylainino, alkylthio, arylainino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkyltllio, alkylsulfinyl, arylsulfinyl, diallcylamino, aininoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, aminothiocarbonylaminoalkyl.
In certain embodiments, coinpounds have the structure of Formula (1) above, Gl is a 5 or 6 meinbered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an N-sulfonamide moiety having structure (XV) or S-sulfonamide (XVI), G3 is an optionally substituted phenyl of structure (XIX) as described in the preceding paragraph, and G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in combination with sulfur, fonns a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wlierein G4 taken in coinbination with sulfur, forms a phosphorothioate diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or Rl and R2 taken together fonn optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. In further embodiments, Gj can be:

S~
N Ni~ O~'~

N HN'N1-~
N O \--~j 021- N ~ IIN NNN N
N J NN
H
N-S, ~/N
N N
N N

LN~ ~ N
N N~
N N
(N
'~~~J
N~N/
Particular embodiments include:

O O
S S N S
O'S~\ ~ 0 O
HN~ ~O N 0 \ HN~SO
F~ /
~

OCH3 oS ~~ Sy 0\ O ~ Slir HN O ~ HN'SpJ'\\N 0 ~ i S
y S
-\~ ~'' HN \S' N 0 N, S\ 1 ~ S~
I \ O

CI ~ ~

0S-N I' o'S Sy N 'O O'N 0 a HN'SO N O

In certain embodiments, compounds have the structure of Formula (1) above, G1 is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is a S-sulfonamide (XVI), and G3 is a 5 or 6 membered optionally substituted heteroaromatic of structure (XVIII), G3 can be a 5 or 6 meinbered heteroaromatic optionally substituted by X3, X4, XS of structure (XX) S, or 6 C'4 Membered RI R2 Heteroaromatic X3 R4 ring S
O~ ~\O X2 X~
X4 X5 (XX) wherein X3, X4 and X5 are each independently cl7osen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylainino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxainido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkyltliio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylainido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylainino, aininoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or arninothiocarbonylaminoalkyl.

In certain embodiments, compounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is a S-sulfonamide (XVI), and G3 is a 5 or 6 membered optionally substituted heteroaromatic of structure (XVIII), G3 can be a 5 or 6 meinbered heteroaromatic optionally substituted by X3, X4, X5 of structure (XX), and G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 taken in coinbination with sulfur, forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in combination with sulfur, forms a phosphorothioate diester or phosphorothioate;
R, and R2 are eacll independently hydrogen, lower alkyl or Rl and R2 taken together form optionally substituted cycloalkyl; and Xl and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility. In further einbodiments, Gl and G3 can each be independently selected from:

O~~J N
N N H

~O <N ~ HN
N O

C2J N ~ IINII N\/ N
N NN N;N~
H
S -S N-S, N
N N N N~ N

N r N-N, ~N NN~ LIJN N~
%/>
\N
Particular embodiments include:

O
N\ S O
' /
O o\S S S ~"
N~NH ~
O I~ N' ~O O
\,N N~
O O
S
O'S /S I O S /O I S~ 11 O N yN N N H~N O
~, ~ ~ o N C
In certain embodiments, compounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an S-sulfonamide (XVI), and G3 is an optionally substituted alkyl.

O
5 or 6 S\G4 Membered R, R2 Heteroaromatic R4 ring N, X, (XVI) In certain embodiments, coinpounds have the structure of Formula (1) above, Gl is a 5 or 6 membered heteroaromatic optionally substituted by Xl, X2 of structure (XIV), G2 is an S-sulfonamide (XVI), G3 is an optionally substituted alkyl, and G4 is an optionally substituted acyl of the formula -C(O)RE , wherein RE is any pharmaceutically acceptable acid, an optionally substituted alkylthiol, wherein G4 talcen in coinbination with sulfur, forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 talcen in combination with sulfur, forms a phosphorothioate diester or phosphorothioate; Rl and R2 are each independently hydrogen, lower alkyl or RI and R2 taken together fonn optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy. R4 can be hydrogen, optionally substituted lower alkyl, or a structural eleinent loiown to confer aqueous solubility. In further embodiments, Gl can be:

p~~~
NJ N O~
H
~J ~N N HN S
N O

I N1~ N N N N ~N I~
~ INIJ ~ N.NJ
H
S NS, N
N N -N~ N
NN N'N, N N N N N~
(N

N-, N
Particular einbodiments include:

N S O
~ o~\ O \0~~ S~
N p p O

S
~S~S 0 lir ~\ ~S~O Dr S Il N N3 N' ~\ p N p In one einbodilnent, a compound having the structure of Fonnula (1), has the structure consisting of:

O
F3CO/ S~
~ ( ~NH 0 OSO

[0053] In another aspect, the invention relates to a compound selected from the goup consisting of the compounds set forth in the examples, or a pharmaceutically acceptable salt, ester, amide, or prodrug thereof:

Exemplary compounds and pharinaceutically acceptable esters or prodrugs thereof the invention include, but are not limited to, disulfide dimers, mercaptans, and thioesters of coinpounds of Formula (I).

Uses of compounds of the invention [0054] In accordance with one aspect, the present invention provides compounds of Formula (I), where each compound is capable of inhibiting the catalytic activity of histone deacetylase (HDAC). In another aspect, the present invention provides phannaceutical compositions comprising compounds of Formula (I), capable of inhibiting the catalytic activity of histone deacetylase (HDAC).

[0055] In accordance with another aspect, the present invention provides compounds of Formula (I), capable of inhibiting the cellular function of HDAC.
In anotller aspect, the present invention provides phannaceutical compositions comprising compounds of Forlnula (I), capable of inhibiting the cellular function of histone deacetylase (HDAC).
[0056] In accordance with yet another aspect of the invention, the present invention provides methods and compositions for treating certain diseases or disease states.
Methods and compositions are provided for using compounds of the invention for treating diseases or disease states including, but not limited to, cancers, autoimmune diseases, tissue damage, central nervous system disorders, neurodegenerative disorders, fibrosis, bone disorders, and disorders in which angiogenesis play a role in pathogenesis, [0057] In accordance with one aspect, methods and compositions of the invention are used for treating cancer. In some embodiments, but without limitation, the tenn cancer refers to and is selected from disorders such as colon cancer, breast cancer, ovarian cancer, lung cancer and prostrate cancer, tumor invasion, tumor growth, tumor metastasis, and cancers of the oral cavity and pharynx (lip, tongue, mouth, pharynx), esophagus, stoinach, small intestine, large intestine, rectum, liver and biliary passages, pancreas, larynx, bone, connective tissue, skin, cervix uteri, corpus endornetriurn, testis, bladder, kidney and other urinary tissues, eye, brain and central nervous system, thyroid and endocrine gland. The term "cancer" also encompasses Hodgkin's disease, non-Hodgkin's lymphoinas, inultiple myeloma and hematopoietic malignancies including leukemias (Cluonlc Lymphocytic Leukeinia) and lymphomas including lymphocytic, granulocytic and monocytic.
Additional types of cancers which may be treated using the coinpounds and methods of the invention include, but are not limited to, adrenocarcinoma, angiosarcoma, astrocytoma, acoustic neuroma, anaplastic astrocytoma, basal cell carcinoma, blastoglioma, chondrosarcoma, choriocarcinoma, chordoma, craniopharyngioma, cutaneous melanoma, cystadenocarcinoina, endotheliosarcoma, embryonal carcinoma, ependymoma, Ewing's tumor, epithelial carcinoma, fibrosarcoma, gastric cancer, genitourinary tract cancers, glioblastoma multifonne, head and neck cancer, hemangioblastoma, hepatocellular carcinoma, hepatoma, Kaposi's sarcoma, large cell carcinoma, cancer of the larynx, leiomyosarcoina, leukeinias, liposarcoma, lyinphatic system cancer, lymphomas, lyinphangiosarcoma, lymphangioendotheliosarcoma, medullary thyroid carcinoma, medulloblastoma, meningioma mesothelioma, myelomas, inyxosarcoma neuroblastoma, neurofibrosarcoina, oligodendroglioma, osteogenic sarcoma, epithelial ovarian cancer, papillary carcinoma, papillary adenocarcinomas, parathyroid tumours, pheochromocytoma, pinealoma, plasmacytomas, retinoblastoma, rhabdomyosarcoma, sebaceous gland carcinoma, seminoma, skin cancers, melanoma, small cell lung carcinoma, squainous cell carcinoma, sweat gland carcinoma, synovioma, thyroid cancer, uveal melanoma, and Wilm's tuinor.
[00581 In accordance with another aspect, methods and compositions of the invention are used for preventing neoplasias including, but not limited to, brain cancer, bone cancer, a leukemia, a lymphoma, epithelial cell-derived neoplasia (epithelial carcinoma) such as basal cell carcinoma, adenocarcinoma, gastrointestinal cancer such as lip cancer, mouth cancer, esophogeal cancer, small bowel cancer and stomach cancer, colon cancer, liver cancer, bladder cancer, pancreas cancer, ovary cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamous cell and basal cell cancers, prostate cancer, renal cell carcinoma, and other known cancers that effect epithelial cells throughout the body. The neoplasia can be selected from gastrointestinal cancer, liver cancer, bladder cancer, pancreas cancer, ovary cancer, prostate cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamous cell and basal cell cancers.

[0059] In accordance with another aspect, metllods and compositions of the invention are used for treating autoimmune diseases including, but not limited to:
autoimmune disease that targets the nervous system, e.g., inultiple sclerosis, myasthenia gravis, autoimmune neuropathies such as Guillain-Barre syndrome, autoimmune uveitis;
autoimmune disease that targets the gastrointestinal system, e.g., Crohn's disease, ulcerative colitis, primary biliary cirrhosis; autoimmune hepatitis; autoimmune disease that targets the blood, e.g., autoimmune heinolytic anemia, pernicious anemia, autoimmune thrombocytopenia; autoimmune disease that targets endocrine glands, e.g., Type 1 or immune-mediated diabetes mellitus, Grave's disease, Hashimoto's thyroiditis, autoimmune oophoritis and orchitis, autoimmune disease of the adrenal gland; autoimmune disease that targets blood vessels, e.g., temporal arteritis, anti-phospholipid syndrome, vasculitides such as Wegener's granulomatosis, Behcet's disease; autoimmune disease that targets multiple organs including the musculoskeletal systein, e.g., rheumatoid arthritis, scleroderma, polymyositis, dermatomyositis, spondyloarthropathies such as ankylosing spondylitis, Sjogren's syndrome; autoimmune disease that targets skin, e.g., psoriasis, dermatitis herpetiformis, pemphigus vulgaris, or vitiligo.
[0060] In accordance with another aspect, methods and compositions of the invention are used for treating disease states characterized by tissue damage, where the disease states include, but are not limited to, vascular diseases, inigraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheuinatic fever, type I diabetes, neuromuscular junction disease including myasthenia gravis, white matter disease including multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, nephritis, hypersensitivity, swelling occurring after injury, myocardial ischemia, and the like.
[0061] In accordance with another aspect, methods and coinpositions of the invention are used for treating the fibrosis which occurs with radiation therapy.
[0062] In accordance with another aspect, methods and compositions of the invention are used for treating subjects having adenomatous polyps, including those with familial adenomatous polyposis (FAP). Additionally, the present coinpounds and methods can be used to prevent polyps from forming in patients at risk of FAP.
[0063] In accordance with another aspect, methods and compositions of the invention are used for treating anemias or thalassemias including, without limitation, sickle cell anemia.

[0064] In accordance with another aspect, methods and coinpositions of the invention are used for treating a cardiovascular condition, e.g., cardiac hypertrophy and heart failure.

[0065] In accordance with another aspect, methods and coinpositions of the invention are used for treating diseases related to an inflainmatory condition including, but not limited to, rheumatoid arthritis (RA), inflainmatory bowel disease (IBD), ulcerative colitis and psoriasis.

[0066] In accordance with another aspect, methods and compositions of the invention are used for treating certain central nervous system disorders including, but not limited to, Parkinson's disease, Alzheimer's disease, Alzheimer's dementia, and central nervous system damage resulting fiom stroke, ischeinia and trauma.
[0067] In accordance with another aspect, methods and compositions of the invention are used for treating a neurological or polyglutamine-repeat disorder including, but not limited to, Huntington's disease, Spinocerebellar ataxia 1(SCA 1), Machado-Joseph disease (MJD)/Spinocerebella ataxia 3 (SCA 3), Kennedy disease/Spinal and bulbar muscular atrophy (SBMA) and Dentatorubral pallidolusyian atrophy (DRPLA).
[0068] In accordance with another aspect, methods and compositions of the invention are used for treating neurodegenerative disorders in which HDAC
iiihlbltlon is useful include nerve degeneration or nerve necrosis in disorders such as hypoxia, hypoglycemia, epilepsy, and in cases of central nervous system (CNS) trauma (such as spinal cord and head injury), hyperbaric oxygen convulsions and toxicity, dementia e.g. pre-senile dementia, and AIDS-related dementia, cachexia, Sydenham's chorea, Huntington's disease, Amyotrophic Lateral Sclerosis, Korsakoffs disease, imbecility relating to a cerebral vessel disorder, sleeping disorders, schizophrenia, depression, depression or other symptoins associated with Premenstrual Syndrome (PMS), anxiety and septic shock.
[0069] In accordance with another aspect, methods and compositions of the invention are used for treating bone diseases, including bone disorders involving osteoclasts and chonrocytes. Without wishing to be limited by this tlleory, it is noted that HDAC activity regulates the process of osteoclastogenesis and chondrocyte differentiation, such that inhibitors of HDAC are also useful in the treatment of all bone disorders involving osteoclasts and chondrocytes.

[0070] In accordance with another aspect, methods and coinpositions of the invention are used for treating ophthalmic diseases and other diseases in which angiogenesis plays a role in pathogeneis, such as glaucoma, retinal ganglion degeneration,occular ischemia, retinitis, retinopathies, uveitis, ocular photophobia, and of inflammation and pain associated with acute injury to the eye tissue.
[0071] Methods and compositions of the invention are used for treating human and non-huinan subjects. Methods and compositions of the invention are suitable for veterinary uses in treating companion animals, exotic animals and farm animals, including mammals, rodents, and the like. In particular embodiments, inethods and compositions of the invention are used for treating horses, dogs,and cats.
[0072] The terms "treat" or "treating" or "therapy" as used herein refer to (1) reducing the rate of progress of a disease, or, in case of cancer reducing the size of the tumor;
(2) inhibiting to some extent furtlier progress of the disease, which in case of cancer may mean slowing to some extent, or preferably stopping, tumor metastasis or tumor growth;
and/or, (3) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the disease. Thus, the term "therapeutically effective amount"
as used herein refers to that amount of the compound being administered which will provide therapy or affect treatment.
[0073J In accordance with certain aspects of the invention, the compounds of the present invention act as anti-tumor compounds and/or inhibit the growth of a tumor, i.e., they are tumor-growtll-iiiliibiting compounds. The tenns "anti-tumor" a.nd "tumor-growth-inhibiting," when modifying the term "coinpound," and the terms "inhibiting"
and "reducing", when modifying the terms "compound" and/or "tumor," mean that the presence of the subject compound is correlated with at least the slowing of the rate of growth of the tuinor. More preferably, the terms "anti-tumor," "tumor-growth-inhibiting,"
"inhibiting," and "reducing" refer to a correlation between the presence of the subject compound and at least the temporary cessation of tumor growtli. The terms "anti-tuinor," "tumor-growth-inhibiting," "inhibiting," and "reducing" also refer to, a correlation between the presence of the compound(s) of the invention and at least the temporary reduction in the mass of the tumor. It is understood that the effectiveness of compounds of the invention as anti-tumor, or tumor-inhibiting, agents may be contribute to their effectiveness in treating cancer, but that the compound of the invention may also act through other mechanisms to exert measured effects on cancer.
[0074] The term "cellular function" refers to the function of HDAC in the cell.
The term "HDAC function" is generally understood to refer to interaction of HDAC with a natural binding partner, and is particularly understood to refer to catalytic activity. The "cellular function" of HDAC is understood to refer not only to the catalyic activity of HDAC
in a cell, but also to the cellular effects of HDAC catalytic activity on the function of the cell.
The term "catalytic activity", in the context of the invention, defines the rate at wllich HDAC
deacetylates a substrate. Catalytic activity can be measured, for example, by determining the amount of a substrate converted to a product as a function of time.
Deacetylation of a substrate occurs at the active-site of HDAC. The active-site is normally a cavity in which the substrate binds to HDAC and is deacetylated.
[0075] The term "substrate" as used herein refers to a molecule deacetylated by HDAC. The substrate is preferably a peptide and more preferably a protein. In some embodiments, the protein is a histone, whereas in other embodiments, the protein is not a histone.

[0076] The term "inhibit" refers to decreasing the cellular function of HDAC.
It is understood that coarnpounds of the present invention may inhibit the cellular function of HDAC by various direct or indirect mechanisms, in particular by direct or indirect inhibition of the catalytic activityof HDAC. The term "activates" refers to increasing the cellular function of HDAC.

[0077] The tenn "modulates" refers to altering the function of HDAC by increasing or decreasing the probability that a complex forms between HDAC and a natural binding partner. A modulator may increase the probability that such a coinplex forms between HDAC and the natural binding partner, or may increase or decrease the probability that a complex forms between HDAC and the natural binding partner depending on the concentration of the compound exposed to HDAC, or may decrease the probability that a complex forms between HDAC and the natural binding partner. A modulator may activate the catalytic activity of HDAC, or may activate or inhibit the catalytic activity of HDAC
depending on the concentration of the compound exposed to HDAC, or may inhibit the catalytic activity of HDAC.
[0078] The term "complex" refers to an assembly of at least two molecules bound to one another. The term "natural binding partner" refers to polypeptides that bind to HDAC
in cells. A change in the interaction between HDAC and a natural binding partner can manifest itself as an increased or decreased probability that the interaction forms, or an increased or decreased concentration of HDAC/natural binding partner complex.

[0079] The term "contacting" as used herein refers to mixing a solution comprising a compound of the invention with a liquid medium bathing the cells of the methods. The solution comprising the compound may also comprise another component, such as dimethylsulfoxide (DMSO), which facilitates the uptake of the compound or compounds into the cells of the metllods. The solution comprising the compound of the invention may be added to the medium bathing the cells by utilizing a delivery apparatus, such as a pipet-based device or syringe-based device.
[0080] The term "monitoring" refers to observing the effect of adding the compound to the cells of the method. The effect can be manifested in a change in cell phenotype, cell proliferation, HDAC catalytic activity, substrate protein acetylation levels, gene expression changes, or in the interaction between HDAC and a natural binding partner.
[0081] The term "effect" describes a change or an absence of a change in cell phenotype or cell proliferation. "Effect" can also describe a change or an absence of a change in the catalytic activity of HDAC. "Effect" can also describe a change or an absence of a change in an interaction between HDAC and a natural binding partner.
[0082] The term "cell phenotype" refers to the outward appearance of a cell or tissue or the function of the cell or tissue. Examples of cell phenotype are cell size (reduction or enlargement), cell proliferation (increased or decreased numbers of cells), cell differentiation (a change or absence of a change in cell shape), cell survival, apoptosis (cell death), or the utilization of a metabolic nutrient (e.g., glucose uptake).
Changes or the absence of changes in cell phenotype are readily measured by techniques known in the art.
The present compounds are usefule for identifying a carbonyl coinpounds that modulates the cellular function of HDAC, comprising the steps of:

a) contacting cells expressing HDAC with the disclosed compounds of and b) measuring an effect of the coinpound or composition.

Optionally the effect is inhibition of the catalytic activity of HDAC. Further the inethods of coinprises measuring histone hyperacetylation. Additionally the effect is a change in cell phenotype and/or the effect is a change in cell proliferation.

Furtlier the disclosed compounds can be used for the manufacture of a medicainent for use in the treatment of a condition mediated by HDAC activity.

A. Pharmaceutical Compositions [0083] The present invention also relates to a pharmaceutical composition comprising a compound of the invention, or a pharmaceutically acceptable salt, solvate, amide, ester, or prodrug thereof, as described herein. and a pharmaceutically acceptable carrier, diluent, or excipient, or a combination thereof.
[0084] The term "pharmaceutical coinposition" refers to a mixture of a coinpound of the invention with other chemical components, such as carriers, diluents or excipients.
The pharmaceutical composition facilitates administration of the compound to an organism.
Multiple techniques of adininistering a compound exist in the art including, but not limited to: intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.

[0085] The term "carrier" refers to relatively nontoxic chemical compounds or agents. Such carriers may facilitate the incorporation of a coinpound into cells or tissues.
For exainple, hulnan serum albuinin (HSA) is a coinmonly utilized canier as it facilitates the uptake of many organic compounds into the cells or tissues of an organism.
[0086] The term "diluent" refers to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents can also be used to stabilize compounds because they can provide a more stable environment. Salts dissolved in buffered solutions (providing pH control) are utilized as diluents in the art. One commonly used buffered solution is phosphate buffered saline. It is a buffer found naturally in the blood systein, Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound.
[0087] The compounds described herein can be administered to a human patient per se, or in phartnaceutical coinpositions where they are mixed with other active ingredients, as in coinbination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences," 20th ed. Edited by Alfonso Gennaro, 2000.
1) Routes Of Administration [0088] Suitable routes of administration include local or systemic routes of administration including, but not limited to, topical, transdermal, oral, rectal, transinucosal, pulmonary, ophthalmic, intestinal, parenteral, intramuscular, subcutaneous, intravenous, intramedullary, intrathecal, direct intraventricular, iiitraperitoneal, intranasal, or intraocular delivery. In certain embodiments, compounds of the invention are adininistered topically, e,g in an ointment, patch, nasal spray, or eye drops/ointinent. In certain embodiments, compounds of the invention are delivered by intestinal, parenteral, intramuscular, subcutaneous, intravenous, intramedullary, intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
[0089] Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot or sustained release formulation. Furthennore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. The liposomes will be targeted to and taken up selectively by the organ.
2) Composition/Formulation [0090] The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
[0091] Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Phannaceutical Sciences, above.
[0092] For intravenous injections, the agents of the invention may be formulated in aqueous solutions, preferably in phannaceutically compatible buffers such as Hanlcs's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be perineated are used in the forinulation. Such penetrants are generally known in the art. For other parenteral injections, the agents of the invention may be fomlulated in aqueous or nonaqueous solutions, preferably with pharmaceutically compatible buffers or excipients. Such excipients are generally known in the art.

[0093] For oral administration, the compounds can be formulated readily by combining the active compounds with pharinaceutically acceptable carriers or excipients well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with one or more compound of the invention, optionally grinding the resulting mixture, and processing the niixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for exainple, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethyl cellulose; or others such as:
polyvinylpyrrolidone (PVP or povidone) or calciurn phosphate. If desired, disintegrating agents may be added, such as the cross-linked croscannellose sodium, polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[0094] Dragee cores are provided witll suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or piginents may be added to the tablets or dragee coatings for identification or to characterize different combinations of active coinpound doses.
[0095] Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All forinulations for oral adininistration should be in dosages suitable for such administration.
[0096] For buccal or sublingual administration, the coinpositions inay take the form of tablets, lozenges, or gels forinulated in conventional manner.
[0097] For administration by inhalation, the coinpounds for use according to the present invention are conveniently delivered in the fonn of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be fonnulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0098] The compounds may be forinulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage fonn, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such fonns as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain fornnulatory agents such as suspending, stabilizing and/or dispersing agents.

[0099] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble forni. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxyinethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.

[00100] Alternatively, the active ingredient niay be in powder form for constitution wit11 a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[00101] The compounds may also be formulated in rectal coinpositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.

[00102] In addition to the formulations described previously, the coinpounds may also be formulated as a depot preparation. Such long acting forinulations may be administered by iinplantation (for example subcutaneously or intrainuscularly) or by intrainuscular injection. Thus, for example, the coinpounds may be formulated with suitable polylneric or hydrophobic materials (for example as an einulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

[00103] A pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent systein may be a 10%
ethanol, 10%
polyethylene glycol 300, 10% polyethylene glycol 40 castor oil (PEG-40 castor oil) with 70%
aqueous solution. This cosolvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systeinic administration. Naturally, the proportions of a cosolvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the cosolvent components may be varied: for exainple, other low-toxicity nonpolar surfactants may be used instead of PEG-40 castor oil, the fraction size of polyethylene glycol 300 may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides maybe included in the aqueous solution.
[00104] Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as N-inethylpyrrolidone also may be employed, although usually at the cost of greater toxicity.
Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
[00105] Many of the compounds of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not liinited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free acid or base forms.

GENERAL SYNTHETIC METHODS FOR PREPARING COMPOUNDS
Molecular einbodiments of the present invention can be synthesized using standard synthetic techniques known to those of skill in the art. Compounds of the present invention can be synthesized using the general synthetic procedures set forth in Scheines I-III.

General procedure for sulfonamide (a): Scheme I

Q a ~~~~ Q
-Ir R35HN R45 0 R31 / S\ I NQ O

SI-I

b ~S\N a S~R3z ~ c Y.O~Q)r-~.-Br R31 ~ 1 ~ 0 R31 R

Reagents: (a) R31SO2CI, Et3N, THF; (b) PTT, THF; (c) KSCOR32, MeOH

General procedure for sulfonamide (b): Scheme II
Q
Rg4SO2Cl + \ ~ a R34/S\N / R~
\ ~ O
0 i b nN~ S O~~ / ~Br O
0 ( x- c /S\ \ (a R34'~ \N \ E Rs4 i ~ R35 d O
O O / ~ R3z \'e~ 0-'0y-SH e R jS~N 34 O R
I

Reagents: (a) pyridine, THF; (b) PTT, THF; (c) N-methyl 2-thiopyridone, EtOH;
(d) NaOH, water; (e) acid chloride, hunig's base, DCM

General Procedure for reverse sulfonamide :

Scheme III

a R34R33N\S ~ I Q
CI~S 0 SIII-1 O~ \O
b 0 S~R32 c Br y R34R33N YQ

\ \ \ \

$f li-3 SI I I-2 Reagents: (a) Amine (R34R33N), THF, pyr; (b) THF, PTT; (c) MeOH, KSC(O)R32 General Procedure for Ring-Fused Sulfonamides Scheme IV

pO 0 :Cr S"CI a N ~ S~ O

H
b O C
SY Br N 0 c SO f / \ O
(a) Pyridine, THF; (b) PTT, THF; (c) KSC(O)CH3, MeOH

Scheme V illustrates the general syntliesis of disulfide embodiments of the present invention.
Scheme V

Rt O R1 O

Rz /~ Br a RZ / S11 --~ I
Rs \ Re R3 ~ R 0 Ra R4 b Rl O
RZ ~ S
R3 ~ I R5 Ry Reagents: (a) KSC(O)CH3, MeOH; (b) NaOH, MeOH

Scheme VI depicts an alternative general scheme for the synthesis of thiol (mercaptan) and disulfide embodiments of the present invention Scheme VI

Ri 0 R, O
R2 a Rp / Br R3 I RS R. ~ I Rs Ry R2 = ~b ::i'ii"- c Rq d Ri 0 R3 ~ I R5 Rq Reagents: (a) PTT, THF; (b) N-methyl 2-thiopyridone, EtOH;
(c) NaOH, water; (d) MeOH, water.

EXAMPLES
[00106] The examples below are non-limiting and are merely representative of various aspects of the invention.
Example 1 o / S
\ ~s/~N \ ~ y F, CFsO I

Thiobenzoic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: The compound tlliobenzoic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl}ester was synthesized according to Scheme I.
Step 1 \ ii S"N
\ H
I /

N-(4-Acetyl-phenyl)-4-trifluoromethoxy-benzenesulfonamide (SI-2): 4'-Amino acetophenone (0.375 g, 2.78 mmol) was dissolved in THF (5 ml) before pyridine (0.674 ml, 8.34 minol) was added, leaving a yellow solution. 4-trifluorometlioxy benzenesulfonylchloride (0.871 g, 3.34 minol) was then added dropwise with stirring. After strirring for 2h, THF and pyridine were removed. The desired sulfonamide (0.848 g, 2.36 minol, 85 %) was recrystallized from ethyl acetate and hexanes. 'H-NMR: (400 MHz, CDC13) 7.89 (m, 4H), 7.29 (d, IH), 7.16 (d, 2H), 6.88 (s, 1H), 2.55 (s, 3H).
LC-MS (ES+):
360 [MH]+ in/e.
Step 2 O
Br \ N
~
/ H

N-[4-(2-Bromo-acetyl)-phenyl]-4-trifluoromethoxy-benzenesulfonamide (SI-3):
The ketone from stepl (0.32 g, 0.868 mmol) was dissolved in THF (9 ml), and phenyltrilnethylammonium tribromide (PTT) (0.368 g, 0.868 mmol) was added as a solid leaving an orange solution which began to deposit a white solid immediately.
Stirring for 1.5 hours leaves a colorless mixture to which water (5 ml) was added. THF was then evaporated and the resulting aqueous mixture was extracted with ethyl acetate. Drying over Na2SO4 and evaporation leaves a white crystalline solid (90% desired mono-brominated material by LC-MS, 5% starting inaterial, 5 % dibrominated) suitable for the next step. LC-MS
(ES-): 436, 43 8 m/e.
Step 3 o I
s \

v ~ \ \S~N 0 ~ H

Thiobenzoic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: oc-Bromoketone (SI-3) (0.3 g, 0.68 mmol) was dissolved in anliydrous THF (5 ml), and thiobenzoic acid (104 mg, 0.75 mmol) was added followed inunediately by potassium carbonate (104 rng, 0.75 mmol). The reaction was stirred for 2 hours. The THF
was evaporated and the residue was taken up in DCM and filtered. After evaporation of the DCM the product was purified by flash chromatography (7:3 EtOAC: hexanes).
Removal of the solvent afforded 57 mg product (0.11 mmols, 16%) as a white solid. 'H-NMR:
(400 MHz, DMSO): 8.00 (t, 3H), 7.98 (d, 2H), 7.61 (t, 2H), 7.50 (t, 2H), 7.32 (d, 2H), 7.22 (d, 2H), 4.52 (s, 2H); LC-MS (ES+): 496 [MH] + m/e.

Example 2 / S \ N
o Y
~N \ 0 I \
/ y Thionicotinic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: Exainple 2 was synfihesized according to the procedure outlined in Example 1.
'H-NMR: (400 MHz, CDC13): 9.11 (s, 1H), 8.86 (d, 1H), 8.30 (d, 1H), 8,00 (d, 2H), 7.98 (d, 2H), 7.44 (t, 1H), 7.33 (d, 2H), 7.28 (d, 2H), 4.60 (s, 2H); LC-MS (ES+): 498 m/e.

Example 3 \V' lol / H
CF3O ~ \

Pentanethioic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: Thioacetic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylainino)-phenyl]-ethyl} ester (SI-4) (0.5 g, 1.15 mmol) was dissolved in anhydrous DMF
(5 ml).
Lithium hydroxide (28 mg, 1.15 mmol) was added and the solution was sonicated until homogeneous and heated to 90 C for 4 hours, The solution was allowed to cool to room temperature and valeryl chloride (150 ing, 1.27 mmol) was added and the mixture stirred for 16 hours. The solution was partioned between 20 ml each water and EtOAc. The organic layer was washed with water, then brine and dried over Na2SO4. The solution was then filtered, stripped of solevent and the product purified by flash chromatography (100% DCM), The solvent was evaporated to afford 107 mg (0.22 mmol, 19%) product as a clear oil, 1H-NMR: (400 MHz, DMSO): 8.07 (d, 2H), 7.60 (d, 2H), 7.40 (d, 2H), 7.25 (d, 2H), 2.40 (s, 2H), 2.10 (t, 2H), 1.42 (t, 2H) 1,11 (t, 2H), 0.90 (t, 3H); LC-MS (ES+): 476 [M]+m/e.
Example 4 QLSO H

Furan-2-carbothioic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: The compound furan-2-carbothioic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester was synthesized according to Scheme II.
Step 1 \/
S~N
\ H
I

N-(4-Acetyl-phenyl)-4-trifluoromethoxy-benzenesulfonamide (SII-1): 4'-Amino acetophenone (0.375 g, 2.78 inmol) was dissolved in THF (5 ml) before pyridine (0.674 ml, 8.34 mmol) was added, leaving a yellow solution. 4-trifluoromethoxy benzenesulfonylehloride (0.871 g, 3.34 mmol) was then added dropwise with stirring. After removal of THF and pyridine, the desired sulfonamide (0.848 g, 2.36 mmol, 85 %) was recrystallized from ethyl acetate and hexanes. 1H-NMR: (400 MHz, CDCl3) 7.89 (m, 4H), 7.29 (d, 1H), 7.16 (d, 2H), 6.88 (s, 1H), 2.55 (s, 3H). LC-MS (ES+): 360 [MH]+m/e.

Sten 2 Br 0i I
S" N
I~ y N-[4-(2-Bromo-acetyl)-phenyl]-4-trifluoromethoxy-benzenesulfonamide (SII-2):
The ketone from step 1(0.32 g, 0.868 nunol) was dissolved in THF (9 ml), and phenyltrimethylainmonium tribroinide (PTT) (0.368 g, 0.868 mmol) was added as a solid leaving an orange solution which began to deposit a white solid iinmediately.
Stirring for 1.5 hours leaves a colorless mixture to which water (5 ml) was added. THF was then evaporated and the resulting aqueous mixture was extracted with ethyl acetate. Drying over Na2SO4 and evaporation leaves a white crystalline solid (90% desired mono-brominated material by LC-MS, 5% starting material, 5 % dibrominated) suitable for the next step. LC-MS
(ES-): 436, 43 8 m/e.

Step 3 / s\ ~
N ~N'v/'1J
\ \S/~ H
I/ X' 1-Methyl-2-{2-oxo-2-[4-(4-trifluoromethoxybenzene- sulphonylamino) phenyl]ethylsulfanyl}-pyridinium bromide (SII-3): The bromoketone from step 2 (0.141 g crude material, 0.322 mmol) was dissolved in ethanol (2 ml) before N-methyl thiopyridone (0.040 g, 0.322 mmol) was added as a solid. The resulting yellow solution was then heated to reflux overnight. Evaporation of the volatiles leaves a residue (75 % by NMR, 0.116 g, 0.240 mmol) suitable for the next step, however, the product may be recrystallized from ethanol if desired. 'H-NMR: (400 MHz, DMSO-d6) 11.21 (s, 1H), 8.90 (d, IH), 8.18 (t, 1H), 8.03 (m, 5H), 7.90 (t, 1H), 7.80 (d, 2H), 7.15 (d, 2H), 5.33 (s, 2H), 4.24 (s, 3H). LC-MS (ES+): 483 [M]+ m/e.
Step 4 SH
\V/

I / H
S\N

N-[4-(2-Mercapto-acetyl)-phenyl]-4-trifluoromethoxy-benzenesulfonamide (SII-4):

SII-3 (4.35 g, 7,72 mmol) was suspended in water (1.7 1) before 2 M NaOH (7.25 ml) was added. Solid NaOH (1 g) was then added, and the resulting mixture was then heated to reflux overnight, producing a red solution. The solution was then acidified to a pH
of 1 and extracted with ethyl acetate. Drying over Na2SO4 and evaporation leaves a red oil.
Throughout the work-up, the alpha-mercapto ketone readily oxidizes to the coiresponding disulfide, which was purified by preparative HPLC (0.582 g, 0.75 mmol, 10 %).
1H-NMR:
(400 MHz, DMSO-d6) 11.09 (bs, 2H), 7.97 (d, 4H), 7.85 (d, 4H), 7.57 (d, 4H), 7.22 (d, 4H), 4.29 (s, 4H). LC-MS (ES+): 781 [MH]+ m/e.
Step 5 I'~ y ~
H

Furan-2-carbothioic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: To thiol SII-4 (0.5 g, 1.33 mmol) in 5 ml degassed anhydrous DCM (5 ml) was added 2-furoyl chloride (190 mg, 1.46 mmol) followed by DIEA (0.19 g, 1.46 mmol). The reaction was stirred for 4 hours and the solvent was evaporated.
The product was first purified by flash chromatography (4:6 EtOAc:hexanes), then recrystallized from EtOAc and finally triturated with DCM to afford 31 mg (0.064 mmol, 5%) product as a white solid. 'H-NMR: (400 MHz, CDC13): 8.00 (d, 2H), 7.94 (d, 2H), 7.64 (d, 1H), 7.35 (d, 2H), 7.29 (d, 1H), 7.22 (d, 2H), 6.60 (t, 1H), 4.50 (s, 2H); LC-MS (ES+): 486 [MH]+
m/e.
Example 5 o S

\ \8~N I 0 I / H

1-(4-Diethylamino-phenyl)-2-mercapto-ethanone was syntllesized according to the procedure described in the preparation of Example 4. 'H-NMR: (400 MHz, CDC13):
7.96 (d, 2H), 7.93 (d, 2H), 7.35 (d, 2H), 7.21 (d, 2H), 4.27 (s, 2H); LC-MS (ES+): 476 [MH]+ m/e, Example 6 s \
I \ S'T 0 0Y
/
/ H o Acetic acid 2-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethylsulfanylcarbonyl}-phenyl ester was synthesized according to the procedure described in the preparation of Example 4. 'H-NMR: (400 MHz, CDC13): 7.99 (d, 2H), 7.95 (d, 2H), 7.60 (t, IH), 7.30 (d, 2H), 7.12 (d, 2H), 7.09 (d, 2H), 4.47 (s, 2H), 2.37 (s, 3H); LC-MS
(ES+): 555 [MH] + m/e.

Example 7 O / ~ N\
4I~ ~ SN O

CF30 I~

4-Dimethylamino-thiobenzoic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl ester was synthesized according to the procedure described in the preparation of Example 4. 1H-NMR: (400 MHz, DMSO): 11.10 (s, 1H), 8.01 (d, 2H), 7.99 (d, 2H), 7.78 (d, 2H), 7.60 (d, 2H), 7.28 (d, 2H), 6.78 (d, 2H), 4.35 (s, 2H), 3.02 (s, 6H); LC-MS (ES+): 539 [MH]+ mle.

Example 8 sYo"
~S"N 0 Thiocarbonic acid 0-methyl ester S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester was synthesized according to the procedure described in the preparation of Example 4. 'H-NMR: (400 MHz, CDC13): 7.96 (d, 2H), 7,92 (d, 2H), 7.38 (d, 2H), 7.21 (d, 2H), 4.37 (s, 2H), 3.83 (s, 3H); LC-MS (ES+):
448 [MH]+ in/e.
Example 9 /I S ~N
\ \S/\N \
I H
CF3O" v Pyridine-2-carbothioic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: Picolinic acid (0.25 g, 2.04 mmol) was dissolved in anhydrous, degassed DCM (5 ml), HATU (775 mg, 2.04 mmol) was added followed by DIEA (0.53 g, 4.1 mmol). The mixture was stirred for 20 min and N-[4-(2-Mercapto-acetyl)-phenyl]-4-trifluoroinethoxy-benzenesulfonamide SII-4 (0.725 g, 1.85 mmol) was added in 5 ml anhydrous, degassed DCM and the mixture was stirred for 16 hours. The reaction was filtered and the solvent evaporated. The product was purified by flash chromatography (1:1 EtOAc:hexanes) to afford 125 mg (0.25 mmol, 14%) product as a white solid. 'H-NMR:
(400 MHz, DMSO): 11.12 (s, 1H), 8.89 (d, 1H), 8.03 (t, 1H), 8.01 (d, 2H), 8.00 (d, 2H), 7.90 (d, 1H), 7.78 (t, 1H), 7.60 (d, 2H), 7.14 (d, 2H), 4.60 (s, 2H); LC-MS (ES+):
497 [MH]} in/e.
Example 10 H
~S\N \ 0 I / H

Tert-Butoxycarbonylamino-thioacetic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester: The compound tert-Butoxycarbonylamino-thioacetic acid S- {2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylainino)-phenyl]-ethyl}ester was synthesized according to the procedures described in the preparation of Example 9. 1H-NMR: (400 MHz, CDC13): 7.94 (d, 2H), 7.88 (d, 2H), 7.31 (d, 2H), 7.21 (d, 2H), 5.24 (bs, 1H), 4.36 (s, 2H), 4.14 (s, 2H), 1.48 (s, 9H); LC-MS (ES-): 448 [MH]" - Boc m/e (major fragment, no parent ion observed).

Example 11 / H

Amino-thioacetic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylaniino)-phenyl]-ethyl} ester: N-Boc thioester from Example 12 (110 mg, 0.2 mmol) was dissolved in anhydrous DCM (1 ml) and TFA (1 ml) was added. The mixture was stirred for minutes and the volatiles removed in vacuo. Recrystallization from EtOAc-hexanes afforded 70 mg product (0.15 mmol, 75%) as an off white solid. 'H-NMR: (400 MHz, DMSO):
11.08 (bs, 1H), 8.00 (d, 2H), 7.99 (d, 2H), 7.60 (d, 2H), 7.14 (d, 2H), 4.61 (s, 2H), 4.21 (s, 2H);
LC-MS (ES+): 449 [MH]+ m/e.

Example 12 s~
\ \S\N ! / lOl Thioacetic acid S-(2-{4-[methyl-(4-trifluoromethoxy-benzenesulfonyl)-amino]phenyl}-2-oxo-ethyl) ester: The compound thioacetic acid S-(2-{4-[methyl-(4-trifluoromethoxy-benzenesulfonyl)-amino]phenyl}-2-oxo-ethyl) ester was synthesized according to the procedures described in the preparation of Example 1. 'H NMR: (400MHz, DMSO-d6) cS
8.00 (d, 2H), 7.63 (d, 2H), 7.60 (d, 2H), 7.38 (d, 2H), 4.50 (s, 2H), 3.10 (s, 3H), 2.40 (s, 3H);
LCMS (ES+): 448 [M]+ m/e.

Example 13 S/-~N 0 CF3 1O I / "L"

Thioacetic acid S-(2-{4-[isopropyl-(4-trifluoromethoxy-benzenesulfonyl)-amino]-phenyl}-2-oxo-ethyl) ester: The compound thioacetic acid S-(2- {4- [isopropyl-(4-trifluoromethoxy-benzenesulfonyl)-alnino]-phenyl}-2-oxo-ethyl) ester was synthesized according to the procedures described in the preparation of Exainple 1. 1H
NMR: (400MHz, DMSO-d6) b 8.02 (d, 2H), 7.90 (d, 2H), 7.60 (d, 2H), 7.22 (d, 2H), 4.56 (s, 2H), 4.43 (m, 1H), 2.40 (s, 3H), 1.00 (d, 6H); LCMS (ES+): 476 [M]" m/e.

Example 14 \s/\ lol CF3~ - I / H

Thioacetic acid S-{2-[2-chloro-4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester: The compound thioacetic acid S-{2-[2-chloro-4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Exainple 1. 1H NMR: (400MHz, DMSO-d6) 611.10 (s, 1 H), 8.00 (d, 2H), 7.99 (d, 1 H), 7.60 (s, 1 H), 7.10 (d, 1 H), 4.3 5(s, 2H), 2.3 6(s, 3H); LCMS (ES+): 467 [M]+ in/e.

Example 15 s\ f \ S~\N ~o( CF3~ ~ / H F
O
Thioacetic acid S-{2-[3-fluoro-4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester: The compound thioacetic acid S-{2-[3-fluoro-4-(4-trifluoroinethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 'H NMR: (400MHz, DMSO-d6) 8 10.94 (s, 1H), 7.98 (d, 2H), 7.81 (d, 2H), 7.60 (d, 2H), 7.49 (t, 1H), 4,42 (s, 2H), 2.39 (s, 3H);
LCMS (ES+): 452 [M] + m/e.

Example 16 oso \ I / s~

H

Thioacetic acid S-{2-oxo-2-[4-(toluene-4-sulfonylamino)-phenyl]-ethyl} ester:
The compound thioacetic acid S-{2-oxo-2-[4-(toluene-4-sulfonylamino)-phenyl]-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR
(DMSO): 10.95 (s, 1H), 7.90 (d, 2H), 7.74 (d, 2H), 7.40 (d, 2H), 7.22 (d, 2H), 4.42 (s, 2H), 2.47 (s, 3H), 2.37 (s, 3H); LC-MS (ES+): 364 [MH]+ m/e.

Example 17 p/o~ s F3C \IS~N ~ I 0 H
Thioacetic acid S-{2-oxo-2-[4-(3-trifluoromethyl-benzenesulfonylamino)-phenyl]-ethyl}
ester: The compound thioacetic acid S-{2-oxo-2-[4-(3-trifluorornethyl-benzenesulfonylamino)-phenyl]-ethyl} ester was syntllesized according to the procedures described in the preparation of Exainple 1. 'H-NMR (DMSO): 11.12 (s, 1H), 8.10 (m, 3H), 7.88 (m, 3H), 7.26 (d, 2H), 4.43 (s, 2H), 2.37 (s, 3H); LC-MS (ES+): 418 [MH]+
in/e.

Example 18 S~l o N ol H
F
Thioacetic acid S-{2-[4-(4-fluoro-2-methyl-benzenesulfonylamino)-phenyl]-2-oxo-ethyl}
ester: The compound thioacetic acid S-{2-[4-(4-fluoro-2--methyl-enzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR (DMSO): 11.16 (s, 1H), 8.08 (t, 1H), 7.90 (d, 2H), 7.31 (m, 2H), 7.19 (d, 2H), 4.42 (s, 2H), 2.62 (s, 3H), 2.37 (s, 3H); LC-MS (ES+):
382 [MH]+
m/e.

Example 19 0,I~
SrJ 0 H
\O I /

Thioacetic acid S-{2-[4-(4-methoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl}
ester:
The compound thioacetic acid S-{2-[4-(4-methoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR (DMSO): 10.86 (s, 1H), 7.90 (d, 2H), 7.79 (d, 2H), 7.23 (d, 2H), 7.11 (d, 2H), 4.42 (s, 2H), 3.83 (s, 3H), 2.37 (s, 3H); LC-MS (ES+): 380 [MH] +
in/e.

Example 20 \ "r oSO s N
H
GI

Thioacetic acid S-{2-[4-(4-chloro-3-trifluoromethyl-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester: The compound thioacetic acid S-{2-[4-(4-chloro-3-trifluoromethyl-benzenesulfonylarnino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. iH-NMR (DMSO):l 1.16 (s, 1H), 8.20 (s, 1H), 8.12 (t, 1H), 7.98 (m, 3H), 7.27 (d, 2H), 4.44 (s, 2H), 2.37 (s, 3H);
LC-MS (ES+):
452 [MH]+ m/e.

Example 21 o~~O
s ~N 0 y ' / H

/O

Thioacetic acid S-{2-[4-(3-methoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl}
ester:
The compound thioacetic acid S-{2-[4-(3-methoxy-benzenesulfonylainino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. IH-NMR (DMSO): 10.95 (s, 1H), 7.92 (d, 2H), 7.49 (t, 1H), 7.41 (d, 1H), 7.34 (s, 1H), 7.24 (m, 3H), 4.43 (s, 2H), 3.80 (s, 3H), 2.37 (s, 3H); LC-MS (ES+):
380 [MH]+ m/e.
Example 22 slir oNo , 0 N
H
Thioacetic acid S-[2-(4-ethanesulfonylamino-phenyl)-2-oxo-ethyl] ester: The compound thioacetic acid S-[2-(4-ethanesulfonylamino-phenyl)-2-oxo-ethyl] ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR
(DMSO):
10.91 (s, 1H), 8.00 (d, 2H), 7.30 (d, 2H), 4.47 (d, 2H), 3.23 (q, 2H), 2.39 (s, 3H), 1.21 (t, 3H);
LC-MS (ES+): 302 [MH]+ m/e.

Example 23 ~ / s~
~ \ ~ N 0 H
Thioacetic acid 2-[4-(3,5-dimethyl-benzenesulfonylamino)-phenyl)-2-oxo-ethyl ester:
The compound thioacetic acid 2-[4-(3,5-dimethyl-benzenesulfonylamino)-phenyl]-2-oxo-ethyl ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR (DMSO): 10.91 (s, 1H), 7.91 (d, 2H), 7.49 (s, 2H), 7.24 (d, 3H), 4.43 (s, 2H), 2.37 (s, 3H), 2.33 (s, 6H); LC-MS (ES+): 378 [MH]} in/e.

Example 24 s~
~~ N o ), H
I
Thioacetic acid S-{2-[4-(1,2-dimethyl-lH-imidazole-4-sulfonylamino)-phenyl]-2-oxo-ethyl} ester: The compound thioacetic acid S-{2-[4-(1,2-diinethyl-lH-imidazole-sulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. iH-NMR (DMSO): 10.92 (s, 1H), 7.91 (d, 2H), 7.27 (d, 2H), 4.43 (s, 2H), 3.57 (s, 3H), 2.37 (s, 3H), 2.26, (s, 3H); LC-MS
(ES+): 368 [MH]+
m/e.

Example 25 , s ~
~s~ No H
/
Thioacetic acid S-{2-oxo-2-[4-(propane-2-sulfonylamino)-phenyl]-ethyl} ester:
The compound thioacetic acid S-{2-oxo-2-[4-(propane-2-sulfonylamino)-phenyl]-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR
(DMSO): 10.38 (s, 1H), 8.00 (d, 2H), 7.36 (d, 2H), 4.47 (s, 2H), 2.39 (s, 3H), 1.25 (s, 7H);
LC-MS (ES+): 316 [MH]} m/e.

Example 26 slir O\ N 0 S' 'H
\0 CI qCI

N-[4-(2-Mercapto-acetyl)-phenyl]-3,4-Dichlorobenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-3,4-Dichlorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Exainple 1. iH-NMR: (CDC13) 7.99(s, 1H), 7.92(d, 2H), 7.68(d, 1H), 7.56(d, IH), 4.34(s, 2H), 2.42(s, 3H); MS: (418.6) Example 27 SITr ~'iN

H
O1~

N-[4-(2-Mercapto-acetyl)-phenyl]-3,4-bis-trifluoromethoxy-benzenesulfonamide The coinpound N-[4-(2-Mercapto-acetyl)-phenyl]-3,4-bis-trifluoromethoxy-benzenesulfonamide was synthesized according to the procedures described in the preparation of Exaniple 1. 1H-NMR: (CDC13) 7.92(d, 2H), 7.51(d, 1 H), 7.31(s, 1 H), 7.21(d, 2H), 6.91(d, IH), 4.38(s, 2H), 2.42(s, 3H); MS: (409,8) Example 28 s"Ir o\ N \ ~ 0 I\ SoH
ci ~ ci N-[4-(2-Mercapto-acetyl)-phenyl]-2,4-Dichlorobenzene-sulfonamide The compound [4-(2-Mercapto-acetyl)-phenyl]-2,4-Dichlorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. iH-NMR: (CDC13) 8.08(d, 1H), 7.91(d, 2H), 7.58(s, 1H), 7.41(d, 1H), 7.21(d, 2H), 4.32(s, 2H), 2.42(s, 3H);
MS: (418.6) Example 29 sY

SN~~ O
O
N-[4-(2-Mercapto-acetyl)-phenyl]-2-(3,5-Dimethylisoxazole)-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-2-(3,5-Dimethylisoxazole)-sulfonainide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR:
(CDC13) 8.01(d, 2H), 7.21(d, 2H), 4.39(s, 2H), 2.63(s, 3H), 2.24(s, 3H), 2.20(s, 3H);
MS: (368.4) Example 30 s o N IQI
S S;
---X O

N-[4-(2-Mercapto-acetyl)-phenyl]-5-(2,4-Dimethyl-1,3-Thiazole)-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-5-(2,4-Dimethyl-1,3-Thiazole)-sulfonamide was synthesized according to the procedures described in the preparation of Exainple 1. IH-NMR: (CDC13) 7.98(d, 2H), 7.24(d, 2H), 4.39(s, 2H), 2.68(s, 3H), 2.59(s, 3H), 2.42(s, 3H);
MS: (384.4) Example 31 sy 0, N ! / 0 N-[4-(2-Mercapto-acetyl)-phenyl]-3-(2,5-Dimethylfuran)-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-3-(2,5-Dimethylfuran)-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR:
(CDC13) 7.98(d, 2H), 7.41(s, 1H), 7.21(d, 2H), 4.39(s, 2H), 2.52(s, 3H), 2.44(s, 3H), 2.21(s, 3H); MS:
(368.6) Example 32 o N ~ / 0 s FF
F
N-[4-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethylbenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethylbenzene-sulfonamide was synthesized according to the procedures described in the preparation of Exainple 1. 'H-NMR: (CDC13) 8.21(d, 2H), 7.98(d, 2H), 7.78(d, 2H), 7.22(d, 2H), 4.39(s, 2H), 2.44(s, 3H);
MS: (418.5) Example 33 OS'N \ t O
I \ O

N-[4-(2-Mercapto-acetyl)-phenyl]-4-benzene-sulfonamide The coznpound N-[4-(2-Mercapto-acetyl)-phenyl]-4-benzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR: (CDC13) 7.88(d, 3H), 7.59(d, 2H), 7.47(d, 2H), 7.34(d, 2H), 4.29(s, 2H), 2.41(s, 3H); MS: (348.6) Example 34 o 0 s'N

F
N-[4-(2-Mercapto-acetyl)-phenyl]-4-Fluorobenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-4-Fluorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Exalnple 1. 1H-NMR: (CDC13) 7.91(d, 2H), 7.89(d, 2H), 7.22(d, 2H), 7.18(d, 2H), 4,38(s, (2H), 2.42(s, 3H); MS: (367.6) Example 35 s OSN \ !
\
I ~ o ~ F
N-[4-(2-Mercapto-acetyl)-phenylJ-2-Fluorobenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-2-Fluorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR: (CDC13) 7.98(d, 1H), 7.89(d, 2H), 7.61(t, 1H), 7.24(d, 2H), 7.22(d, 2H), 4.37(s, 2H), 2.41(s, 3H);
MS: (367.6) Example 36 s"r p 0 ' N \ I
s I ~ o Ci N-[4-(2-Mercapto-acetyl)-phenyl]-4-Chlorobenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-4-Chlorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR: (CDC13) 7,85(d, 2H), 7.78(d, 2H), 7.60(d, 2H), 7.40(d, 2H), 4.28(s, 2H), 2.41(s, 3H); MS: (384.1) Example 37 slir O O
\N pso CI
N-[4-(2-Mercapto-acetyl)-phenyl]-3-Chlorobenzene-sulfonamide The coinpound N-[4-(2-Mercapto-acetyl)-phenyl]-3-Chlorobenzene-sulfonaznide was synthesized according to the procedures described in the preparation of Example 1. IH-NMR: (CDC13) 7.98(d, 2H), 7.90(s, 1 H), 7.78(d, 1 H), 7.60(d, 114), 7.46(t, 1 H), 7.21(d, 2H), 4.38(s, 2H), 2.41(s, 3H); MS:
(384.1) Example 38 s"r cC0 N-[4-(2-Mercapto-acetyl)-phenyl]-2-Chlorobenzene-sulfonamide The compound N-[4-(2-Mercapto-acetyl)-phenyl]-2-Chlorobenzene-sulfonamide was synthesized according to the procedures described in the preparation of Exainple 1. 1H-NMR: (CDC13) 8.18(d, 1H), 7.90(d, 2H), 7.58(d, 2H), 7.43(d, 1H), 7.22(d, 2H), 4.38(s, 2H), 2.42(s, 3H);
MS: (384.1) Example 39 os N g/\

F \~O
]F' N-[3-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethoxybenzene-sulfonamide The compound N-[3-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethoxybenzene-sulfonainide was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR: (CDC13) 7.90(d, 2H), 7.82(d, 1H), 7.65(s, 1H), 7.44(d, 2H), 7.37(d, 1H), 7.24(d, 1H), 4.39(s, 2H), 2.43(s, 3H); MS: (433.2) Example 40 0 ,N ~
~ S~ O
FF O I /

t F O~

N-[2-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethoxybenzene-sulfonamide The compound N-[2-(2-Mercapto-acetyl)-phenyl]-4-Trifluoromethoxybenzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR: (CDC13) 7.91(d, 2H), 7.82(d, 1H), 7.79(t, 1H), 7.58(dd, 1H), 7.24(d, 2H), 7,19(d, 1H), 4.39(s, 2H), 2.42(s, 3H); MS: (433.2) Example 41 sY
OS'N 0 O

O
F
F F
1V-[4-(2-Mercpto-acetyl)-phenylJ-3-Trifluoromethoxybenzene-sulfonamide The compound .N-[4-(2-Mercpto-acetyl)-phenyl]-3-Trifluoromethoxybenzene-sulfonamide was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR: (CDC13) 7.96(d, 2H), 7.81(d, 1 H), 7.72(s, 1 H), 7.59(dd, 1H), 7.43(d, 1 H), 7.21(d, 2H), 4.38(s, 2H), 2.41(s, 3H); MS: (433.2) Example 42 s4 ~
s, ~~N N
F ~ S
F~O I / O
F

Thioacetic acid-S-{2-[2-(3,4-Di-Trifluoromethoxy-benzenesulfonylamino)-4-methyl-thiazol-5-yl]-2-oxo-ethyl}-ester The compound thioacetic acid-S-{2-[2-(3,4-Di-Trifluoromethoxy-benzenesulfonylamino)-4-methyl-thiazol-5-yl]-2-oxo-ethyl}-ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR: (CDC13) 8.01(d, 2H), 7.25(d, 2H), 2.60(s, 3H), 2.41(s, 3H); MS: (454.8) Example 43 s ~
Q~N N
~ S
~
CI ~
CI
Thioacetic acid-S-{2-[2-(3,4-Dichlorobenzenesulfonylamino)- 4-methyl-thiazol-5-yl]-2-oxo-ethyl}-ester The compound thioacetic acid-S-{2-[2-(3,4-Dichlorobenzenesulfonylamino)- 4-methyl-thiazol-5-yl]-2-oxo-ethyl}-ester was synthesized according to the procedures described in the preparation of Example 1. 'H-NMR:
(CDC13) 8.00(s, 1H), 7.79(d, 1H), 7.49(d, 1H), 4.02(s, 2H), 2.58(s, 3H), 2.41(s, 3H);
MS: (439.6) Example 45 i S 11 CF3'O O
p a~:"
N\S
Thioacetic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-phenylsulfamoyl)-phenyl]-ethyl}
ester:
Step 1 H I \

// \\
~\ /
CF311 o 0 0 4-Acetyl-N-(4-trifluoromethoxy-phenyl)-benzenesulfonamide: 4-Trifluoromethoxy aniline (3.4 g, 19.21 minol)was dissolved in THF (25 ml) before pyridine (4.44 ml, 54.88 mmol) was added. 4-Acetyl-benzenesulfonyl chloride (4.0 g, 18.29 mmol) was then added as a solid, and the resulting dark solution was stirred for 10 minutes. The volatiles were then removed, and the resulting residue was suspended in THF. Excess Et3N was then added, and the mixture was stirred for several minutes before the solids were filtered.
The mother liquor was then evaporated to a solid which was purified by flash chroinatography (EtOAc/hexanes) to yield the desired compound as a white solid. (5.5 g(15.31 mmol, 84 %).
Step 2 Br CFa~O 0 0 /\ all, N\S

4-(2-Bromo-acetyl)-N-(4-trifluoromethoxy-phenyl)-benzenesulfonamide: The bromoketone intermediate was synthesized according to the procedures described in the preparation of Example 44 Step 2.

~ I \

// \\
CF3~0 ~ O O
~N\S 0 Thioacetic acid S-{2-oxo-2-[4-(4-trifluoromethoxy-phenylsulfamoyl)-phenyl)-ethyl}
ester: The desired compound was synthesized according to the procedures described in the preparation of Example 44 step 3. 1H NMR: (400MHz, DMSO): 10.74 (s, 1H), 8.08 (d, 2H), 7.92 (d, 2H), 7.23 (d, 2H), 7.20 (d, 2H), 4.57 (s, 2H), 2.38 (s, 3H); LC-MS
(ES+): 360 [MH]+
m/e.

Example 46 s"r O N \ I 0 9"s, O

F
N- [4-(2-Mercapto-acetyl)-phenyl)-3-Fluorobenzene-sulfonamide:
The compound N-[4-(2-Mercapto-acetyl)-phenyl]-3-Fluorobenzene-sulfonainide was synthesized according to the procedures described in the preparation of Exatnple 1. IH-NMR: (CDC13) 7.98(d, 2H), 7.68(d, 1H), 7.60(d, 1H), 7.51(dd, 1H), 7.22 (d, 2H), 4.38(s, 2H), 2.41(s, 3H); LC-MS (ES+): 367.6 [MH]+ m/e Example 47 O

/ ~ S1, I SH F O
O :

Thioacetic acid S-{2-[2-fluoro-4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester The coinpound thioacetic acid S-{2-[2-fluoro-4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-2-oxo-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. , 1H-NMR (DMSO): 11.37 (bs, 1H), 8.02 (dd, 2H), 7.79 (t, 1H), 7.61 (d, 2H), 7.06 (m, 2H), 4.33 (d, 2H), 2,35 (s, 3H). LC-MS
(ES+): 452 [MH]+ m/e.

Example 48 O
~ S
~ y N i 0-0 S
O
O
Thioacetic acid S-[2-(1-benzenesulfonyl-2,3-dihydro-lH-indol-5-yl)-2-oxo-ethyl] ester The compound thioacetic acid S; [2-(1-benzenesulfonyl-2,3-dihydro-lH-indol-5-yl)-2-oxo-ethyl] ester was synthesized according to Scheme IV.

Step 1 O
CO
S
O
1-(1-Senzenesutfonyl-2,3-dihydro-lH-indol-5-yl)-ethanone 1-(2,3-Dihydro-lH-indol-5-yl)-ethanone (0.200 g, 1.24 mmol) was dissolved in THF (2 inl) before pyridine (0.303 ml, 3.75 mmol) was added, leaving a yellow solution.
The benzenesulfonyl chloride (0.220 g, 1.24 mmol) was then added, as a solid, with stirring. The reaction mixture was stirred at 40 C for 5h. The mixture was cooled and the THF and pyridine were reinoved in vacuo. The desired sulfonamide (0.310 g, 1.02 mmol, 82 %) was recrystallized from ethyl acetate and hexanes. It had LC-MS (ES+): 302 [MH] +
in/e Step 2 O
Br N CO--'~ ~ \~ S~O
.,. O
1-(1-Benzenesulfonyl-2,3-dihydro-IH-indol-5-yl)-2-bromo-ethanone: The ketone from step 1(0.310 g, 1.02 mmol) was dissolved in THF (3 ml), and phenyltrimethylammonium tribromide (PTT) (0.386 g, 1.02 mmol) was added as a solid leaving an orange solution which began to deposit a white solid iinmediately. Stirring for 1.5 hours leaves a colorless mixture to which water (5 ml) was added. THF was then evaporated and the resulting aqueous mixture was extracted with ethyl acetate. Drying over Na2SO4 and evaporation leaves a white crystalline solid (85% desired mono-brominated material by LC-MS, 5% starting material, 10% dibrominated) suitable for the next step. LC-MS (ES+): 381, 379 m/e.

Step 3 O
Sllr N I O
~'0 Thioacetic acid S-[2-(1-benzenesulfonyl-2,3-dihydro-lH-indol-5-yl)-2-oxo-ethyl] ester The mono-brominated sulfonamide from step 2(0.194 g, 0.509 mmol) was was dissolved in methanol (2 ml) before potassiuin thioacetate (0.0639 g, 0.560 minol) was added as a solid.
LC-MS of the resulting yellow solution shows the reaction is coinplete in minutes.
Evaporation of the volatiles leaves a tan residue which was taken up into dichloroinethane (4 ml), during which the disulfide of the thioacetic acid was deposited and filtered. The desired thioester could then be recrystallized fioin dichloroinethane/hexanes (0.080 g, 0.214 mmol, 42%). LC-MS (ES+): 376 [MH]+ m/e. 1H-NMR (DMSO): 7.87 (d, 3H), 7.78 (s, 1H), 7.7 (m, 1H), 7.57 (m, 3H), 4.41 (s, 2H), 3.99 (t, 2H), 3.06 (t, 2H), 2.34 (s, 3H) Example 49 O

~
~N

/ \, So0 Thioacetic acid S-[2-(1-benzenesulfonyl-lH-indol-5-yl)-2-oxo-ethyl] ester The compound thioacetic acid S-[2-(1-benzenesulfonyl-lH-indol-5-yl)-2-oxo-ethyl] ester was synthesized according to the procedures described in the preparation of Example 48.
1H-NMR (CDC13): 8.22 (d, 1H), 8.07 (m, 1H), 7.98 (m, 1H), 7.90 (m, 2H), 7.66 (d, 1H), 7.57 (m, IH), 7.47 (m, 2H), 6.76 (m, 1H), 4.42 (s, 2H), 2.40 (s, 3H). LC-MS (ES+):
374 [MH]+
m/e Example 50 / sY
N

SNH
\
Thioacetic acid S-{2-[6-(3-methoxy-benzenesulfonylamino)-pyridin-3-yl]-2-oxo-ethyl}ester The compound thioacetic acid S-{2-[6-(3-methoxy-benzenesulfonylamino)-pyridin-3-yl]-2-oxo-ethyl}ester was synthesized according to the procedures described in the preparation of Exainple 1. 1H-NMR: (CDC13) 8.91(s, 1 H), 8.21(d, IH), 7.51(d, 1 H), 7.44(d, 1H), 7.42(d, 1H), 7.38(s, 1H), 7,09(d, 1H), 4.21(s, 2H), 3.82(s, 3H), 2.41(s, 3H) ppm. MS:
(380.05) Example 51 O
s N S

~N~
~
I H
/

Thioacetic acid S-{2-oxo-2-[4-(pyridine-2-sulfonylamino)-phenyl]-ethyl} ester The coinpound Thioacetic acid S-{2-oxo-2-[4-(pyridine-2-sulfonylamino)-phenyl]-ethyl} ester was synthesized according to the procedures described in the preparation of Example 1. 1H-NMR (DMSO): 11.19 (bs, 1H), 8.71 (d, 1H), 8.10 (in, 2H), 7.89 (d, 2H), 7.67 (m, 1H), 7.28 (d, 2H), 4.41 (s, 2H), 2.36 (s, 3H). LC-MS (ES+): 351 [MH]+ m/e.

Example 52 O
A~y \%
a~~ SN O
H

Thioacetic acid S-{2-oxo-2-[4-(pyridine-3-sulfonylamino)-phenyl]-ethyl} ester The compound Thioacetic acid S-{2-oxo-2-[4-(pyridine-3-sulfonylainino)-phenyl]-ethyl} ester was synthesized according to the procedures described in the preparation of Exainple 1 tH-NMR (DMSO): 11.16 (bs, 1H), 8.99 (d, 1H), 8.81 (dd, 1H), 8.23 (in, 1H), 7.92 (d, 2H), 7.64 (dd, 1H), 7.26 (d, 2H), 4.42 (s, 2H), 2.36 (s, 3H). LC-MS (ES+): 351 [MH]+
m/e.

Example 53 O
y DO S
N O
N
~ S~O

Thioacetic acid S-{2-oxo-2-[1-(pyridine-2-sulfonyl)-2,3-dihydro-lH-indol-5-yl]-ethyl}
ester The coinpound thioacetic acid S-{2-oxo-2-[1-(pyridine-2-sulfonyl)-2,3-dihydro-lH-indol-5-yl]-ethyl} was synthesized according to the procedures described in the preparation of Example 48. 1H-NMR (DMSO): 8.67 (m, IH), 8.11 (m, 2H), 7.82 (m, 2H), 7.50 (m, 1H), 7.43 (d, 1H), 4.42 (s, 2H), 4.23 (t, 2H), 3.15 (t, 2H), 2.36 (s, 3H). LC-MS
(ES+): 377 [MH]+
m/e.

Example 54 O
O / S~PHOH
o .i1 I 11 ~S. ~ O
F3C\ ~ / H
O
Thiophosphoric acid S-{2-oxo-2-[4-(4-trifluoromethoxy-benzenesulfonylamino)-phenyl]-ethyl} ester The compouiid from Example 1, Step 2 (0.200 g, 0.461 mmol) was dissolved in methyl alcohol (3 mL). Na3SPO3 (0.091 g, 0,508 mmol) was added as a solid, fornning a heterogeneous suspension. The suspension was stirred at room temperature.
After 1.5 hours a white precipitate was present. This precipitate was filtered and found to be the undesired disulfide of the starting sulfonainide. The methyl alcohol was then removed and dichloromethane was added to the crude reaction mixture. A white solid precipitated out of the dichloromethane solution which was collected by filtration. The solid was further purified by mass triggered HPLC to give the desired compound (0.050 g, 0.106 mmol, 23%). 'H-NMR (DMSO): 11.06 (s, 1H), 7.98 (in, 2H), 7.88 (in, 2H), 7.59 (d, 2H), 7.23 (m, 2H), 4.16 (d, 2H). LC-MS (ES+): 471 [MH]+ m/e.

The following compounds can be made using the methods as described above and when made should have similar activity as those made above.

G3 ~eGZ'G~S\G4 , wherein Gl is selected from the following:

H

N N
ol ~ N N s N~ N
I H NJ N
v ~ > > > > >
N1 N~ N
N" ~ N
vN N" N N~N and N
> > > > >
G2 is selected from the following:

N'U"I AN i rS.N N, R3 R3 ~ R3 and R4 G3 is selected from the following:

H

Ol NJ NJ H N N~\
> > > , I ~ > > >
N rN N

~ , N NN N~N N,NJ
and and G4 is selected from the following:

-Yk ~ .~ o\ .~
YO ~
~ o 0 ~ 0 0 / I

o' yo / \
O O A N N A \\
)r 0 0 ~ 0 0 s~s X

0~ +N\ S' X

~
~ 0 , , , , , S OH ~\g NHz ~\S NH2 S H
//~ ~ ~ "*,r N I I
OH CO2H COZMe CO2H 0 ~ ~ > >
,S\S H
S' N

CO2MeO -P(O)(OH)2 , and Additionally, the following compounds can be made using the methods as described above.

iGz, ,~is~
~3 ~~ G4, wherein GI is selected from the following:

l/ ~ I ~ I (J3 / I N\N ~ I NN

and G2 is selected from the following:

and G3 is selected from the following:

'~'J 0 0 NN S N1 N
olNJ N J O~~ H N~r N,J\ JJ N
> > > > > > > > > >
CN~ y N N

NI-~ N , N N and N.NJ
and G4 is selected from the following:

~ i~ N
.~ \
y Y**"~~ .~' \
O O O O O O
~
.~ \~ yo i Y(N o O 0~ s~ \ N / I / \~
O O O O O
~ N~ N~ ~ OH ~ OH
0 ~I ~ ~ ~1 54S-~, S / OH '\g~NH2 ,~\S~NHz S~ N
~
OH CO2H CO2Me CO2H 0 H

\S~N~ P O OH
COzMeO , aud -~ ~~ )2 The prophetic examples disclosed herein may optionally be substituted as described above.
The activity of the above mentioned compounds as HDAC inhibitors has generally been shown by the following assays. The other compounds listed above, which may not yet been made or tested, are predicted to generally have activity in these assays as well.
Inhibition Assays:
In vits o HDAC-inhibition Assay:

[00107] This assay measures a compound's ability to inhibit acetyl-lysine deacetylation in vitro and was used as both a primary screening method as well as for IC5o detenninations of confirmed inhibitors. The assay is performed in vitro using an HDAC
enzyme source (e.g. partially purified nuclear extract or iinmunopurified HDAC
complexes) and a proprietary fluorescent substrate/developer system (HDAC Quantizyine Fluor de Lys Fluorescent Activity Assay, BIOMOL). The assay is run in 1,536-well Greiner white-bottom plates using the following volumes and order of addition:
Step 1: Enzyme (2.5 ul) source added to plate (from refrigerated container) Step 2: Compounds (50 nl) added with pin transfer device Step 3: Fluor de Lys (2.5 ul) substrate added, incubate at RT, 30 minutes Step 4: Developer (5 ul) solution is added (containing TSA), to stop reaction Step 5: Plate Reader - data collection The deacetylated fluorophore is excited with 360 nm light and the emitted light (460 nm) is detected on an automated fluorometric plate reader (Aquest, Molecular Devices).
Cellular Histone Hyperacetylation Assays:
[001081 These two secondary assays evaluates a compound's ability to iiihibit HDAC in cells by ineasuring cellular histone acetylation levels. The cytoblot facilitates quantitative EC50 information for cellular HDAC inhibition. Transformed cell lines (e.g.
HeLa, A549, MCF-7) are cultured under standard inedia and culture conditions prior to plating.
For Cytoblot:
[001091 Cells (approx. 2,500/well) are allowed to adhere 10-24 hours to wells of a 384-well Greiner PS assay plate in media containing 1-5% serum. Cells are treated with appropriate cornpound and specific concentrations for 0 to 24 hours. Cells are washed once with PBS (60 ul) and then fixed (95% ethanol, 5% acetic acid or 2% PFA) for 1 ininute at RT
(30 ul). Cells are blocked with 1% BSA for 1 hour and washed and stained witll antibody (e.g. anti-Acetylated Histone H3, Upstate Biotechnology), followed by washing and incubation with an appropriate secondary antibody conjugated to HRP or fluorophore. For luminescence assays, signal is generated using Luminol substrate (Santa Cruz Biotechnology) and detected using an Aquest plate reader (Molecular Devices).
For Iinmunoblot:
[00110J Cells (4 x 105/well) are plated into Conling 6-well dish and allowed to adhere overnight. Cells are treated with compound at appropriate concentration for 12-18 hours at 37 degrees. Cells are washed with PBS on ice. Cells are dislodged with rubber policeman and lysed in buffer containing 25 mM Tris, pH7.6; 150 mM NaC1, 25 mM
MgC12, 1% Tween-20, and nuclei collected by centriguation (7500g). Nuclei are washed once in 25 mM Tris, pH7.6; 10 mM EDTA, collected by centrifugation (7500g). Supematant is removed and histones are extracted using 0.4 M HCl. Samples are centrifuged at 14000g and supernatants are precipitated in 1 ml cold acetone. The histone pellet is dissolved in water and histones are separated and analyzed by SDS-PAGE Coomassie and immunobloting (anti-acetylated histone antibodies, Upstate Biotechnology) using standard techniques.
Differential Cytotoxicity AssU:
[00111] HDAC inhibitors display differential cytotoxicity toward certain transformed cell lines. Cells are cultured according to standard ATCC
recommended conditions that are appropriate to each cell type. Compounds were tested for their ability to kill different cell types (normal and transformed) using the ATPlite luminescence ATP
detection assay system (Perkin Elmer). Assays are run in either 384-well or 1536-well Greiner PS plates. Cells (30 ul or 5 ul, respectively) are dispensed using either rnultichannel pipette for 384-well plates, or proprietary Kalypsys bulk liquid dispenser for 1536-well plates.
Compounds added using proprietary pin-transfer device (500 nL or 5 nL) and incubated 5 to 30 hours prior to analysis. Luminescence is measured using Aquest plate reader (Molecular Devices).
[00112] The activity of some of the coinpounds of the invention are shown in Table 1, below. Syntllesis of each compound is described in the Exainple listed in the left column.
Table 1 Example COMPOUND In vitro IC50 Cellular IC50 No. ( M) ( M) 02 O s 'H <1 <10 \
H

04 0 <1 % S \
, S
F3C_ 1/

06 S <1 <1 F3C- ~ H 0 07 ~ <1 <10 ~i ~~s ~ N
H

, <10 <1 09 <1 <1 .~~P
S~N O
F,C_p ~ H

00 <1 ry p FaC~~ r H

-,-S <1 \ ~5 O \ I --~-NHz F3Cy H
12 i 5~f <1 <10 \ S\N ~ ~ I I
F~C~p I r I
13 5~ <10 p O r r /N ~ 0 F3C, C ~ I
14 p <1 <50 ''s' F,C\ I / H \ CI

0 <1 ~~5~~ ~
N
I ~ F
F,C-16 p 5Y <1 <1 \,p Jr H
17 0 0 S <1 <10 F3C \ S~ N ~ ~ =
H

18 s <1 <1 HJ~Iy I~~s 0 F
0 <1 <1 r~ 11 F,C S~H ~ p CI

<1 <1 01\ //
N O
H

22 0 S <1 <10 H
23 0 <1 <10 N O
0p H
S
24 <10 N /~ i 5~ <1 /~S\H \ 0 N
/
O
25 \0 ~ I <1 N \ ICI
H
27 s <1 <1 O ~ ~N i 0 / H
~O
28 0 ~Sy <1 <1 S_ 0 H
29 <1 <10 H
N
30 0 <1 <1 O
S 4S/, N~I
NN H

31 0 <1 <10 O

33 S~ <1 <10 H '' 0 ~
34 s~ <1 <1 , I ~ .ON ' 0 H
F ~
35 <1 <1 H O

36 0 5~ <1 <1 ~

CI
37 5\/ <1 <1 G oNJ~ 'C' H

38 <1 <1 'i~
H' 39 N <1 <1 , I /

40 ~s <10 >50 I\"
s~
p 41 0 p 0 SY <1 <1 F3C- 5 N \ I 0 ~ , 42 oF' 0 <50 N 5 S~
or ~ -r~ I ol 43 a ci 0 <50 N~/
o~ N N~ ICI

45 ~. 0 s~ <1 <10 N ~ J~
~ N~~\~\ C
F3C'p ~ f p O

46 <1 <1 O S~

y H
F
47 0 <1 <1 0%/i / ( S
~

' S\H \
CFy 48 <1 <10 5~

r S)o 49 0 <1 <1 S' /

N ~I I{
/ \\r- so 50 <1 <1 N:P' I

\INH ~
I
/
o~
51 <1 <1 sy O
I

52 0 <1 <10 Wi / S
~
NI \ S~H \ 0 /

53 <1 <1 S' ~

N I ~IOI( N
'O 0 54 0 oH
sI,,oH
O'\ ri H
F,C~,O
~5\N \ 0 A blank cell indicates that the value was not determined.

All references cited above are incorporated herein by reference in their entirety.

From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims (73)

1. A compound having structural formula (I), or a pharmaceutically acceptable salt, amide, ester, or prodrug thereof, wherein G1 is optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl, or G1 and R3 taken together form an optionally substituted heterocycloalkyl or heteroaryl, both of which can be optionally substituted;
G2 is an N-sulfonamide moiety having structure (II), an S-sulfonamide moiety having structure (III), or an amide of the form -NR3C(O)- or -C(O)NR3-:

G3 is optionally substituted phenyl or optionally substituted 5 or 6 membered heteroaryl, or optionally substituted alkyl, wherein alkyl is preferably C2-C7;
R1 and R2 are each independently selected from the group consisting of hydrogen, lower alkyl, halogen and perhaloalkyl, or R1 and R2 taken together form optionally substituted cycloalkyl;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted lower alkyl, optionally substituted aryl, and optionally substituted alkaryl; or a structural element known to confer aqueous solubility; or R3 and R4 taken together form an optionally substituted heterocycloalkyl;
G4 is chosen from the group consisting of optionally substituted acyl, wherein taken in combination with sulfur forms a thioester, optionally substituted alkyl thiol, wherein G4 taken in combination with sulfur forms a disulfide, and -P(O)(OR5)2 or -P(O)(OH)2, wherein G4 taken in combination with sulfur, forms a phosphorothioate diester or phosphorothioate; and each R5 is independently selected from the group consisting of hydrogen, alkyl, aryl, and arylalkyl.
2. The compounds of Claim 1 wherein G1 is an optionally substituted phenyl having structure (IV) or (V):

wherein X1 and X2 are each independently selected from the group consisting of hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkoxy.
3. The compound of Claim 2, wherein G2 is N-sulfonamide moiety having structure (VI) or (VII) wherein R3 may combine with X1 or X2 to form an optionally substituted bicyclic heterocycle or heteroaryl.
4. The compound of Claim 3 wherein G3 comprises is an optionally substituted phenyl of structure (VIII) or (IX) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl,, dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, aminothiocarbonylaminoalkyl.
5. The compound of Claim 4 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
6. The compound of Claim 5 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
7. The compound of Claim 6 wherein the compound has the structure selected from a group consisting of:

8. The compound of Claim 3 wherein G3 comprises an 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5, wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
9. The compound of Claim 8 wherein G3 is selected from a group consisting of:

10. The compound of Claim 9 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
11. The compound of Claim 10 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
12. The compound of Claim 11 wherein the compound has the structure selected from a group consisting of:

13. The compound of Claim 3 wherein G3 comprises an optionally substituted alkyl.
14. The compound of Claim 13 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
15. The compound of Claim 14 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
16. The compound of Claim 15 wherein the compound has the structure selected from a group consisting of:

17. The compound of Claim 2, wherein G2 is S-sulfonamide moiety having structure (X) or (XI);

18. The compound of Claim 17 wherein G3 comprises is an optionally substituted phenyl of structure (XII) or (XIII) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, aminothiocarbonylaminoalkyl.
19. The compound of Claim 18 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
20. The compound of Claim 19 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility
21. The compound of Claim 20 wherein the compound has the structure selected from a group consisting of:

22. The compound of Claim 17 wherein G3 comprises an 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5, wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
23. The compound of Claim 22 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
24. The compound of Claim 23 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility
25. The compound of Claim 24 wherein the compound has the structure selected from a group consisting of:

26. The compound of Claim 17 wherein G3 comprises an optionally substituted alkyl.
27. The compound of Claim 26 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
28. The compound of Claim 27 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
29. The compound of Claim 28 wherein the compound has the structure selected from a group consisting of:

30. The compound of Claim 1 wherein G1 comprises a 5 or 6 membered heteroaromatic optionally substituted by X1, X2 of structure (XIV);

wherein X1 and X2 are each independently selected from the group consisting of hydrogen, halogen, hydroxyl, optionally substituted lower alkyl, optionally substituted lower alkoxy.
31. The compound of Claim 30 wherein G2 is a sulfonamide having an N-sulfonainide moiety of structure (XV) or an S-sulfonamide moiety of structure (XVI):

32. The compound of Claim 31 wherein G3 comprises an optionally substituted phenyl of structure (XVII):

wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
33. The compound of Claim 32 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
34. The compound of Claim 33 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
35. The compound of Claim 34 wherein G1 is selected from a group consisting of;

36. The compound of Claim 35 wherein the compound has the structure selected from a group consisting of:

37. The compound of Claim 31 wherein G3 comprises an 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5 of structure (XVIII) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
38. The compound of Claim 37 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
39. The compound of Claim 38 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
40. The compound of Claim 39 wherein G1 and G3 is each independently selected from a group consisting of:

41. The compound of Claim 40 wherein the compound has the structure selected from a group consisting of:

42. The compound of Claim 31, structure (XV) wherein G3 comprises an optionally substituted alkyl.

43. The compound of Claim 42 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
44. The compound of Claim 43 wherein R3 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
45. The compound of Claim 44 wherein G1 is selected from a group consisting of:

46. The compound of Claim 45 wherein the compound has the structure selected from a group consisting of:

47. The compound of Claim 32 wherein G3 comprises an optionally substituted phenyl of structure (XIX):

wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
48. The compound of Claim 47 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
49. The compound of Claim 48 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
50. The compound of Claim 49 wherein G1 is selected from a group consisting of:

51. The compound of Claim 50 wherein the compound has the structure selected from a group consisting of:

52. The compound of Claim 32 wherein G3 comprises an 5 or 6 membered heteroaromatic optionally substituted by X3, X4, X5 of structure (XX) wherein X3, X4 and X5 are each independently chosen from a group consisting of hydrogen, perhaloaryloxy, alkanoylalkyl, N-aryl-N-alkylamino, heterocyclylalkoxy, heterocyclylthio, hydroxyalkoxy, carboxamidoalkoxy, alkoxycarbonylalkoxy, alkoxycarbonylalkenyloxy, aralkanoylalkoxy, aralkenoyl, N-alkylcarboxamido, N-haloalkylcarboxamido, N-cycloalkylcarboxamide, N-arylcarboxamidoalkoxy, cycloalkylcarbonyl, cyanoalkoxy, heterocyclylcarbonyl, carboxy, heteroaralkylthio, heteroaralkoxy, cycloalkylamino, acylalkyl, acylalkoxy, aroylalkoxy, heterocyclyloxy, aralkylaryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, haloalkylthio, alkanoyloxy, alkoxy, alkoxyalkyl, cycloalkoxy, cycloalkylalkoxy, hydroxyl, amino, thio, nitro, alkylamino, alkylthio, arylamino, aralkylamino, arylthio, arylthioalkyl, alkylsulfonyl, heteroarylthio, heteroarylsulfonyl, heterocyclylsulfonyl, arylsulphonyl, heteroarylsulfonyl, heterocyclylsulphonyl, heterocyclylthio, alkanoyl, alkenoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl, haloalkanoyl, alkyl, alkenyl, alkynyl, alkenyloxy, alkylenedioxy, haloalkylenedioxy, cycloalkyl, cycloalkylalkanoyl, halo, haloalkyl, haloalkoxy, hydroxyhaloalkyl, hydroxyhaloalkoxy, hydroxyalkyl, aryl, aryloxy, aralkoxy, saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, heteroaralkyl, arylalkenyl, carboalkoxy, alkoxycarboxamido, alkylamidocarboylamido, arylamidocarboylamido, carboalkoxyalkyl, carboalkoxyalkenyl, carboamido, carboxamidoalkyl, cyanocycloalkylalkyl, cycloalkenyl, alkoxycarbonyl, aralkylthio, alkylthio, alkylsulfinyl, arylsulfinyl, dialkylamino, aminoalkyl, , dialkylaminoalkyl, aminoaryl, alkylaminoaryl, acylamino, aminocarbonylalkoxy, aminocarbonylamino, aminothiocarbonylamino, or aminothiocarbonylaminoalkyl.
53. The compound of Claim 52 wherein wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
54. The compound of Claim 53 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
55. The compound of Claim 54 wherein G1 and G3 is each independently selected from a group consisting of:

56. The compound of Claim 55 wherein the compound has the structure selected from a group consisting of:

57. The compound of Claim 31, having structure (XVI) wherein G3 comprises an optionally substituted alkyl.

58. The compound of Claim 57 wherein G4 is an optionally substituted acyl of the formula -C(O)R E, wherein R E is any pharmaceutically acceptable acid, or a phosphoryl moiety that taken together with sulfur forms a phosphorothioic acid; R1 and R2 are each independently hydrogen, lower alkyl or R1 and R2 taken together form optionally substituted cycloalkyl; and X1 and X2 are each independently selected from hydrogen, halogen, hydroxyl, lower alkyl, lower perhaloalkyl, lower alkoxy and lower perhaloalkoxy.
59. The compound of Claim 58 wherein R4 is selected from the group comprising hydrogen, optionally substituted lower alkyl, or a structural element known to confer aqueous solubility.
60. The compound of Claim 59 wherein G1 is selected from a group consisting of:

61. The compound of Claim 60 wherein the compound has the structure selected from a group consisting of:

62. The compound of Claim 1 wherein the compound has the structure consisting of:

63. The compound of Claim 1, wherein R3 is a structural element known to confer aqueous solubility.
64. The compound of Claim 63, wherein R3 is N-piperazinylethyl.
65. The compound of Claim 63, wherein R3 is N-morpholinylethyl.
66. The compound of Claim 63, wherein R3 is 1,3-dihydroxy-2N-propanoyl.
67. The compound of Claim 1, wherein R4 is a structural element known to confer aqueous solubility.
68. The compound of Claim 67, wherein R4 is N-piperazinylethyl.
69. The compound of Claim 67, wherein R4 is N-morpholinylethyl.
70. The compound of Claim 67, wherein R4 is 1,3-dihydroxy-2N-propanoyl.
71. A pharmaceutical composition comprising the compound of Claim 1 in a mixture with at least one carrier, diluent or excipient.
72. The compound of Claim 1 wherein the compound or pharmaceutically acceptable salt, amide, ester or prodrug thereof is capable of inhibiting the catalytic activity of histone deacetylase (HDAC).
73. A method of treating a HDAC related disease in a patient in need thereof by administering a compound of claim 1 to said patient.
CA002567835A 2004-06-10 2005-06-09 Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease Abandoned CA2567835A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
PCT/US2004/018502 WO2004110418A2 (en) 2003-06-10 2004-06-10 Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
US10/865,743 US7271195B2 (en) 2003-06-10 2004-06-10 Carbonyl compounds as inhibitors of histone deacetylase for the treatment of disease
USPCT/US2004/018502 2004-06-10
US10/865,743 2004-06-10
US63501904P 2004-12-09 2004-12-09
US60/635,019 2004-12-09
PCT/US2005/020770 WO2005120515A2 (en) 2004-06-10 2005-06-09 Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease

Publications (1)

Publication Number Publication Date
CA2567835A1 true CA2567835A1 (en) 2005-12-22

Family

ID=35503650

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002567835A Abandoned CA2567835A1 (en) 2004-06-10 2005-06-09 Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease

Country Status (5)

Country Link
EP (1) EP1773398A2 (en)
JP (1) JP2008515777A (en)
AU (1) AU2005251816A1 (en)
CA (1) CA2567835A1 (en)
WO (1) WO2005120515A2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006028104A (en) * 2004-07-16 2006-02-02 Kyowa Hakko Kogyo Co Ltd Histone deacetylase inhibitor
AR050552A1 (en) * 2004-09-02 2006-11-01 Osi Pharm Inc MERCAPTOAMIDS AS INHIBITORS OF HISTONA DEACETILASE
TW200730485A (en) * 2005-07-29 2007-08-16 Kalypsys Inc Multicyclic sulfonamide compounds as inhibitors of histone deacetylase for the treatment of disease
WO2008010985A2 (en) * 2006-07-20 2008-01-24 Merck & Co., Inc. Phosphorus derivatives as histone deacetylase inhibitors
US7786139B2 (en) 2006-11-21 2010-08-31 Omeros Corporation PDE10 inhibitors and related compositions and methods
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2436263C2 (en) * 1974-07-27 1983-02-17 Hoechst Ag, 6000 Frankfurt Thiazolidine derivatives and processes for their preparation
DE2533821A1 (en) * 1975-07-29 1977-02-17 Hoechst Ag THIAZOLIDE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
SE7700500L (en) * 1976-01-20 1977-07-21 Hoechst Ag THIAZOLIDINE DERIVATIVES AND PROCEDURES FOR THEIR PREPARATION
CA2404002A1 (en) * 2000-03-24 2001-09-27 Methylgene, Inc. Inhibitors of histone deacetylase
KR100462045B1 (en) * 2001-06-21 2004-12-16 네오뷰코오롱 주식회사 An metal cap for encapsulating an organic electro-luminescence device and a fabricating method thereof
CA2476586C (en) * 2002-03-13 2011-11-01 Janssen Pharmaceutica N.V. Sulfonyl-derivatives as novel inhibitors of histone deacetylase

Also Published As

Publication number Publication date
AU2005251816A1 (en) 2005-12-22
EP1773398A2 (en) 2007-04-18
WO2005120515A2 (en) 2005-12-22
JP2008515777A (en) 2008-05-15
WO2005120515A3 (en) 2006-03-30

Similar Documents

Publication Publication Date Title
US7381749B2 (en) Sulfonamides as inhibitors of histone deacetylase for the treatment of disease
US7470722B2 (en) Multicyclic sulfonamide compounds as inhibitors of histone deacetylase for the treatment of disease
ES2327269T3 (en) N-SULFONYLPIRROLES AND ITS USE AS INHIBITORS OF HISTONE DEACETILASE.
JP5278983B2 (en) New uses of amide compounds
RU2141478C1 (en) Amides, pharmaceutical composition, methods of decrease of apolipoprotein b secretion, compound
AU2003217393B8 (en) Ansamycins having improved pharmacological and biological properties
US20080194681A1 (en) Novel Inhibitors of Histone Deacetylase for the Treatment of Disease
US8106051B2 (en) Utilities of amide compounds
CA3030581C (en) Apoptosis inhibitors
CA2600531A1 (en) Pyrimidine compounds and methods of use
CA2567835A1 (en) Novel sulfonamides as inhibitors of histone deacetylase for the treatment of disease
AU2008305294A1 (en) Pyridin-2 -yl-amino-1, 2, 4 -thiadiazole derivatives as glucokinase activators for the treatment of diabetes mellitus
JP2016037467A (en) Sulfonamide derivatives and pharmaceutical applications thereof
JP2008515777A5 (en)
WO2007045962A2 (en) Novel hdac inhibitors
MX2007002246A (en) Processes for the preparation of isothiazole derivatives.
WO2012098501A1 (en) Febuxostat co-crystals
Calderone et al. Heterocyclic analogs of benzanilide derivatives as potassium channel activators. IX
GB2154585A (en) Prodrugs of antiinflammatory oxicams
MX2015002624A (en) Non-annulated thiophenylamides as inhibitors of fatty acid binding proteini(fabp) 4 and/or 5.
JP3165866B2 (en) Sulfur-containing heterocyclic compounds
JPS6239583A (en) Benzodioxazole derivative and production thereof
JP2566843B2 (en) Benzothiazine derivatives, their preparation and their application as pharmaceuticals or as synthetic intermediates for pharmaceuticals
KR101630243B1 (en) Novel compounds, pharmaceutically acceptable salts thereof or optical isomer thereof, preparation method thereof and pharmaceutical composition for prevention or treatment of the viral diseases containing the same as an active ingredient
EP0298040B1 (en) 1,3-dithiol-2-ylidene derivatives

Legal Events

Date Code Title Description
FZDE Discontinued
FZDE Discontinued

Effective date: 20100609